Skip to main content
Erschienen in: BMC Cancer 1/2020

Open Access 01.12.2020 | Research article

Global targetome analysis reveals critical role of miR-29a in pancreatic stellate cell mediated regulation of PDAC tumor microenvironment

verfasst von: Shatovisha Dey, Sheng Liu, Tricia D. Factora, Solaema Taleb, Primavera Riverahernandez, Lata Udari, Xiaoling Zhong, Jun Wan, Janaiah Kota

Erschienen in: BMC Cancer | Ausgabe 1/2020

Abstract

Background

Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive forms of malignancies with a nearly equal incidence and mortality rates in patients. Pancreatic stellate cells (PSCs) are critical players in PDAC microenvironment to promote the aggressiveness and pathogenesis of the disease. Dysregulation of microRNAs (miRNAs) have been shown to play a significant role in progression of PDAC. Earlier, we observed a PSC-specific downregulation of miR-29a in PDAC pancreas, however, the mechanism of action of the molecule in PSCs is still to be elucidated. The current study aims to clarify the regulation of miR-29a in PSCs and identifies functionally important downstream targets that contribute to tumorigenic activities during PDAC progression.

Methods

In this study, using RNAseq approach, we performed transcriptome analysis of paired miR-29a overexpressing and control human PSCs (hPSCs). Enrichment analysis was performed with the identified differentially expressed genes (DEGs). miR-29a targets in the dataset were identified, which were utilized to create network interactions. Western blots were performed with the top miR-29a candidate targets in hPSCs transfected with miR-29a mimic or scramble control.

Results

RNAseq analysis identified 202 differentially expressed genes, which included 19 downregulated direct miR-29a targets. Translational repression of eight key pro-tumorigenic and -fibrotic targets namely IGF-1, COL5A3, CLDN1, E2F7, MYBL2, ITGA6 and ADAMTS2 by miR-29a was observed in PSCs. Using pathway analysis, we find that miR-29a modulates effectors of IGF-1-p53 signaling in PSCs that may hinder carcinogenesis. We further observe a regulatory role of the molecule in pathways associated with PDAC ECM remodeling and tumor-stromal crosstalk, such as INS/IGF-1, RAS/MAPK, laminin interactions and collagen biosynthesis.

Conclusions

Together, our study presents a comprehensive understanding of miR-29a regulation of PSCs, and identifies essential pathways associated with PSC-mediated PDAC pathogenesis. The findings suggest an anti-tumorigenic role of miR-29a in the context of PSC-cancer cell crosstalk and advocates for the potential of the molecule in PDAC targeted therapies.
Hinweise

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12885-020-07135-2.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ADAMTS2
ADAM Metallopeptidase With Thrombospondin Type 1 Motif 2
CLDN1
Claudin-1
COL5A3
Collagen alpha-3(V)
CPM
Counts per million
DEGs
Differentially expressed genes
E2F7
E2F Transcription Factor 7
ECM
Extracellular Matrix
EMT
Epithelial mesenchymal transition
FSTL1
Follistatin like 1
GO
Gene Ontology
IGF-1
Insulin-like growth factor 1
INS
Insulin
ITGA6
Integrin alpha-6
KEGG
Kyoto Encyclopedia of Genes and Genomes
LAMC1
Laminin subunit gamma 1
MAPK
Mitogen-activated protein kinase
miRNA
microRNA
MYBL2
MYB Proto-Oncogene like 2
PDAC
Pancreatic Ductal Adenocarcinoma
PSCs
Pancreatic stellate cells
qRT-PCR
Quantitative Real-time Polymerase Chain Reaction
STRING
Search Tool for the Retrieval of Interacting Genes/Proteins
UTR
Untranslated region

