Skip to main content
Erschienen in: Molecular Cancer 1/2014

Open Access 01.12.2014 | Research

Aberrant gene expression in mucosa adjacent to tumor reveals a molecular crosstalk in colon cancer

verfasst von: Rebeca Sanz-Pamplona, Antoni Berenguer, David Cordero, David G Molleví, Marta Crous-Bou, Xavier Sole, Laia Paré-Brunet, Elisabet Guino, Ramón Salazar, Cristina Santos, Javier de Oca, Xavier Sanjuan, Francisco Rodriguez-Moranta, Victor Moreno

Erschienen in: Molecular Cancer | Ausgabe 1/2014

Abstract

Background

A colorectal tumor is not an isolated entity growing in a restricted location of the body. The patient’s gut environment constitutes the framework where the tumor evolves and this relationship promotes and includes a complex and tight correlation of the tumor with inflammation, blood vessels formation, nutrition, and gut microbiome composition. The tumor influence in the environment could both promote an anti-tumor or a pro-tumor response.

Methods

A set of 98 paired adjacent mucosa and tumor tissues from colorectal cancer (CRC) patients and 50 colon mucosa from healthy donors (246 samples in total) were included in this work. RNA extracted from each sample was hybridized in Affymetrix chips Human Genome U219. Functional relationships between genes were inferred by means of systems biology using both transcriptional regulation networks (ARACNe algorithm) and protein-protein interaction networks (BIANA software).

Results

Here we report a transcriptomic analysis revealing a number of genes activated in adjacent mucosa from CRC patients, not activated in mucosa from healthy donors. A functional analysis of these genes suggested that this active reaction of the adjacent mucosa was related to the presence of the tumor. Transcriptional and protein-interaction networks were used to further elucidate this response of normal gut in front of the tumor, revealing a crosstalk between proteins secreted by the tumor and receptors activated in the adjacent colon tissue; and vice versa. Remarkably, Slit family of proteins activated ROBO receptors in tumor whereas tumor-secreted proteins transduced a cellular signal finally activating AP-1 in adjacent tissue.

Conclusions

The systems-level approach provides new insights into the micro-ecology of colorectal tumorogenesis. Disrupting this intricate molecular network of cell-cell communication and pro-inflammatory microenvironment could be a therapeutic target in CRC patients.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1476-4598-13-46) contains supplementary material, which is available to authorized users.

Competing interest

The authors have declared that no competing interests exist.

Authors’ contributions

RSP and VM designed the study, conceived the experiments and wrote the article. RSP and AB carried out the experiments. DC performed transcriptional networks. DGM performed inmunohistochemistry and helped to draft the manuscript. XSo, MCB, LPB, and EG analyzed data. CS, JO, XSa, FRM and RS provided samples and clinical data. All authors critically reviewed and had final approval of the article.
Abkürzungen
BIANA
Biological interactions and network analysis
CRC
Colorectal cancer
DEG
Differentially expressed genes
ECM
Extracellular matrix
EMT
Epithelial to mesenchymal transition
DETF
Differentially expressed transcription factors
MSS
Microsatellite stable
PPIN
Protein-protein interaction network
TF
Transcription factor.

Background

Colorectal cancer (CRC) is a complex disease in which many genes, proteins, and molecular processes are implicated. Proteins do not work independently in a tumor cell, but are organized into co-regulated units or pathways that perform a common biological function [1]. Relevant molecular mechanisms involved in cancer are gene regulation, signaling, cell metabolism, and the connections between them, among others [2]. In addition to the tumor cell intrinsic complexity, increasing data support the main role of tumor microenvironment in the mechanisms of CRC progression [35]. Tumor microenvironment is composed by a heterogeneous population of stromal cells such as fibroblasts and immune cells, extracellular matrix components and secreted factors. All these components work orchestrated by molecular transducers like integrins engaging cell-cell and cell-matrix signaling that in turn enhance tumor growth [6].
Besides, a colorectal tumor is not an isolated entity growing in a restricted location of the body. An active communication exists not only between different cell communities within the tumor bulk but also between the tumor and the non-tumor distant mucosa. Hence, the patient’s gut environment constitutes the framework where the tumor evolves and this relationship promotes and includes a complex and tight correlation with inflammation, blood vessels formation, nutrition and gut microbiome composition [7]. Consequently, studying the micro-ecology context of a tumor is central to understand colorectal carcinogenesis. The tumor influence on environment could both promote an anti-tumor and a pro-tumor response. Some microenvironments, particularly those associated with tissue injury, are favorable for progression of mutant cells, whereas others restrict it. Cancer cells can also instruct surrounding tissues to undergo changes that promote malignancy [8].
Field cancerization or the field-effect is a theory first described by Slaughter et al. in oral carcinoma [9]. In the initial phase of the multistep carcinogenesis, a stem cell acquires genetic alterations and forms a “patch”, a clonal unit of altered daughter cells. Further alterations convert the “patch” into a field of pre-neoplastic cells. Although only one cell becomes tumoral, the remaining field (adjacent mucosa) continues in a “pre-neoplastic-state” composed of morphologically normal, but biologically altered epithelial cells. Since this field is a pre-tumor site predisposed towards development of cancer, this hypothesis could explain local recurrences after surgery [10].
Understanding the complex ways in which cancer cells interact with their surroundings, both locally in the tumor organ and systemically in the body as a whole has implications for effective cancer prevention and therapy. In contrast to the gene-centric view, a systems biology approach (defined as the analysis of the molecular relationship between genes and proteins as a whole) can be useful to depict a global view of the cancer disease not only as a tumor cell but as an intricate systemic disease [11].
In this study, mRNA expression from paired tumor (T) and adjacent mucosa from CRC patients (A) and mRNA from mucosa healthy donors (H) were measured using microarrays. The inclusion of samples from healthy subjects has allowed us assessing whether adjacent mucosa from colon cancer patients differs from healthy donors’ mucosa possibly due to the tumor presence. Indeed, a number of differentially expressed genes (DEG) were found between these two entities (A vs. H). Considering their level of expression in tumor tissues, these DEGs were classified as “Tumor-like”, “Trend” or “Adjacent-specific” (A vs. T) patterns. To explain the mechanisms that regulate these patterns of differential expression, networks mimicking transcription regulation were used to search for those transcription factors directly influencing DEG. Then, a systems biology approach using PPIN was applied to describe a crosstalk between cytokines and other proteins secreted by the tumor and receptors activated in the adjacent colon tissue; and vice versa, providing new insights into the micro-ecology of colorectal tumorigenesis. Finally, relevant cytokines and receptors up-regulated in tumor tissue were identified comparing T vs. H expression (Figure 1). Further elucidation of these interactions could be helpful in the development of novel therapeutic strategies oriented to disrupt this molecular crosstalk.

Results

Characterization of differentially expressed genes between adjacent and healthy mucosa

A principal component analysis (PCA) was done to explore the variability of the transcriptomic data from our 246 samples (Figure 2A). As expected, tumor samples appeared as an independent cluster (T in red). Surprisingly, adjacent paired mucosa (A in blue) were also clearly separated from healthy mucosa (H in green), reflecting a large number of differentially expressed genes (DEG) between them. A total of 895 genes were differentially expressed at FDR < 1% and log2 mean difference > 1 between adjacent and healthy mucosa (Additional file 1: Table S1). Interestingly, 88% of these genes were over-expressed in adjacent mucosa (Figure 2B).
The functional enrichment analysis of these genes identified the classical pathways involved in cancer and were highlighted by a significant enrichment of functions related to Inhibition of matrix metalloproteinases, Cell adhesion molecules, cytokine-cytokine receptor interaction, TGF-beta signaling pathway, integrin signaling pathway, complement and coagulation cascades, wound healing, response to external stimulus, inflammatory response and soluble fraction, among others (see complete list in Additional file 2: Table S2, Additional file 3: Table S3 and Additional file 4: Figure S1). This functional analysis suggested an active reaction of the adjacent mucosa related to the presence of the tumor or a more passive reaction induced by factors released from the tumor.
Public transcriptomic data analyzing adjacent and healthy mucosa were used to validate the list of DEG. As a result, 60% of the genes were validated at FDR 1%. At FDR 5%, 91% of the genes were validated (Additional file 5: Table S4 and Additional file 4: Figure S2). These results should be interpreted with caution because each sample type was analyzed in different experiments and, though we normalized the data jointly, we cannot exclude strong batch or laboratory effects. We could not find a dataset like ours, in which healthy and adjacent colon mucosa were analyzed simultaneously.
Figure 3A shows a hierarchical clustering performed with the set of DEG between adjacent mucosa (A) and healthy mucosa (H). Interestingly, the three different tissues were perfectly classified, including the tumors (T) that did not participate in the gene selection. Regarding genes, three patterns of expression were identified as shown in Figure 3B: a) “Tumor-like” (A = T > H or H > A = T) when genes in A had similar pattern as T (349 genes); b) “Trend” (T > A > H or H > A > T) when genes in A had an intermediate expression between H and T (132 genes); and c) “Adjacent-specific” (A < (T,H) or A > (T,H)), when genes were specifically de-regulated in A when compared to either T or H, irrespective of the relationship between T and H (414 genes). The size of this latter group was a surprise that lead us to explore in detail a crosstalk between the tumor and the adjacent mucosa.
Regarding enriched functions for these gene patterns, Tumor-like functions included AP-1 transcription factor network, COX reactions or activation of AP-1, whereas Adjacent-specific functions were enriched in axon guidance, PPAR signaling pathway or BMP2 signaling pathway, among others. These results suggest different functions for each gene expression pattern, though Integrin signaling pathway, complement cascade, adhesion or Interferon signaling were functions shared by the two patterns (see complete list in Additional file 6: Table S5).
Adjacent mucosa samples appeared divided into two groups in the hierarchical clustering analysis (Figure 3A). The smallest of them, with 24 samples, was characterized by high expression in most of adjacent-specific genes. A PCA performed with these adjacent-specific genes showed that the second component was capturing the specificity of this sample cluster and that adjacent mucosa were more similar to tumor than to healthy mucosa (Figure 3C). In fact, the original PCA analysis with all genes also identified these adjacent mucosa samples as highly variable in the second component (Figure 2A).
These clusters were not associated with the clinical parameters gender, age and tumor progression neither with technical parameters RNA integrity value (RIN), 260/230 ratio and plate. In addition, a functional analysis including differentially expressed genes between these two clusters did not show specific functions but essentially those described as characteristic of adjacent mucosa. These results suggest that the smaller cluster of adjacent samples was just an extreme phenotype of these samples. Interestingly, this pattern was also observed in the validation dataset (see heatmap in Additional file 4: Figure S2).