Background

Despite considerable advancement in the knowledge of pathogenesis and therapeutics of pancreatic ductal adenocarcinoma (PDAC) in recent years, the disease continues to remain as one of the deadliest malignancies. PDAC ranks as the seventh leading cause of cancer-related deaths worldwide [1] and the fourth in the United States [2]. This rapidly metastatic cancer is characterized by abundant desmoplastic reactions around pancreatic tumors mediated by the pancreatic stellate cells (PSCs) [35]. PSCs remain in quiescent state in normal pancreas, with a low extracellular-matrix (ECM) producing capacity. During pancreatic injury or inflammation, PSCs are activated by pro-inflammatory cytokines and growth factors to differentiate into myofibroblasts, expressing alpha smooth muscle actin (α-SMA) [3, 6, 7]. The transformed and activated stromal PSCs interact with the tumor cells, proliferate and produce ECM proteins and growth factors promoting fibrosis, pancreatitis and pancreatic cancer [4, 8, 9].
MicroRNAs (miRNAs) are a class of small (~ 22 nucleotide long), non-coding RNAs in multicellular organisms, which modulate key cellular mechanisms of proliferation, metabolism and apoptosis via post-transcriptional regulation of hundreds of genes [10]. miRNAs are initially generated as primary transcripts (pri-miRNA) from inter- and intragenic chromosomal regions predominantly via RNA polymerase II mediated transcription, and are then further processed by the Drosha RNase III enzyme to produce short hairpin pre-miRNAs [11]. Pre-miRNAs are exported to the cytoplasm by exportin 5, where they are further processed by the exonuclease III enzyme Dicer, in a complex, to generate mature miRNA. Mature miRNA, along with Agonaute 2, forms an RNA-dependent silencing complex and binds to the 3′-UTRs of the target gene mRNAs with imperfect complementarity to cause their degradation or translational suppression [11, 12]. Accumulating evidences have shown the involvement of miRNAs in regulation of pathological processes of variety of diseases including oncogenesis [1214]. Studies have further demonstrated the association of dysregulated miRNAs in stromal cells with progression of different types of cancer, including pancreatic cancer, indicating the potential of miRNAs in developing targeted therapies [1520].
In our previous work, we found microRNA-29a (miR-29a) to be pre-dominantly an anti-fibrotic molecule in PDAC, where miR-29a was significantly downregulated in activated PSCs and fibroblasts of murine and human PDAC as compared to normal pancreas, resulting in enhanced stromal extracellular matrix (ECM) deposition in PDAC microenvironment [21]. In addition, co-culture of pancreatic cancer cells with miR-29a overexpressing PSCs resulted in significant reduction in colony formation ability of the cancer cells and stromal deposition [21]. Thus, given the anti-fibrotic and tumor suppressive role of miR-29a in PSC-mediated PDAC progression, in the current study, we sought to decipher the mechanism of miR-29a in PSC regulation by identifying some of the key downstream target genes of the molecule, which also have critical functional implications in stromal remodeling and PDAC pathogenesis. Here we show for the first time that miR-29a concatenates genes belonging to key pathways associated with PDAC microenvironment, indicating the importance of the molecule in PSC-mediated PDAC stromal accumulation, suggestive of the potential of miR-29a as a therapeutic target for normalization of PDAC stroma.

Methods

Cell culture

Primary human pancreatic stellate cells (hPSCs) (3830, ScienCell Research Laboratories Carlsbad, California) were cultured in Dulbecco’s Modified Eagle Medium (DMEM, 11965092, Life Technologies, Carlsbad, CA) supplemented with 10% FBS in a humidified 5% CO2 incubator at 37 °C. hPSCs were authenticated using short tandem repeat profiling, and were regularly tested for mycoplasma contamination (MycoAlert, Lonza). All cells used in this study were less than passage 9.

Transfection

To overexpress miR-29a, hPSC cells were seeded at 1 X 105 cells/well in 6 well-plates for 24 h and transfected with control (CN-001000-01, GE Dharmacon, Lafeyette, CO) or miR-29a mimic (C-300504-07, GE Dharmacon, Lafeyette, CO) using DharmaFECT 1 Reagent (T-2001-01, GE Dharmacon, Lafeyette, CO) following manufacturer’s instructions. Total protein or RNA was isolated 48 h post-transfection for western blot or qPCR analyses, respectively.