Transcriptional regulation of differentially expressed genes between adjacent and healthy mucosa

We hypothesized that this differential expression could be triggered by a transcriptional program, activated only in adjacent mucosa by the presence of the tumor, and normally silenced in healthy mucosa. This hypothesis was supported by the GSEA results, in which 312 transcription factors motifs were found to be statistically associated with the adjacent mucosa phenotype (nominal p-value < 0.01) but none was found associated to healthy mucosa phenotype (Additional file 3: Table S3).
To further explore this hypothesis, transcriptional networks were inferred and compared using gene expression data of adjacent and healthy mucosa (see Additional file 4: Figure S3). Venn diagram in Figure 4A shows the overlap between nodes of each network. The vast majority of healthy mucosa nodes were also active in adjacent mucosa network whereas 3120 new nodes appeared specific to the adjacent mucosa and 668 nodes disappeared from the network. As expected, DEG between adjacent and healthy mucosa were over-represented in the new active nodes of the adjacent mucosa network (empirical p-value < 10-4) suggesting that DEG are not only performing common functions but also co-regulated in a sub-transcriptional network not active in healthy mucosa samples. Out of 895 DEG, 60 (13%) were transcription factors (TF), and random re-sampling of genes among the complete dataset revealed that DEG were significantly enriched in TF (empirical p-value < 0.001). Among these 60 TF, 35 were specific of the adjacent mucosa transcriptional network.
TF were ranked taking into account the total number of their targets (degree) and the proportion of targets in our DEG list. This rank suggested sub-networks specifically active in adjacent mucosa tissue. TF with higher rank were more specific of adjacent mucosa, and showed higher values of eccentricity (a topological network measure of the spreading of a node in the network) and lower values of closeness centrality (Table 1).
Table 1
List of transcription factors differentially expressed between adjacent and healthy mucosa samples
Gene symbol
Connections
DEG connections
Rank
P-value
Eccentricity
Closenness centrality
Healthy mucosa network
FOSB
4
4
1.000
<1.00E-04
9
0.209
NO
JUN
2
2
1.000
<1.00E-04
11
0.151
NO
NR4A2
5
5
1.000
<1.00E-04
9
0.204
NO
OSR2
12
12
1.000
<1.00E-04
8
0.234
NO
ZBTB16
1
1
1.000
<1.00E-04
9
0.200
NO
EGR2
10
9
0.900
<1.00E-04
8
0.247
NO
NR4A3
9
8
0.889
<1.00E-04
8
0.249
NO
EBF1
162
143
0.883
<1.00E-04
7
0.294
YES
HEY2
24
21
0.875
<1.00E-04
8
0.266
NO
NR4A1
16
14
0.875
<1.00E-04
8
0.240
NO
PRRX1
30
26
0.867
<1.00E-04
7
0.289
NO
EGR1
13
11
0.846
<1.00E-04
9
0.217
YES
FOS
6
5
0.833
<1.00E-04
10
0.178
NO
JUNB
5
4
0.800
<1.00E-04
10
0.178
NO
MEOX2
75
60
0.800
<1.00E-04
7
0.303
NO
ZFPM2
133
106
0.797
<1.00E-04
7
0.296
NO
ERG
76
60
0.789
<1.00E-04
8
0.287
NO
TSHZ2
109
85
0.780
<1.00E-04
7
0.302
NO
FOXC1
27
21
0.778
<1.00E-04
8
0.268
NO
HLF
84
65
0.774
<1.00E-04
7
0.301
NO
MEIS2
133
100
0.752
<1.00E-04
8
0.305
NO
CREB5
32
24
0.750
<1.00E-04
8
0.278
NO
PRDM6
83
62
0.747
<1.00E-04
7
0.293
NO
GLIS2
79
58
0.734
<1.00E-04
7
0.302
NO
HAND2
138
101
0.732
<1.00E-04
7
0.289
NO
EGR3
11
8
0.727
<1.00E-04
8
0.263
NO
SOX18
40
29
0.725
<1.00E-04
7
0.274
NO
ZNF423
79
57
0.722
<1.00E-04
8
0.303
YES
PHOX2B
60
42
0.700
<1.00E-04
8
0.275
YES
KLF7
133
92
0.692
<1.00E-04
7
0.313
NO
GLI3
217
150
0.691
<1.00E-04
7
0.325
NO
MEIS1
113
77
0.681
<1.00E-04
8
0.301
YES
KLF2
15
10
0.667
<1.00E-04
8
0.261
YES
TSHZ3
80
52
0.650
<1.00E-04
8
0.302
YES
NKX2-3
75
47
0.627
<1.00E-04
8
0.290
NO
BNC2
252
148
0.587
<1.00E-04
7
0.334
NO
PITX2
26
15
0.577
<1.00E-04
9
0.244
YES
PRDM8
70
40
0.571
<1.00E-04
7
0.304
YES
NR2F2
141
78
0.553
<1.00E-04
7
0.334
YES
TSC22D3
29
16
0.552
<1.00E-04
7
0.282
NO
PBX3
186
102
0.548
<1.00E-04
7
0.337
YES
ZEB1
264
139
0.527
<1.00E-04
7
0.343
YES
FOXF1
62
31
0.500
<1.00E-04
7
0.312
YES
ZNF532
110
52
0.473
<1.00E-04
7
0.326
YES
CAMTA1
68
30
0.441
<1.00E-04
7
0.318
YES
JAZF1
172
74
0.430
<1.00E-04
7
0.334
NO
AFF3
31
11
0.355
<1.00E-04
7
0.295
YES
NFIC
131
43
0.328
<1.00E-04
7
0.339
YES
ZEB2
74
24
0.324
<1.00E-04
7
0.324
YES
TCF4
408
129
0.316
<1.00E-04
6
0.371
YES
BCL6
36
11
0.306
<1.00E-04
7
0.309
NO
NR1H4
77
22
0.286
<1.00E-04
8
0.282
YES
MAFB
125
23
0.184
<1.00E-04
6
0.313
YES
HOXB13
93
14
0.151
<1.00E-04
8
0.264
NO
ATF3
4
3
0.750
0.0003
10
0.184
NO
ZBTB20
149
13
0.087
0.0183982
7
0.356
YES
THRB
12
2
0.167
0.0986901
8
0.259
YES
HOXB6
2
0
0.000
1
8
0.227
YES
IFI16
5
0
0.000
1
7
0.247
YES
NEUROD1
12
0
0.000
1
8
0.236
NO
Genes from the AP-1 complex (Fosb and Jun) ranked first in the TF list. The AP-1 subunits Fos, Junb, Mafb and Atf3 also appeared in the list. Previous GSEA analysis also had revealed as most significant motive “Genes with promoter regions [-2 kb,2 kb] around transcription start site containing the motif TGACTCANNSKN which matches annotation for JUN” (p-value = 0.002, and FDR q-value = 0.015, Additional file 4: Figure S4). A high correlation existed between the expression of Jun and Fos AP-1 subunits in adjacent mucosa but not in healthy mucosa (Spearman’s correlation 0.67 and 0.23 respectively; Figure 4B). Interestingly, these TF belonged to the “tumor-like” genes pattern (Figure 4C). Fos, Jun, Fosb and Junb did not appear in healthy mucosa transcriptional network highlighting their idiosyncratic role in adjacent mucosa. The family of transcription factors NR4A1, NR4A2 and NR4A3 also ranked in top positions. Other TF such as GLI3, BCN2, EBF1 and ZEB1 were also significant because of their high rank in the network and large number of DEG targets.

Deciphering a crosstalk between adjacent mucosa and tumor through a protein-protein interaction network

Changes in adjacent mucosa not detected in healthy mucosa might be a direct response in front of tumor stimulus based on a physical crosstalk between the cells (Figure 1). This molecular communication could be through the direct interaction between secreted proteins and their corresponding membrane receptors. The following strategy was applied to identify interactions compatible with this hypothesis: 1) Search for over-expressed genes in tumors compared to healthy mucosa in addition to previous DEG. 2) Identify those that code for secreted proteins and membrane receptors. 3) Construct a protein interaction network with the selected genes. 4) Identify interaction pairs that reflect cellular communication in both directions: from tumor to adjacent (efferent pathway) and vice versa (afferent pathway).
From the 788 over-expressed genes in adjacent mucosa vs. healthy mucosa, 324 (41%) corresponded to secreted (n = 111) or membrane (n = 213) genes. In addition, 442 genes (250 secreted and 192 membrane) over-expressed in tumors were included in the analysis. A level 0 (only direct interactions) protein-protein interaction network was retrieved using the 766 up-regulated secreted/membrane genes in adjacent and tumor samples as input. The resulting network included 291 nodes connected by 596 interactions, the majority of them integrated in a giant component (Figure 5A). A functional analysis of this network revealed cell adhesion, response to external stimulus, response to wounding, and anatomical structure development as the most statistically significant functions (Additional file 7: Table S6).
A curated analysis of the network revealed 84 crosstalk interactions (Table 2), 61 of them efferent (tumor secreted proteins linked to a receptor in adjacent mucosa tissue), and 23 afferent (adjacent mucosa secreted proteins linked to a receptor in tumor). Figure 5B shows an abstraction of the original network restricted to crosstalk interactions. It is remarkable that 6 out of 23 afferent interactions (26%) included members of the Slit family of secreted proteins, which emerged as relevant players in tumor crosstalk determining the adjacent mucosa response. In the network, Slit2 and Slit3 were redundantly activating Robo1, Robo2, Robo4 and ITGA1 receptors in tumor. Slit family followed an adjacent-specific pattern of expression (see Figure 6A). Due to its importance, and as a proof of concept of the overall strategy of gene selection, inmunohistochemical staining was done to asses the protein expression of Slit2 and the receptor Robo2. Slit2 was expressed in adjacent epithelial cells and also in stromal cancer cells. Robo2 was expressed in both epithelial and stromal cells in cancer tissue but not in adjacent tissue (Figure 6B). Other interesting afferent crosstalk pairs involved LRP8 receptor in tumor activated by a double stimulus of RELN and CLU proteins secreted by adjacent mucosa, and VIP, an intestinal peptide that causes vasodilatation, linked to MME receptor in tumor cells.
Table 2
Afferent and efferent pairs in the crosstalk network
AFFERENT PATHWAYS
SECRETED BY ADJACENT MUCOSA
LOCATED IN TUMOR MEMBRANE
Gene symbol
Function
Gene symbol
Function
SLIT2
Cell migration/axon guidance
ITGA1
Integrin-mediated cellular signalling and axon guidance
SLIT2
ROBO2
Cell migration/axon guidance
SLIT2
ROBO1
SLIT3
ROBO2
SLIT3
ROBO1
SLIT2
ROBO4
Cell migration/angiogenesis
FGF7
Growth factor activity
FGFR4
Regulation of cell proliferation, differentiation and migration
PTN
Growth factor with mitogenic activity
SDC3
Organization of cell shape
TIMP2
Metalloendopeptidase inhibitor activity
MMP14
Metalloendopeptidase activit (angiogenesis, cell proliferation…)
MMP3
Regulation of cell migration
MMP14
SPOCK1
Cell adhesion
MMP14
ISG15
Interferon-mediated signaling pathway
NEDD4
Virus-host interaction
VIP
Intestinal peptide that causes vasodilation
MME
Cellular response to cytokine stimulus
MYOC
Anatomical structure morphogenesis
CD81
Cell proliferation
RELN
Neuron migration
LRP8
Cytokine-mediated signalling pathway
CLU
Platelet and complement activation
LRP8
SERPING1
Innate immunity
SELE
Inflammatory response
CYR61
Cell adhesion/chemotaxis
ITGA1
integrin-mediated signaling pathway
FIGF
Angiogenesis
ITGA9
Integrin-mediated signaling pathway, cell adhesion
HBEGF
Growth factor activity
CD82
Metastasis suppressor gene
CXCL12
Immune response
CXCR4
Inflammatory response
CFH
Innate immunity
IGDCC4
Inmunoglobulin
CFD
Innate immunity
IGDCC4
 
EFFERENT PATHWAYS
SECRETED BY TUMOR
LOCATED IN ADJACENT MUCOSA MEMBRANE
Gene symbol
Function
Gene symbol
Function
IL8
Inflammatory response
DARC
Inflammatory response
MIF
Inflammatory response
CALD1
Cellular component movement
PLA1A
Lipid catabolic process
CALD1
PF4
Immune response and cytokine-mediated signaling pathway
LDLR
Lipid transport and metabolism
WNT5A
In the presence of ROR2, inhibits the canonical Wnt pathway
ROR2
Wnt receptor signaling pathway
REG3A
Inflammatory response
SDC2
Wound healing/carbohydrate metabolic process
SPARC
Regulation of cell proliferation
SDC2
KAL1
Extracellular matrix structural constituent
SDC2
PF4
Immune response and cytokine-mediated signaling pathway
SDC2
FN1
Extracellular matrix structural constituent involved in multiple cellular functions
SDC2
FN1
ITGA5
Angiogenesis/cell adhesion/wound healing
FN1
IGDCC4
Inmunoglobulin
FN1
CD36
Antigen processing and presentation, lipid storage, cell adhesion
FN1
PECAM1
Cell adhesion, signal transduction
TNC
Guidance of migrating neurons
CNTN1
Notch signaling pathway, cell adhesion
TNC
 
ITGA5
Angiogenesis/cell adhesion/wound healing
TNC
 
ITGA9
Integrin-mediated signaling pathway, cell adhesion
COL18A1
Inhibits endothelial cell proliferation and angiogenesis
ITGA5
Angiogenesis/cell adhesion/wound healing
SFRP2
Wnt receptor signaling pathway
ITGA5
SPP1
Cell adhesion, response to vitamin D
ITGA5
SPP1
ITGA9
Integrin-mediated signaling pathway, cell adhesion
AGT
Renin-angiotensin system
AGTR1
Inflammatory response, Rho protein signal transduction, Renin-angiotensin system
AGT
CTSG
Immune response
APOC2
Lipid metabolism
IGDCC4
Inmunoglobulin
PPBP
Chemotaxis and inmune response
IGDCC4
PPBP
CTSG
Immune response
TAC1
Peptide which excite neurons, and are potent vasodilators
TACR2
Response to stress
CXCL5
Chemotaxis and inmune response
DARC
Inflammatory response
CCL2
Chemotaxis and inflammatory response
DARC
C4A
Inflammatory response
IGDCC4
Inmunoglobulin
IGHG1
Innate inmune response
IGDCC4
 
PCSK9
Cellular response to starvation/cholesterol metabolic proces
LDLR
Lipid transport and metabolism
MMP9
Proteolysis
ITGA5
Angiogenesis/cell adhesion/wound healing
MMP9
RECK
Blood vessel maturation
VEGFA
Growth factor active in angiogenesis
NRP2
Angiogenesis
PGF
Growth factor active in angiogenesis
NRP2
ADAM12
Epidermal growth factor receptor signaling pathway
ITGA9
Integrin-mediated signaling pathway, cell adhesion
COL4A1
Angiogenesis
CD93
Macrophage activation, cell-cell adhesion
COL4A1
CD36
Antigen processing and presentation, lipid storage, cell adhesion
COL1A1
Positive regulation of cell migration/positive regulation of epithelial to mesenchymal transition
DDR2
Cell adhesion, ossification
COL1A1
ITGA5
Angiogenesis/cell adhesion/wound healing
COL1A1
CD93
Macrophage activation, cell-cell adhesion
COL1A1
CD36
Antigen processing and presentation, lipid storage, cell adhesion
COL1A2
Transforming growth factor beta receptor signaling pathway/platelet activation/leukocyte migration
CD93
Macrophage activation, cell-cell adhesion
COL1A2
CD36
Antigen processing and presentation, lipid storage, cell adhesion
COL3A1
Integrin-mediated signaling pathway/blood vessel develpment
DDR2
Cell adhesion, ossification
COL6A1
Axon guidance/cell adhesion
CD36
Antigen processing and presentation, lipid storage, cell adhesion
COL6A3
Axon guidance/cell adhesion
ITGA5
Angiogenesis/cell adhesion/wound healing
COL4A2
Cellular response to transforming growth factor beta stimulus/axon guidance/angiogenesis
CD93
Macrophage activation, cell-cell adhesion
COL4A2
CD36
Antigen processing and presentation, lipid storage, cell adhesion
LAMA4
Cell adhesion
ITGA5
Angiogenesis/cell adhesion/wound healing
CFB
Complement activation
IGDCC4
Inmunoglobulin
SEMA3F
Cell migration
NRP2
Angiogenesis
EFNA3
Cell-cell signalling
EPHA3
Cell adhesion and migration
C3
Complement activation/Fatty acid metabolism
CTSG
Immune response
INHBA
Cell surface receptor signaling pathway
TGFBR3
Negative regulation of transforming growth factor beta receptor signaling pathway
LIF
Growth factor activity
LIFR
Cell proliferation
FN1
Cell adhesion, cell motility, wound healing
FGFR1
Cell proliferation, differentiation and migration
IGHG1
Complement activation
FGFR1
ELN
Extracellular matrix organization, cell proliferation
FGFR1
C3
Complement activation
FGFR1
 
Efferent interactions were more numerous and included interleukins (IL-8), extracellular-matrix components (Fibronectin, Collagen) or molecules related to invasion like SPARC linked with receptors such as integrins or complement receptors (see Table 2 for specific pairs). Interestingly, the vascular endothelial growth factor receptor NRP2, over-expressed in adjacent mucosa, interacted with a plethora of candidate activating secreted factors from tumors such as VEGFA or SEMA3F. Another interesting finding was the over-expression of LIF in tumor, whose receptor LIFR was over-expressed in adjacent mucosa but not in the tumor. These results were highly indicative of an active crosstalk between cells in the gut microenvironment that triggers an intra-cellular signaling response. The protein-protein interaction network also revealed autocrine signals within tumor or adjacent mucosa. For example, the vascular endothelial growth factor receptor FLT1 was found linked with its ligand VEGFA, both over-expressed in tumor samples.
The bulk tumor includes a mixture of epithelial and active stromal cells. In order to assess which compartment was predominantly expressing the proteins involved in the identified crosstalk interactions, we used the expression data described by Calon et al. [4] who analyzed profiles of each cell population sorted from human CRC. As a result, the vast majority of genes were over-expressed in the stromal compartment (i.e. collagens, interleukins) indicating their active role in the remodeling of the surrounding microenvironment (Additional file 8: Table S7).
To look for hypothetical relationships explaining the communication loop between TF and membrane receptors activated in CRC-adjacent mucosa, a network using as seed proteins AP-1 and membrane receptors was retrieved. Only experimentally-determined interactions were used to construct this level 1 PPIN (including proteins working as bridges between seed proteins that add information to the studied system). As a result, a strong physical interaction between these two cellular components (the extracellular one and the nuclear one) was found. Twenty-one membrane receptors (out of 22) interact with each other through linker proteins to transduce a cellular signal across the extracellular matrix and membrane, finally activating TF belonging to the AP-1 complex (Additional file 4: Figure S5). It is remarkable the close relationship found between ITGA9, ITGA5, CD36, CD93, TGFBR3 and RECK receptors. Also, this analysis revealed a direct path from ROR2 receptor and the AP-1 transcriptional sub-network, being the ligand WNT5A (up-regulated in tumor tissues) the activator of this signal.