RNA extraction

Total RNA from cultured cells were extracted using the RNeasy plus Mini kit (74,134, Qiagen, Venlo, Netherlands) following manufacturer’s protocol. The concentration and purity of the extracted RNAs were measured using a Nanodrop 2000 Spectrophotometer (Thermo Fisher Scientific, Carlsbad, CA).

RNAseq

For RNAseq, the quality and integrity of the extracted RNA were evaluated by a Bioanalyzer 2100 (Agilent technologies, CA). Samples with RNA Integrity Number (RIN) > 7.0 were used for RNAseq. cDNA libraries were prepared using the TruSeq RNA library kit (Illumina Inc., San Diego, CA). The libraries were amplified and then sequenced on an Illumina Hiseq.2000 instrument (San Diego, CA) with 100 bp paired end reads per sample. The quality of the sequence data was analyzed using FastQC [22]. The reads were mapped to the human genome (hg38) using STAR (v.2.5) [23]. Uniquely mapped sequencing reads were assigned to genes based on Gencode 25 using featureCounts (v1.6.2) [24]. Genes with read count per million (CPM) < 0.5 in two or more samples were filtered out and gene expression profiles were normalized using trimmed mean of M values (TMM) method. Differentially expressed genes (DEGs) were assessed by cutoff p-value of less than 0.05 after false discovery rate (FDR) adjustment with amplitude of fold change (FC) of gene expression greater than 2 linear FC.

Target prediction, functional enrichment and network analysis

Conserved miR-29a target genes were obtained using TargetScan (v7.1). The hypergeometric model was adopted to identify the overlap between DEGs and miR-29a predicted targets.
Functional enrichment analysis of the gene ontology (GO) terms and KEGG pathway analysis were performed using R package to investigate the biological functions and pathways of the identified genes. The protein-protein interaction networks of the genes were explored using the STRING database, version 11 [25].

Quantitative real time PCR (qRT-PCR)

RNA was reverse transcribed to cDNA using High capacity cDNA Reverse Transcription kit (4368814, Thermo Fisher Scientific, Carlsbad, CA) with random primers for genes or custom primer pool for miRNA (Thermo Fisher Scientific, Carlsbad, CA). To measure mature miR-29a expressions, TaqMan qRT-PCR reactions were set up using TaqMan Fast Advanced Mastermix (4444557, Applied Biosystems Foster City, CA) with TaqMan probe and primers for mature miR29a (002112, Applied Biosystems, Foster City, CA) or U6 snRNA (001973, Applied Biosystems, Foster City, CA). To assay the mRNA levels of genes, qRT-PCRs were performed with PowerUp SYBR Green Mastermix (A25742, Applied Biosystems, Foster City, CA) and custom primers Table S1). miRNA and mRNA qRT-PCR were normalized to U6 and ACTB respectively. Samples were run in triplicates in a 10 μl final volume using ABI 7500 Real-Time PCR machine with standard settings. Relative expressions were analyzed using ΔΔCT method.

Western blot

Protein lysates were prepared with RIPA Buffer (PI-89900, Thermo Fisher Scientific, Carlsbad, CA) and quantified using BCA Protein Assay Kit (23,225, Pierce Biotechnology, Waltham, CA). Equal amounts of total protein were loaded onto NuPAGE 4–12% Bis-Tris Gels (NP0323, Invitrogen, Carlsbad, CA). After electrophoresis, the gels were electrotransferred onto polyvinylidene fluoride membranes, blocked with 5% dry non-fat milk and incubated overnight at 4 °C with specific primary antibodies. The membranes were washed and then probed with corresponding HRP conjugated goat anti-mouse (31,430, Thermo Fisher Scientific, Carlsbad, CA) or goat anti-rabbit (31,460, Thermo Fisher Scientific, Carlsbad, CA) antibodies at 1:5000 dilution. To develop the blots, ECL detection kit (34,096, Thermo Fisher Scientific, Carlsbad, CA) was utilized and the images were captured on an Amersham Imager 600 (GE Healthcare, Chicago, IL). Densitometry analysis was performed using Image J software to quantify each protein band, which were then normalized against loading control GAPDH. The primary antibodies used in this study were anti-IGF-1 (ab9572, Abcam, Cambridge, MA), anti-COL5A3 (PA5–77257, Thermo Fisher Scientific, Carlsbad, CA), anti-E2F7 (ab56022, Abcam, Cambridge, MA), anti-MYBL2 (PA546845, Thermo Fisher Scientific, Carlsbad, CA), anti-ITGA6 (3750, Cell Signaling Technology, Danvers, MA), anti-CLDN1 (4933S, Cell Signaling Technology, Danvers, MA), anti-ADAMTS2 (3485, Cell Signaling Technology, Danvers, MA), and anti-GAPDH (MA5–15738, Thermo Fisher Scientific, Carlsbad, CA).