Discussion

There is clear evidence of the relevance of the tumor-microenvironment crosstalk for carcinogenesis [1215]. Here we describe altered patterns of expression of the adjacent mucosa from colon cancer patients that could be a direct response against the tumor or induced by the tumor. The analysis of transcriptional profiles and the regulatory networks derived from them allowed us identifying the pathways involved in tumor-microenvironment crosstalk.
We can not discard that at least part of the differences found between adjacent and healthy mucosa were explained by the existence of a pre-neoplastic field in the gut. Studies of adjacent mucosa of the head and neck tumors indicate that such fields can expand more than 7 cm in diameter [10]. Nevertheless, a study in CRC by Jothy S. et al. reported a gradient of carcinoembryonic antigen (CEA) expression expanding only 5 cm. from the peritumor area [16]. In our study, adjacent tissue from patients was dissected from the proximal tumor resection margin, with a minimum distance of 10 cm. However, a recent paper by Hawthorn et Mojica suggests that the field effect cancerization could be evident up to 10 cm. from the tumor [17].
Previous studies usually have compared paired tumor and adjacent mucosa tissues, which can result in misleading interpretations. We have used a large sample of healthy mucosa as reference for gene expression comparisons and have identified a large number of DEG that can be grouped into three altered patterns: “tumor-like”, “trend”, and “adjacent-specific”. Our conclusion is that adjacent normal mucosa is not so normal. In fact, studies that only compare tumor and adjacent mucosa may miss good cancer biomarkers candidates, because many genes are deregulated in adjacent mucosa mimicking the tumor expression.
The predominant functions of DEG are mainly related to response to stimulus, extracellular matrix (ECM) remodeling, organ morphogenesis, and cell adhesion. Remodeling of the ECM network though controlled proteolysis regulates tissue tension, generate pathways for migration, and release ECM protein fragments to direct normal developmental processes such as branching morphogenesis [8]. Collagens are major components of the ECM of which basement membrane type IV and interstitial matrix type I are the most prevalent. Abnormal expression, proteolysis and structure of these collagens influence cellular functions to elicit multiple effects on tumors, including proliferation, initiation, invasion, metastasis, and therapy response [18]. It has been described that integrins that connect various cell types play a vital role in the survival of a growing tumor mass by orchestrating signaling pathways activated through cell-cell and cell-matrix interactions [6]. In our system, integrins ITGA5 and ITGA9 emerged as active signal transducers, occupying central positions in the cellular networks. This result suggests that integrins are not only vital proteins in tumor cells but also in normal-adjacent cells. Moreover, our results indicate that proteins implicated in the described crosstalk are predominantly over-expressed by the tumor stroma. This result underscores the important role of this compartment in CRC carcinogenesis.
One important finding is that DEG are enriched in transcription factors. This indicates the existence of a transcriptional program driving the altered expression pattern observed in adjacent mucosa. A loop including members of the AP-1 family of transcription factors emerged as the most significant one in the analysis. Interestingly, these TF are over-expressed in both adjacent mucosa and tumor tissue. AP-1 members homo or hetero dimerize to assemble the activator protein 1 (AP-1). AP-1 transcription factor acts synergistically with SMAD3/SMAD4 component and is implicated in the regulation of a variety of cellular processes including proliferation and survival, differentiation, growth, apoptosis, cell migration, and inflammation [19, 20]. Topologically, these nodes have a low centrality but a high eccentricity in the transcriptional network. This result can be a little controversial since it is widely accepted that the more centered a node is the more important their functional role in the studied system [21]. However, a recent publication postulates that nodes with high eccentricity could be quickly activated by external factors [22]. This observation could explain the radial position of AP-1 members Jun, Fos, FosB and JunB into the transcriptional network as important fast effectors mediating response against the tumor.
We hypothesized that cytokines and other signaling proteins secreted by the tumor activate membrane receptors of adjacent mucosa cells that initiate this transcription factor activity. Tumor-secreted growth factors act as paracrine agents distorting the normal tissue homeostasis. In turn, tumors are both maintained or attacked by signals from the surrounding microenvironment inducing stromal reaction, angiogenesis and inflammatory responses. To gain insight into the molecular mechanisms underlying this phenomenon, a bi-tissue PPIN analysis strategy was performed to extract patterns of receptor activation in both directions from adjacent mucosa to tumor and vice-versa. Robo genes appeared as the most recurrently receptors activated in tumor membrane by Slit family of proteins. Slits have been implicated in regulating a variety of life activities, such as axon guidance, neuronal migration, neuronal morphological differentiation, tumor metastasis, angiogenesis and heart morphogenesis [23]. Several studies have demonstrated dual roles for Slit and Robo in cancer, acting as both oncogenes and tumor suppressors [24]. This bi-functionality is also observed in their roles as axon guidance cues in the developing nervous system, where they both attract and repel neuronal migration [25]. In CRC, Slit2 up-regulation has been reported as beneficial for the overall survival of patients [26]. Slit is under-expressed in patients with metastatic colorectal cancer and their over-expression in cells resulted in an inhibition of cell migration through AKT-GSK3β signaling pathway. In our data, no significant association between Slit2 or Slit3 level of expression and prognosis was found.
CLU-RELN-LRP8 was other afferent axis to consider for further analysis. CLU codifies the protein Clusterin that has been described as both tumor suppressor and pro-survival factor in colon cancer depending on the intra- and extracellular microenvironment crosstalk [27]. In fact, it has been reported that Clusterin is a protein that shares the intracellular information with the microenvironment and it also experiences a systemic diffusion, acting as a factor that synergistically interacts with their surrounding microenvironment [28]. Moreover, it has been proposed as a diagnostic biomarker in colon cancer [29]. The other CRC-mucosa-secreted protein activating LRP8 receptor in tumor is Reelin (RELN), a glycoprotein that plays an important role in neuronal migration through the activation of lipoproteins receptors such as LRP8 [30]. Also, Reelin has been proposed as a pro-metastatic factor due to their role in cancer cell migration through TGF-β pathway activation [31].
Efferent pathways were also of interest. LIF is a member of the IL6 family of cytokines that displays pleiotropic effects on various cell types and organs [32, 33]. In our system, its receptor LIFR was expressed in the colonic epithelium. It has been reported that LIF stimulates the Jak/STAT pathway to produce nitric oxide (NO) [34, 35]. Based on this, we hypothesize that, in our model, tumor LIF activates Jak-STAT pathway in normal epithelial cells through LIFR receptor leading to NO release and the subsequent creation of a pro-inflammatory environment. Moreover, in our model, Angiotensinogen (AGT) was produced by the tumor and their receptor (AGTR1) was located in membrane from adjacent tissue. Since Angiotensinogen is the precursor form of the active peptide Angiotensin, the pair AGT-AGTR1 makes up the renin-angiotensin system (RAS), usually associated with cardiovascular homeostasis but recently associated with tumor growth [36]. RAS could play a synergistic effect with LIF inducing NO production, leading to inflammation, macrophage infiltration and tumor-induced fibrosis. In addition to their pro-inflammatory role, it has been reported that NO can activate notch-signaling pathway leading to the induction of tumors [37].
Conceptually, an active sub-network includes differentially expressed and connected proteins in a given phenotype. Here we have described a sub-network including membrane receptors over-expressed in normal adjacent tissue acting together in cell-adhesion and with functions on cell surface signal transduction that finally activate the AP-1 transcription factor. ROR2 has emerged as an important link in the crossroad between cell surface entering signal and Fos/Jun transcriptional role as previously described [38]. ROR2 is tyrosine-kinase receptor that plays an important role in developmental morphogenesis [39] and in our network it was activated by the tumor-secreted WNT5A, a WNT pathway signaling mediator.
We do not exclude the possibility that genes having a pivotal role in crosstalk between adjacent and tumor tissue also have a direct relationship with prognosis. In our data, expression of Fos and Jun were found to be protective when over-expressed in adjacent but not in tumor tissue (log-rank p-value = 0.042). Further studies are needed to experimentally corroborate this hypothesis and to test the utility of these transcription factors as prognosis biomarkers. Nevertheless, a complex equilibrium between positively pro-survival and pro-apoptotic signals given by the microenvironment ultimately influences the tumor growth and their plasticity. This could be one of the reasons why prognosis signatures that only take into account tumor but not adjacent tissue expression fail to accurate predict patients’ outcome [40].
The study has some methodological and technical limitations. Though we obtained adjacent mucosa from the farthest resection margin and usually required at least 10 cm, it is possible that some of the variability observed among adjacent mucosa might be related to the distance to the tumor that we cannot analyze. Also, despite a careful dissection of tumor blocks before RNA extraction was done, a normal adjacent tissue infiltration can exist in some tumor samples. Regarding analytical methods, the network analysis only considered well-annotated genes. Some TFs were excluded from the transcriptional network analysis due to their low variability in our data. For these reasons, some genes with a putative role in colon tissue remodeling could have been missed. In fact, we did not find TGF-β, proposed as an important microenvironment modifier [4] because its probeset had very low expression level in our microarray. Finally, our study only included colon specimens, which could raise a concern about generalizability of the results. However, we have previously analyzed that the expression levels are very similar in colon and rectal tumors [41] and this has been confirmed in the TCGA study [42].

Conclusions

In conclusion, gene expression in cells comprising normal adjacent tissue in CRC patients is not so normal and this could have important implications in colorectal cancer prognosis and progression. A systems-level approach has been useful to gain insight into the molecular mechanisms by which adjacent mucosa activates a transcriptomic program in response to cytokines and other signaling proteins secreted by the tumor. We hypothesize that a crosstalk exists, not only between different cell communities within the tumor bulk, but also between colorectal tumor cells and adjacent mucosa, which reacts against the tumor like against a wound. Tumor-secreted growth factors act as paracrine agents distorting the normal tissue homeostasis. In turn, tumors are both maintained and/or attacked by signals from the surrounding microenvironment inducing stromal reaction, angiogenesis and inflammatory responses. Disrupting this intricate molecular network of cell-cell communication and signal transduction could be a therapeutic target in CRC patients.

Methods

Patients and samples

A set of 98 paired adjacent normal and tumor tissues from CRC patients and 50 colon mucosa from healthy donors (246 samples in total) were included in this work. Patients were selected to form a homogeneous clinical group of stage II, microsatellite stable (MSS) colorectal tumors. All had been treated with radical surgery, had not received adjuvant therapy and had a minimum follow up of three years. Adjacent normal tissue from patients was dissected from the proximal tumor resection margin with a minimum distance of 10 cm. Healthy donors were invited to participate in this study when they underwent a colonoscopy indicated for screening or symptoms with no evidence of lesions in the colon or rectum (Additional file 9: Table S8). In this paper we use tumor (T), adjacent mucosa (A) and healthy mucosa (H) to designate the different tissue origins for the samples analyzed. All patients were recruited at the Bellvitge University Hospital (Spain) and the Ethics Committee approved the protocol. Written informed consent from patients and healthy donors was required for inclusion in this study.

Differential expression analysis

RNA extracted from each sample was hybridized in Affymetrix chips Human Genome U219. After a quality control assessment following Affymetrix standards, data was normalized using the RMA algorithm [43]. Both raw and normalized data are available in the NCBI’s Gene Expression Omnibus (GEO) database [44] through accession number GSE44076.
Prior to the identification of differentially expressed genes, a filter was applied to remove low variability probes (n = 15,533), which mostly corresponded to non-hybridized and saturated measures. The remaining 33,853 probes showed a standard deviation greater than 0.3 and were considered for further analysis. A t-test was used to identify differences in gene expression between apparently normal adjacent mucosa from CRC patients (A) and mucosa from healthy donors (H). A probe was considered differentially expressed when it was significant at 1% FDR (q-value method) and showed an absolute log2 mean difference higher than 1 (double expression). The same criteria were applied to identify differentially expressed genes between tumor (T) and healthy mucosa (H).
To attempt a validation of the differentially expressed genes, the same methods were applied to compare samples of healthy colonic mucosa (n = 13) and adjacent mucosa (n = 24) extracted from public datasets GSE38713 [45] and GSE23878 [46].

Functional analysis

Pathway enrichment analysis was performed using two methods. First, Sigora R package [47] was used, which focuses on genes or gene-pairs that are (as a combination) specific to a single pathway. Sigora contains pre-computed data for human pathways in the KEGG [48], BIOCARTA [49], NCI [50], INOH [51] and REACTOME [52] repositories. Second, the gene set enrichment analysis (GSEA) algorithm was also applied, which uses the ranking of differences to identify pathways from a large list of pre-specified sets [53].

Analysis of transcription factors

Transcriptional networks attempt to translate gene expression correlations into transcriptional relationships to reconstruct regulatory loops between transcription factors and their target genes. Transcriptional regulation networks had been previously inferred using the ARACNe algorithm [54], which identifies direct regulatory associations between transcription factors and targets from mutual information measures of co-expression. The associations, represented as a transcriptional network, were used to identify and characterize transcription factors de-regulated in adjacent mucosa from patients when compared to healthy mucosa. Deregulated transcription factors were ranked using a score that took into account both topological parameters of the network and the node expression values. This score divided the number of deregulated nodes linked to each transcription factor by the total number of nodes linked to the transcription factor. To assess statistical significance, a p-value was calculated by re-sampling 1000 times random lists of genes. For each transcription factor, the Network Analyzer module [55] from Cytoscape [56] was used to extract the topological parameters closeness centrality and eccentricity. Only those genes annotated as transcription factor based on experimental data were used in this analysis, whereas those annotated “in silico” were not considered [57, 58].

Protein-protein interaction network construction and analysis

Protein interaction data can be represented as networks were nodes represent proteins and edges represent physical interactions between them. BIANA software (Biological Interactions and Network Analysis) was used to retrieve such networks [59]. BIANA builds networks by selecting interacting partners for an initial set of seed proteins (i.e., the relevant proteins), combining experimentally-determined data from DIP [60], MIPS [61], HPRD [62], BIND [63] and the human interactions from two high-throughput experiments [64, 65]. The integration of multiple sources of interaction data into a single repository allows working with an extensive set of interactions. For our analysis, only human and experimentally-determined interactions were taken into account. Cytoscape software and its plug-ins were used to analyze and visualize the networks.

Cellular classification of tumor proteins implicated in the crosstalk

Proteins were classified as “epithelial” or “stromal” on the basis of their gene level of expression in specific cellular subtypes. For this classification, normalized data from the public dataset GSE39396 was used, which included 24 samples corresponding to different human CRC cell populations: epithelial, endothelial, fibroblasts and leukocytes [4].

Immunohistochemistry

Slices of paraffin-embedded tissue (4 μm thick) from 5 pairs of matched samples adjacent-mucosa tumor tissue were used. For antigen retrieval, the slides were boiled after deparaffinization in a pressure cooker for 10 minutes in citrated buffer (8.2 mM tri-sodium citrate and 1.98 mM citric acid, pH6) for Robo2 detection and in EDTA buffer (1 mM EDTA, 0.05% Tween-20, pH8) for Slit2 detection. Endogenous peroxidase was blocked with 3% H2O2 during 20 minutes. After blocking during 30 minutes with 1/5 dilution of goat serum, primary antibodies were incubated overnight at 4°C. Primary antibodies were rabbit polyclonal against Slit2 (Abcam, ab111128) and rabbit polyclonal against Robo2 (Prestige Antibodies, HPA013371), diluted both 1:100 in antibody diluent (Dako, Copenhagen, Denmark). Reaction was visualized using EnVision anti-rabbit antibody system, and developed using DAB-Plus Kit (Dako). Slides were counterstained with Harry’s modified haematoxylin. As negative control we used EnVision anti-rabbit antibody system and displayed no reactivity against any antigen.

Acknowledgements

We would like to thank Carmen Atencia, Pilar Medina, and Isabel Padrol for her expert assistance. This study was supported by the European Commission grant FP7-COOP-Health-2007-B HiPerDART. Also the Instituto de Salud Carlos III grants (FIS PI08-1635, PI09-01037 and FISPI11-01439), CIBERESP CB07/02/2005, the Spanish Association Against Cancer (AECC) Scientific Foundation, and the Catalan Government DURSI grant 2009SGR1489. Sample collection was supported by the Xarxa de Bancs de Tumors de Catalunya sponsored by Pla Director d’Oncología de Catalunya (XBTC).
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 credited. The Creative Commons Public Domain Dedication waiver ( https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​ ) applies to the data made available in this article, unless otherwise stated.

Competing interest

The authors have declared that no competing interests exist.

Authors’ contributions

RSP and VM designed the study, conceived the experiments and wrote the article. RSP and AB carried out the experiments. DC performed transcriptional networks. DGM performed inmunohistochemistry and helped to draft the manuscript. XSo, MCB, LPB, and EG analyzed data. CS, JO, XSa, FRM and RS provided samples and clinical data. All authors critically reviewed and had final approval of the article.
Anhänge

Electronic supplementary material

Literatur
1.
Zurück zum Zitat Hornberg JJ, Bruggeman FJ, Westerhoff HV, Lankelma J: Cancer: a Systems Biology disease. Biosystems. 2006, 83: 81-90. 10.1016/j.biosystems.2005.05.014CrossRefPubMed Hornberg JJ, Bruggeman FJ, Westerhoff HV, Lankelma J: Cancer: a Systems Biology disease. Biosystems. 2006, 83: 81-90. 10.1016/j.biosystems.2005.05.014CrossRefPubMed
2.
Zurück zum Zitat Hanahan D, Weinberg RA: Hallmarks of cancer: the next generation. Cell. 2011, 144: 646-674. 10.1016/j.cell.2011.02.013CrossRefPubMed Hanahan D, Weinberg RA: Hallmarks of cancer: the next generation. Cell. 2011, 144: 646-674. 10.1016/j.cell.2011.02.013CrossRefPubMed
3.
Zurück zum Zitat Berdiel-Acer M, Bohem ME, Lopez-Doriga A, Vidal A, Salazar R, Martinez-Iniesta M, Santos C, Sanjuan X, Villanueva A, Mollevi DG: Hepatic carcinoma-associated fibroblasts promote an adaptative response in colorectal cancer cells that inhibit proliferation and apoptosis: nonresistant cells die by nonapoptotic cell death. Neoplasia. 2011, 13: 931-946.PubMedCentralCrossRefPubMed Berdiel-Acer M, Bohem ME, Lopez-Doriga A, Vidal A, Salazar R, Martinez-Iniesta M, Santos C, Sanjuan X, Villanueva A, Mollevi DG: Hepatic carcinoma-associated fibroblasts promote an adaptative response in colorectal cancer cells that inhibit proliferation and apoptosis: nonresistant cells die by nonapoptotic cell death. Neoplasia. 2011, 13: 931-946.PubMedCentralCrossRefPubMed
4.
Zurück zum Zitat Calon A, Espinet E, Palomo-Ponce S, Tauriello DV, Iglesias M, Cespedes MV, Sevillano M, Nadal C, Jung P, Zhang XH, Byrom D, Riera A, Rossell D, Mangues R, Massague J, Sancho E, Batlle E: Dependency of colorectal cancer on a TGF-beta-Driven program in stromal cells for metastasis initiation. Cancer Cell. 2012, 22: 571-584. 10.1016/j.ccr.2012.08.013PubMedCentralCrossRefPubMed Calon A, Espinet E, Palomo-Ponce S, Tauriello DV, Iglesias M, Cespedes MV, Sevillano M, Nadal C, Jung P, Zhang XH, Byrom D, Riera A, Rossell D, Mangues R, Massague J, Sancho E, Batlle E: Dependency of colorectal cancer on a TGF-beta-Driven program in stromal cells for metastasis initiation. Cancer Cell. 2012, 22: 571-584. 10.1016/j.ccr.2012.08.013PubMedCentralCrossRefPubMed
5.
Zurück zum Zitat de la Cruz-Merino L, Henao Carrasco F, Vicente Baz D, Nogales Fernandez E, Reina Zoilo JJ, Codes Manuel de Villena M, Pulido EG: Immune microenvironment in colorectal cancer: a new hallmark to change old paradigms. Clin Dev Immunol. 2011, 2011: 174149PubMedCentralCrossRefPubMed de la Cruz-Merino L, Henao Carrasco F, Vicente Baz D, Nogales Fernandez E, Reina Zoilo JJ, Codes Manuel de Villena M, Pulido EG: Immune microenvironment in colorectal cancer: a new hallmark to change old paradigms. Clin Dev Immunol. 2011, 2011: 174149PubMedCentralCrossRefPubMed
6.
8.
Zurück zum Zitat Egeblad M, Nakasone ES, Werb Z: Tumors as organs: complex tissues that interface with the entire organism. Dev Cell. 2010, 18: 884-901. 10.1016/j.devcel.2010.05.012PubMedCentralCrossRefPubMed Egeblad M, Nakasone ES, Werb Z: Tumors as organs: complex tissues that interface with the entire organism. Dev Cell. 2010, 18: 884-901. 10.1016/j.devcel.2010.05.012PubMedCentralCrossRefPubMed
9.
Zurück zum Zitat Slaughter DP, Southwick HW, Smejkal W: Field cancerization in oral stratified squamous epithelium; clinical implications of multicentric origin. Cancer. 1953, 6: 963-968. 10.1002/1097-0142(195309)6:5<963::AID-CNCR2820060515>3.0.CO;2-QCrossRefPubMed Slaughter DP, Southwick HW, Smejkal W: Field cancerization in oral stratified squamous epithelium; clinical implications of multicentric origin. Cancer. 1953, 6: 963-968. 10.1002/1097-0142(195309)6:5<963::AID-CNCR2820060515>3.0.CO;2-QCrossRefPubMed
10.
Zurück zum Zitat Braakhuis BJ, Tabor MP, Kummer JA, Leemans CR, Brakenhoff RH: A genetic explanation of Slaughter's concept of field cancerization: evidence and clinical implications. Cancer Res. 2003, 63: 1727-1730.PubMed Braakhuis BJ, Tabor MP, Kummer JA, Leemans CR, Brakenhoff RH: A genetic explanation of Slaughter's concept of field cancerization: evidence and clinical implications. Cancer Res. 2003, 63: 1727-1730.PubMed
11.
Zurück zum Zitat Kitano H: Systems biology: a brief overview. Science. 2002, 295: 1662-1664. 10.1126/science.1069492CrossRefPubMed Kitano H: Systems biology: a brief overview. Science. 2002, 295: 1662-1664. 10.1126/science.1069492CrossRefPubMed
12.
Zurück zum Zitat Grizzi F, Bianchi P, Malesci A, Laghi L: Prognostic value of innate and adaptive immunity in colorectal cancer. World J Gastroenterol. 2013, 19: 174-184. 10.3748/wjg.v19.i2.174PubMedCentralCrossRefPubMed Grizzi F, Bianchi P, Malesci A, Laghi L: Prognostic value of innate and adaptive immunity in colorectal cancer. World J Gastroenterol. 2013, 19: 174-184. 10.3748/wjg.v19.i2.174PubMedCentralCrossRefPubMed
13.
Zurück zum Zitat Kipanyula MJ, Seke Etet PF, Vecchio L, Farahna M, Nukenine EN, Nwabo Kamdje AH: Signaling pathways bridging microbial-triggered inflammation and cancer. Cell Signal. 2013, 25: 403-416. 10.1016/j.cellsig.2012.10.014CrossRefPubMed Kipanyula MJ, Seke Etet PF, Vecchio L, Farahna M, Nukenine EN, Nwabo Kamdje AH: Signaling pathways bridging microbial-triggered inflammation and cancer. Cell Signal. 2013, 25: 403-416. 10.1016/j.cellsig.2012.10.014CrossRefPubMed
14.
Zurück zum Zitat Malfettone A, Silvestris N, Paradiso A, Mattioli E, Simone G, Mangia A: Overexpression of nuclear NHERF1 in advanced colorectal cancer: association with hypoxic microenvironment and tumor invasive phenotype. Exp Mol Pathol. 2012, 92: 296-303. 10.1016/j.yexmp.2012.03.004CrossRefPubMed Malfettone A, Silvestris N, Paradiso A, Mattioli E, Simone G, Mangia A: Overexpression of nuclear NHERF1 in advanced colorectal cancer: association with hypoxic microenvironment and tumor invasive phenotype. Exp Mol Pathol. 2012, 92: 296-303. 10.1016/j.yexmp.2012.03.004CrossRefPubMed
15.
Zurück zum Zitat Mojica W, Hawthorn L: Normal colon epithelium: a dataset for the analysis of gene expression and alternative splicing events in colon disease. BMC Genomics. 2010, 11: 5- 10.1186/1471-2164-11-5PubMedCentralCrossRefPubMed Mojica W, Hawthorn L: Normal colon epithelium: a dataset for the analysis of gene expression and alternative splicing events in colon disease. BMC Genomics. 2010, 11: 5- 10.1186/1471-2164-11-5PubMedCentralCrossRefPubMed
16.
Zurück zum Zitat Jothy S, Slesak B, Harlozinska A, Lapinska J, Adamiak J, Rabczynski J: Field effect of human colon carcinoma on normal mucosa: relevance of carcinoembryonic antigen expression. Tumour Biol. 1996, 17: 58-64. 10.1159/000217967CrossRefPubMed Jothy S, Slesak B, Harlozinska A, Lapinska J, Adamiak J, Rabczynski J: Field effect of human colon carcinoma on normal mucosa: relevance of carcinoembryonic antigen expression. Tumour Biol. 1996, 17: 58-64. 10.1159/000217967CrossRefPubMed
17.
Zurück zum Zitat Hawthorn L, Lan L, Mojica W: Evidence for Field Effect Cancerization in Colorectal Cancer. Genomics. 2013, 13: 00206-1. Hawthorn L, Lan L, Mojica W: Evidence for Field Effect Cancerization in Colorectal Cancer. Genomics. 2013, 13: 00206-1.
18.
Zurück zum Zitat Egeblad M, Rasch MG, Weaver VM: Dynamic interplay between the collagen scaffold and tumor evolution. Curr Opin Cell Biol. 2010, 22: 697-706. 10.1016/j.ceb.2010.08.015PubMedCentralCrossRefPubMed Egeblad M, Rasch MG, Weaver VM: Dynamic interplay between the collagen scaffold and tumor evolution. Curr Opin Cell Biol. 2010, 22: 697-706. 10.1016/j.ceb.2010.08.015PubMedCentralCrossRefPubMed
19.
Zurück zum Zitat Eferl R, Wagner EF: AP-1: a double-edged sword in tumorigenesis. Nat Rev Cancer. 2003, 3: 859-868. 10.1038/nrc1209CrossRefPubMed Eferl R, Wagner EF: AP-1: a double-edged sword in tumorigenesis. Nat Rev Cancer. 2003, 3: 859-868. 10.1038/nrc1209CrossRefPubMed
20.
Zurück zum Zitat Vesely PW, Staber PB, Hoefler G, Kenner L: Translational regulation mechanisms of AP-1 proteins. Mutat Res. 2009, 682: 7-12. 10.1016/j.mrrev.2009.01.001CrossRefPubMed Vesely PW, Staber PB, Hoefler G, Kenner L: Translational regulation mechanisms of AP-1 proteins. Mutat Res. 2009, 682: 7-12. 10.1016/j.mrrev.2009.01.001CrossRefPubMed
21.
Zurück zum Zitat Lu C, Hu X, Wang G, Leach LJ, Yang S, Kearsey MJ, Luo ZW: Why do essential proteins tend to be clustered in the yeast interactome network?. Mol Biosyst. 2010, 6: 871-877. 10.1039/b921069eCrossRefPubMed Lu C, Hu X, Wang G, Leach LJ, Yang S, Kearsey MJ, Luo ZW: Why do essential proteins tend to be clustered in the yeast interactome network?. Mol Biosyst. 2010, 6: 871-877. 10.1039/b921069eCrossRefPubMed
22.
Zurück zum Zitat Xu K, Bezakova I, Bunimovich L, Yi SV: Path lengths in protein-protein interaction networks and biological complexity. Proteomics. 2011, 11: 1857-1867. 10.1002/pmic.201000684CrossRefPubMed Xu K, Bezakova I, Bunimovich L, Yi SV: Path lengths in protein-protein interaction networks and biological complexity. Proteomics. 2011, 11: 1857-1867. 10.1002/pmic.201000684CrossRefPubMed
23.
Zurück zum Zitat Mehlen P, Delloye-Bourgeois C, Chedotal A: Novel roles for Slits and netrins: axon guidance cues as anticancer targets?. Nat Rev Cancer. 2011, 11: 188-197. 10.1038/nrc3005CrossRefPubMed Mehlen P, Delloye-Bourgeois C, Chedotal A: Novel roles for Slits and netrins: axon guidance cues as anticancer targets?. Nat Rev Cancer. 2011, 11: 188-197. 10.1038/nrc3005CrossRefPubMed
24.
Zurück zum Zitat Legg JA, Herbert JM, Clissold P, Bicknell R: Slits and Roundabouts in cancer, tumour angiogenesis and endothelial cell migration. Angiogenesis. 2008, 11: 13-21. 10.1007/s10456-008-9100-xCrossRefPubMed Legg JA, Herbert JM, Clissold P, Bicknell R: Slits and Roundabouts in cancer, tumour angiogenesis and endothelial cell migration. Angiogenesis. 2008, 11: 13-21. 10.1007/s10456-008-9100-xCrossRefPubMed
26.
Zurück zum Zitat Chen WF, Gao WD, Li QL, Zhou PH, Xu MD, Yao LQ: SLIT2 inhibits cell migration in colorectal cancer through the AKT-GSK3beta signaling pathway. Int J Colorectal Dis. 2013, 28: 933-940. 10.1007/s00384-013-1641-9CrossRefPubMed Chen WF, Gao WD, Li QL, Zhou PH, Xu MD, Yao LQ: SLIT2 inhibits cell migration in colorectal cancer through the AKT-GSK3beta signaling pathway. Int J Colorectal Dis. 2013, 28: 933-940. 10.1007/s00384-013-1641-9CrossRefPubMed
27.
Zurück zum Zitat Mazzarelli P, Pucci S, Spagnoli LG: CLU and colon cancer. The dual face of CLU: from normal to malignant phenotype. Adv Cancer Res. 2009, 105: 45-61.CrossRefPubMed Mazzarelli P, Pucci S, Spagnoli LG: CLU and colon cancer. The dual face of CLU: from normal to malignant phenotype. Adv Cancer Res. 2009, 105: 45-61.CrossRefPubMed
28.
Zurück zum Zitat Pucci S, Mazzarelli P, Nucci C, Ricci F, Spagnoli LG: CLU "in and out": looking for a link. Adv Cancer Res. 2009, 105: 93-113.CrossRefPubMed Pucci S, Mazzarelli P, Nucci C, Ricci F, Spagnoli LG: CLU "in and out": looking for a link. Adv Cancer Res. 2009, 105: 93-113.CrossRefPubMed
29.
Zurück zum Zitat Rodriguez-Pineiro AM, Garcia-Lorenzo A, Blanco-Prieto S, Alvarez-Chaver P, Rodriguez-Berrocal FJ, Cadena MP, Martinez-Zorzano VS: Secreted clusterin in colon tumor cell models and its potential as diagnostic marker for colorectal cancer. Cancer Invest. 2012, 30: 72-78. 10.3109/07357907.2011.630051CrossRefPubMed Rodriguez-Pineiro AM, Garcia-Lorenzo A, Blanco-Prieto S, Alvarez-Chaver P, Rodriguez-Berrocal FJ, Cadena MP, Martinez-Zorzano VS: Secreted clusterin in colon tumor cell models and its potential as diagnostic marker for colorectal cancer. Cancer Invest. 2012, 30: 72-78. 10.3109/07357907.2011.630051CrossRefPubMed
30.
Zurück zum Zitat Senturk A, Pfennig S, Weiss A, Burk K, Acker-Palmer A: Ephrin Bs are essential components of the Reelin pathway to regulate neuronal migration. Nature. 2011, 472: 356-360. 10.1038/nature09874CrossRefPubMed Senturk A, Pfennig S, Weiss A, Burk K, Acker-Palmer A: Ephrin Bs are essential components of the Reelin pathway to regulate neuronal migration. Nature. 2011, 472: 356-360. 10.1038/nature09874CrossRefPubMed
31.
Zurück zum Zitat Yuan Y, Chen H, Ma G, Cao X, Liu Z: Reelin is involved in transforming growth factor-beta1-induced cell migration in esophageal carcinoma cells. PLoS One. 2012, 7: e31802- 10.1371/journal.pone.0031802PubMedCentralCrossRefPubMed Yuan Y, Chen H, Ma G, Cao X, Liu Z: Reelin is involved in transforming growth factor-beta1-induced cell migration in esophageal carcinoma cells. PLoS One. 2012, 7: e31802- 10.1371/journal.pone.0031802PubMedCentralCrossRefPubMed
32.
Zurück zum Zitat Mathieu ME, Saucourt C, Mournetas V, Gauthereau X, Theze N, Praloran V, Thiebaud P, Boeuf H: LIF-dependent signaling: new pieces in the Lego. Stem Cell Rev. 2012, 8: 1-15.PubMedCentralCrossRefPubMed Mathieu ME, Saucourt C, Mournetas V, Gauthereau X, Theze N, Praloran V, Thiebaud P, Boeuf H: LIF-dependent signaling: new pieces in the Lego. Stem Cell Rev. 2012, 8: 1-15.PubMedCentralCrossRefPubMed
33.
Zurück zum Zitat Rockman SP, Demmler K, Roczo N, Cosgriff A, Phillips WA, Thomas RJ, Whitehead RH: Expression of interleukin-6, leukemia inhibitory factor and their receptors by colonic epithelium and pericryptal fibroblasts. J Gastroenterol Hepatol. 2001, 16: 991-1000. 10.1046/j.1440-1746.2001.02588.xCrossRefPubMed Rockman SP, Demmler K, Roczo N, Cosgriff A, Phillips WA, Thomas RJ, Whitehead RH: Expression of interleukin-6, leukemia inhibitory factor and their receptors by colonic epithelium and pericryptal fibroblasts. J Gastroenterol Hepatol. 2001, 16: 991-1000. 10.1046/j.1440-1746.2001.02588.xCrossRefPubMed
34.
Zurück zum Zitat Park JI, Strock CJ, Ball DW, Nelkin BD: The Ras/Raf/MEK/extracellular signal-regulated kinase pathway induces autocrine-paracrine growth inhibition via the leukemia inhibitory factor/JAK/STAT pathway. Mol Cell Biol. 2003, 23: 543-554. 10.1128/MCB.23.2.543-554.2003PubMedCentralCrossRefPubMed Park JI, Strock CJ, Ball DW, Nelkin BD: The Ras/Raf/MEK/extracellular signal-regulated kinase pathway induces autocrine-paracrine growth inhibition via the leukemia inhibitory factor/JAK/STAT pathway. Mol Cell Biol. 2003, 23: 543-554. 10.1128/MCB.23.2.543-554.2003PubMedCentralCrossRefPubMed
35.
Zurück zum Zitat Stempelj M, Kedinger M, Augenlicht L, Klampfer L: Essential role of the JAK/STAT1 signaling pathway in the expression of inducible nitric-oxide synthase in intestinal epithelial cells and its regulation by butyrate. J Biol Chem. 2007, 282: 9797-9804. 10.1074/jbc.M609426200CrossRefPubMed Stempelj M, Kedinger M, Augenlicht L, Klampfer L: Essential role of the JAK/STAT1 signaling pathway in the expression of inducible nitric-oxide synthase in intestinal epithelial cells and its regulation by butyrate. J Biol Chem. 2007, 282: 9797-9804. 10.1074/jbc.M609426200CrossRefPubMed
36.
Zurück zum Zitat Ager EI, Neo J, Christophi C: The renin-angiotensin system and malignancy. Carcinogenesis. 2008, 29: 1675-1684. 10.1093/carcin/bgn171CrossRefPubMed Ager EI, Neo J, Christophi C: The renin-angiotensin system and malignancy. Carcinogenesis. 2008, 29: 1675-1684. 10.1093/carcin/bgn171CrossRefPubMed
37.
Zurück zum Zitat Charles N, Ozawa T, Squatrito M, Bleau AM, Brennan CW, Hambardzumyan D, Holland EC: Perivascular nitric oxide activates notch signaling and promotes stem-like character in PDGF-induced glioma cells. Cell Stem Cell. 2010, 6: 141-152. 10.1016/j.stem.2010.01.001CrossRefPubMed Charles N, Ozawa T, Squatrito M, Bleau AM, Brennan CW, Hambardzumyan D, Holland EC: Perivascular nitric oxide activates notch signaling and promotes stem-like character in PDGF-induced glioma cells. Cell Stem Cell. 2010, 6: 141-152. 10.1016/j.stem.2010.01.001CrossRefPubMed
38.
Zurück zum Zitat Nomachi A, Nishita M, Inaba D, Enomoto M, Hamasaki M, Minami Y: Receptor tyrosine kinase Ror2 mediates Wnt5a-induced polarized cell migration by activating c-Jun N-terminal kinase via actin-binding protein filamin A. J Biol Chem. 2008, 283: 27973-27981. 10.1074/jbc.M802325200CrossRefPubMed Nomachi A, Nishita M, Inaba D, Enomoto M, Hamasaki M, Minami Y: Receptor tyrosine kinase Ror2 mediates Wnt5a-induced polarized cell migration by activating c-Jun N-terminal kinase via actin-binding protein filamin A. J Biol Chem. 2008, 283: 27973-27981. 10.1074/jbc.M802325200CrossRefPubMed
39.
Zurück zum Zitat Kani S, Oishi I, Yamamoto H, Yoda A, Suzuki H, Nomachi A, Iozumi K, Nishita M, Kikuchi A, Takumi T, Minami Y: The receptor tyrosine kinase Ror2 associates with and is activated by casein kinase Iepsilon. J Biol Chem. 2004, 279: 50102-50109. 10.1074/jbc.M409039200CrossRefPubMed Kani S, Oishi I, Yamamoto H, Yoda A, Suzuki H, Nomachi A, Iozumi K, Nishita M, Kikuchi A, Takumi T, Minami Y: The receptor tyrosine kinase Ror2 associates with and is activated by casein kinase Iepsilon. J Biol Chem. 2004, 279: 50102-50109. 10.1074/jbc.M409039200CrossRefPubMed
40.
Zurück zum Zitat Sanz-Pamplona R, Berenguer A, Cordero D, Riccadonna S, Sole X, Crous-Bou M, Guino E, Sanjuan X, Biondo S, Soriano A, Jurman G, Capella G, Furlanello C, Moreno V: Clinical value of prognosis gene expression signatures in colorectal cancer: a systematic review. PLoS One. 2012, 7: e48877- 10.1371/journal.pone.0048877PubMedCentralCrossRefPubMed Sanz-Pamplona R, Berenguer A, Cordero D, Riccadonna S, Sole X, Crous-Bou M, Guino E, Sanjuan X, Biondo S, Soriano A, Jurman G, Capella G, Furlanello C, Moreno V: Clinical value of prognosis gene expression signatures in colorectal cancer: a systematic review. PLoS One. 2012, 7: e48877- 10.1371/journal.pone.0048877PubMedCentralCrossRefPubMed
41.
Zurück zum Zitat Sanz-Pamplona R, Cordero D, Berenguer A, Lejbkowicz F, Rennert H, Salazar R, Biondo S, Sanjuan X, Pujana MA, Rozek L, Giordano TJ, Ben-Izhak O, Cohen HI, Trougouboff P, Bejhar J, Sova Y, Rennert G, Gruber SB, Moreno V: Gene expression differences between colon and rectum tumors. Clin Cancer Res. 2011, 17: 7303-7312. 10.1158/1078-0432.CCR-11-1570PubMedCentralCrossRefPubMed Sanz-Pamplona R, Cordero D, Berenguer A, Lejbkowicz F, Rennert H, Salazar R, Biondo S, Sanjuan X, Pujana MA, Rozek L, Giordano TJ, Ben-Izhak O, Cohen HI, Trougouboff P, Bejhar J, Sova Y, Rennert G, Gruber SB, Moreno V: Gene expression differences between colon and rectum tumors. Clin Cancer Res. 2011, 17: 7303-7312. 10.1158/1078-0432.CCR-11-1570PubMedCentralCrossRefPubMed
42.
Zurück zum Zitat Network CGA: Comprehensive molecular characterization of human colon and rectal cancer. Nature. 2012, 487: 330-337. 10.1038/nature11252CrossRef Network CGA: Comprehensive molecular characterization of human colon and rectal cancer. Nature. 2012, 487: 330-337. 10.1038/nature11252CrossRef
43.
Zurück zum Zitat Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U, Speed TP: Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics. 2003, 4: 249-264. 10.1093/biostatistics/4.2.249CrossRefPubMed Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U, Speed TP: Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics. 2003, 4: 249-264. 10.1093/biostatistics/4.2.249CrossRefPubMed
44.
45.
Zurück zum Zitat Planell N, Lozano JJ, Mora-Buch R, Masamunt MC, Jimeno M, Ordas I, Esteller M, Ricart E, Pique JM, Panes J, Salas A: Transcriptional analysis of the intestinal mucosa of patients with ulcerative colitis in remission reveals lasting epithelial cell alterations. Gut. 2013, 62: 967-976. 10.1136/gutjnl-2012-303333CrossRefPubMed Planell N, Lozano JJ, Mora-Buch R, Masamunt MC, Jimeno M, Ordas I, Esteller M, Ricart E, Pique JM, Panes J, Salas A: Transcriptional analysis of the intestinal mucosa of patients with ulcerative colitis in remission reveals lasting epithelial cell alterations. Gut. 2013, 62: 967-976. 10.1136/gutjnl-2012-303333CrossRefPubMed
46.
Zurück zum Zitat Uddin S, Ahmed M, Hussain A, Abubaker J, Al-Sanea N, AbdulJabbar A, Ashari LH, Alhomoud S, Al-Dayel F, Jehan Z: Genome-wide expression analysis of Middle Eastern colorectal cancer reveals FOXM1 as a novel target for cancer therapy. Am J Pathol. 2011, 178: 537-547. 10.1016/j.ajpath.2010.10.020PubMedCentralCrossRefPubMed Uddin S, Ahmed M, Hussain A, Abubaker J, Al-Sanea N, AbdulJabbar A, Ashari LH, Alhomoud S, Al-Dayel F, Jehan Z: Genome-wide expression analysis of Middle Eastern colorectal cancer reveals FOXM1 as a novel target for cancer therapy. Am J Pathol. 2011, 178: 537-547. 10.1016/j.ajpath.2010.10.020PubMedCentralCrossRefPubMed
47.
Zurück zum Zitat Foroushani AB, Brinkman FS, Lynn DJ: Pathway-GPS and SIGORA: identifying relevant pathways based on the over-representation of their gene-pair signatures. PeerJ. 2013, 1: e229PubMedCentralCrossRefPubMed Foroushani AB, Brinkman FS, Lynn DJ: Pathway-GPS and SIGORA: identifying relevant pathways based on the over-representation of their gene-pair signatures. PeerJ. 2013, 1: e229PubMedCentralCrossRefPubMed
49.
Zurück zum Zitat Nishimura D: A view from the web BioCarta. Biotech Software & Internet Report. 2004, 2: 117-120.CrossRef Nishimura D: A view from the web BioCarta. Biotech Software & Internet Report. 2004, 2: 117-120.CrossRef
50.
Zurück zum Zitat Schaefer CF, Anthony K, Krupa S, Buchoff J, Day M, Hannay T, Buetow KH: PID: the Pathway Interaction Database. Nucleic Acids Res. 2009, 37: D674-D679. 10.1093/nar/gkn653PubMedCentralCrossRefPubMed Schaefer CF, Anthony K, Krupa S, Buchoff J, Day M, Hannay T, Buetow KH: PID: the Pathway Interaction Database. Nucleic Acids Res. 2009, 37: D674-D679. 10.1093/nar/gkn653PubMedCentralCrossRefPubMed
51.
Zurück zum Zitat Yamamoto S, Sakai N, Nakamura H, Fukagawa H, Fukuda K, Takagi T: INOH: ontology-based highly structured database of signal transduction pathways. Database (Oxford). 2011, 2011: bar052 Yamamoto S, Sakai N, Nakamura H, Fukagawa H, Fukuda K, Takagi T: INOH: ontology-based highly structured database of signal transduction pathways. Database (Oxford). 2011, 2011: bar052
52.
Zurück zum Zitat Vastrik I, D'Eustachio P, Schmidt E, Gopinath G, Croft D, de Bono B, Gillespie M, Jassal B, Lewis S, Matthews L, Wu G, Birney E, Stein L: Reactome: a knowledge base of biologic pathways and processes. Genome Biol. 2007, 8: R39- 10.1186/gb-2007-8-3-r39PubMedCentralCrossRefPubMed Vastrik I, D'Eustachio P, Schmidt E, Gopinath G, Croft D, de Bono B, Gillespie M, Jassal B, Lewis S, Matthews L, Wu G, Birney E, Stein L: Reactome: a knowledge base of biologic pathways and processes. Genome Biol. 2007, 8: R39- 10.1186/gb-2007-8-3-r39PubMedCentralCrossRefPubMed
53.
Zurück zum Zitat Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES, Mesirov JP: Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 2005, 102: 15545-15550. 10.1073/pnas.0506580102PubMedCentralCrossRefPubMed Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES, Mesirov JP: Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 2005, 102: 15545-15550. 10.1073/pnas.0506580102PubMedCentralCrossRefPubMed
54.
Zurück zum Zitat Margolin AA, Nemenman I, Basso K, Wiggins C, Stolovitzky G, Dalla Favera R, Califano A: ARACNE: an algorithm for the reconstruction of gene regulatory networks in a mammalian cellular context. BMC Bioinformatics. 2006, 7 (Suppl 1): S7-10.1186/1471-2105-7-S1-S7.PubMedCentralCrossRefPubMed Margolin AA, Nemenman I, Basso K, Wiggins C, Stolovitzky G, Dalla Favera R, Califano A: ARACNE: an algorithm for the reconstruction of gene regulatory networks in a mammalian cellular context. BMC Bioinformatics. 2006, 7 (Suppl 1): S7-10.1186/1471-2105-7-S1-S7.PubMedCentralCrossRefPubMed
55.
Zurück zum Zitat Assenov Y, Ramirez F, Schelhorn SE, Lengauer T, Albrecht M: Computing topological parameters of biological networks. Bioinformatics. 2008, 24: 282-284. 10.1093/bioinformatics/btm554CrossRefPubMed Assenov Y, Ramirez F, Schelhorn SE, Lengauer T, Albrecht M: Computing topological parameters of biological networks. Bioinformatics. 2008, 24: 282-284. 10.1093/bioinformatics/btm554CrossRefPubMed
56.
Zurück zum Zitat Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T: Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003, 13: 2498-2504. 10.1101/gr.1239303PubMedCentralCrossRefPubMed Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T: Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003, 13: 2498-2504. 10.1101/gr.1239303PubMedCentralCrossRefPubMed
57.
Zurück zum Zitat Vaquerizas JM, Kummerfeld SK, Teichmann SA, Luscombe NM: A census of human transcription factors: function, expression and evolution. Nat Rev Genet. 2009, 10: 252-263. 10.1038/nrg2538CrossRefPubMed Vaquerizas JM, Kummerfeld SK, Teichmann SA, Luscombe NM: A census of human transcription factors: function, expression and evolution. Nat Rev Genet. 2009, 10: 252-263. 10.1038/nrg2538CrossRefPubMed
58.
Zurück zum Zitat Wingender E, Schoeps T, Donitz J: TFClass: an expandable hierarchical classification of human transcription factors. Nucleic Acids Res. 2013, 41: D165-D170. 10.1093/nar/gks1123PubMedCentralCrossRefPubMed Wingender E, Schoeps T, Donitz J: TFClass: an expandable hierarchical classification of human transcription factors. Nucleic Acids Res. 2013, 41: D165-D170. 10.1093/nar/gks1123PubMedCentralCrossRefPubMed
59.
Zurück zum Zitat Garcia-Garcia J, Guney E, Aragues R, Planas-Iglesias J, Oliva B: Biana: a software framework for compiling biological interactions and analyzing networks. BMC Bioinformatics. 2010, 11: 56- 10.1186/1471-2105-11-56PubMedCentralCrossRefPubMed Garcia-Garcia J, Guney E, Aragues R, Planas-Iglesias J, Oliva B: Biana: a software framework for compiling biological interactions and analyzing networks. BMC Bioinformatics. 2010, 11: 56- 10.1186/1471-2105-11-56PubMedCentralCrossRefPubMed
60.
Zurück zum Zitat Xenarios I, Fernandez E, Salwinski L, Duan XJ, Thompson MJ, Marcotte EM, Eisenberg D: DIP: The database of interacting proteins: 2001 update. Nucleic Acids Res. 2001, 29: 239-241. 10.1093/nar/29.1.239PubMedCentralCrossRefPubMed Xenarios I, Fernandez E, Salwinski L, Duan XJ, Thompson MJ, Marcotte EM, Eisenberg D: DIP: The database of interacting proteins: 2001 update. Nucleic Acids Res. 2001, 29: 239-241. 10.1093/nar/29.1.239PubMedCentralCrossRefPubMed
61.
Zurück zum Zitat Pagel P, Kovac S, Oesterheld M, Brauner B, Dunger-Kaltenbach I, Frishman G, Montrone C, Mark P, Stumpflen V, Mewes HW, Ruepp A, Frishman D: The MIPS mammalian protein-protein interaction database. Bioinformatics. 2005, 21: 832-834. 10.1093/bioinformatics/bti115CrossRefPubMed Pagel P, Kovac S, Oesterheld M, Brauner B, Dunger-Kaltenbach I, Frishman G, Montrone C, Mark P, Stumpflen V, Mewes HW, Ruepp A, Frishman D: The MIPS mammalian protein-protein interaction database. Bioinformatics. 2005, 21: 832-834. 10.1093/bioinformatics/bti115CrossRefPubMed
62.
Zurück zum Zitat Peri S, Navarro JD, Kristiansen TZ, Amanchy R, Surendranath V, Muthusamy B, Gandhi TK, Chandrika KN, Deshpande N, Suresh S, Rashmi BP, Shanker K, Padma N, Niranjan V, Harsha HC, Talreja N, Vrushabendra BM, Ramya MA, Yatish AJ, Joy M, Shivashankar HN, Kavitha MP, Menezes M, Choudhury DR, Ghosh N, Saravana R, Chandran S, Mohan S, Jonnalagadda CK, Prasad CK: Human protein reference database as a discovery resource for proteomics. Nucleic Acids Res. 2004, 32: D497-D501. 10.1093/nar/gkh070PubMedCentralCrossRefPubMed Peri S, Navarro JD, Kristiansen TZ, Amanchy R, Surendranath V, Muthusamy B, Gandhi TK, Chandrika KN, Deshpande N, Suresh S, Rashmi BP, Shanker K, Padma N, Niranjan V, Harsha HC, Talreja N, Vrushabendra BM, Ramya MA, Yatish AJ, Joy M, Shivashankar HN, Kavitha MP, Menezes M, Choudhury DR, Ghosh N, Saravana R, Chandran S, Mohan S, Jonnalagadda CK, Prasad CK: Human protein reference database as a discovery resource for proteomics. Nucleic Acids Res. 2004, 32: D497-D501. 10.1093/nar/gkh070PubMedCentralCrossRefPubMed
63.
Zurück zum Zitat Alfarano C, Andrade CE, Anthony K, Bahroos N, Bajec M, Bantoft K, Betel D, Bobechko B, Boutilier K, Burgess E, Cavero R, D'Abreo C, Donaldson I, Dorairajoo D, Dumontier MJ, Dumontier MR, Earles V, Farrall R, Feldman H, Garderman E, Gong Y, Gonzaga R, Grytsan V, Gryz E, Gu V, Haldorsen E, Halupa A, Haw R, Hrvojic A: The Biomolecular Interaction Network Database and related tools 2005 update. Nucleic Acids Res. 2005, 33: D418-D424.PubMedCentralCrossRefPubMed Alfarano C, Andrade CE, Anthony K, Bahroos N, Bajec M, Bantoft K, Betel D, Bobechko B, Boutilier K, Burgess E, Cavero R, D'Abreo C, Donaldson I, Dorairajoo D, Dumontier MJ, Dumontier MR, Earles V, Farrall R, Feldman H, Garderman E, Gong Y, Gonzaga R, Grytsan V, Gryz E, Gu V, Haldorsen E, Halupa A, Haw R, Hrvojic A: The Biomolecular Interaction Network Database and related tools 2005 update. Nucleic Acids Res. 2005, 33: D418-D424.PubMedCentralCrossRefPubMed
64.
Zurück zum Zitat Rual JF, Venkatesan K, Hao T, Hirozane-Kishikawa T, Dricot A, Li N, Berriz GF, Gibbons FD, Dreze M, Ayivi-Guedehoussou N, Klitgord N, Simon C, Boxem M, Milstein S, Rosenberg J, Goldberg DS, Zhang LV, Wong SL, Franklin G, Li S, Albala JS, Lim J, Fraughton C, Llamosas E, Cevik S, Bex C, Lamesch P, Sikorski RS, Vandenhaute J, Zoghbi HY: Towards a proteome-scale map of the human protein-protein interaction network. Nature. 2005, 437: 1173-1178. 10.1038/nature04209CrossRefPubMed Rual JF, Venkatesan K, Hao T, Hirozane-Kishikawa T, Dricot A, Li N, Berriz GF, Gibbons FD, Dreze M, Ayivi-Guedehoussou N, Klitgord N, Simon C, Boxem M, Milstein S, Rosenberg J, Goldberg DS, Zhang LV, Wong SL, Franklin G, Li S, Albala JS, Lim J, Fraughton C, Llamosas E, Cevik S, Bex C, Lamesch P, Sikorski RS, Vandenhaute J, Zoghbi HY: Towards a proteome-scale map of the human protein-protein interaction network. Nature. 2005, 437: 1173-1178. 10.1038/nature04209CrossRefPubMed
65.
Zurück zum Zitat Stelzl U, Worm U, Lalowski M, Haenig C, Brembeck FH, Goehler H, Stroedicke M, Zenkner M, Schoenherr A, Koeppen S, Timm J, Mintzlaff S, Abraham C, Bock N, Kietzmann S, Goedde A, Toksoz E, Droege A, Krobitsch S, Korn B, Birchmeier W, Lehrach H, Wanker EE: A human protein-protein interaction network: a resource for annotating the proteome. Cell. 2005, 122: 957-968. 10.1016/j.cell.2005.08.029CrossRefPubMed Stelzl U, Worm U, Lalowski M, Haenig C, Brembeck FH, Goehler H, Stroedicke M, Zenkner M, Schoenherr A, Koeppen S, Timm J, Mintzlaff S, Abraham C, Bock N, Kietzmann S, Goedde A, Toksoz E, Droege A, Krobitsch S, Korn B, Birchmeier W, Lehrach H, Wanker EE: A human protein-protein interaction network: a resource for annotating the proteome. Cell. 2005, 122: 957-968. 10.1016/j.cell.2005.08.029CrossRefPubMed
Metadaten
Titel
Aberrant gene expression in mucosa adjacent to tumor reveals a molecular crosstalk in colon cancer
verfasst von
Rebeca Sanz-Pamplona
Antoni Berenguer
David Cordero
David G Molleví
Marta Crous-Bou
Xavier Sole
Laia Paré-Brunet
Elisabet Guino
Ramón Salazar
Cristina Santos
Javier de Oca
Xavier Sanjuan
Francisco Rodriguez-Moranta
Victor Moreno
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2014
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/1476-4598-13-46

Weitere Artikel der Ausgabe 1/2014

Molecular Cancer 1/2014 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

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