Statistical analysis

All data were expressed as mean ± standard error of the mean (SEM) of three independent experiments. Statistical analysis was performed by ANOVA or Student’s t test. Statistical significance is indicated as *p < 0.05 or **p < 0.01 or ***p< 0.001.

Results

RNAseq and identification of DEGs

RNAseq libraries were constructed using RNAs from control and miR-29a overexpressing hPSCs to generate global miR-29a targetome. Overexpression of miR-29a in the transfected hPSCs was verified by qPCR (Fig. 1a). Sequencing was performed with 2X 100 bp paired end reads. This yielded sequence reads ranging from 17 to 34 million pairs, of which 90–92% aligned to the hg19 genome assembly (Table 1). Quantile normalization with log2 transformation of number of counts per million (CPM) was performed and quality of raw sequencing reads and depth were verified for differential expression testing between the control and miR-29a overexpressing PSCs. For identification of DEGs, genes were plotted in a volcano plot by their log10 P values with FDR (q value) < 0.05 against log 2 fold change (FC) (Fig. 1b). This identified 90 downregulated and 106 upregulated genes with FDR < 0.05 and log FC < -1 or > + 1 respectively (Table S2). Next, inputting the DEG IDs into the TargetScan database, we identified 20 putative direct miR-29a targets among the identified DEGs- 19 of which were downregulated and one was upregulated (Fig. 1c). Among the downregulated miR-29a targets, IGF-1 exhibited the highest fold change, followed by COL5A3, E2F7, CLDN1, and MYBL2. DPYSL3 was the only upregulated target that met the screening criteria.
Table 1
RNA-Seq read counts and mapping statistics. Ctrl (Control) and miR-29a OE (overexpressing) represent hPSCs transfected with Control and miR-29a mimics respectively. R1, R2 and R3 are the three experimental replicates
Sample ID
Total Reads
Mapped Reads
Mapped High Quality Reads
Read Mapping Ratio
Percentage mapped to gene
hPSC Ctrl R1
38,372,969
35,691,075
35,333,674
92.07%
90.27
hPSC Ctrl R2
25,288,719
22,628,590
22,389,149
88.53%
90.09
hPSC Ctrl R3
18,759,093
16,958,204
16,698,047
89.01%
92.21
hPSC miR-29a-OE R1
36,899,783
34,288,605
33,965,913
92.05%
90.89
hPSC miR-29a-OE R2
20,971,993
18,594,180
18,318,771
87.35%
91.59
hPSC miR-29a-OE R3
33,726,399
29,480,256
29,128,167
86.37%
90.30

GO term enrichment and pathway analysis of downregulated genes

GO analysis of the DEGs with an FDR < 0.05 revealed that the downregulated (target and non-target) genes were significantly enriched in several PDAC relevant biological processes such as regulation of mitosis and cell cycle, cell migration and motility, cellular adhesion, cell proliferation, extracellular matrix organization and cytokine signaling (Table 2). Among the 19 miR-29a predicted downregulated target genes, IGF-1, CLDN1 and ITGA6 were enriched in regulation of cell motility/migration (Table 2). COL5A3, ADAMTS2, ITGA6, LAMC1 and IGF-1 associated with mechanisms of ECM remodeling. While ITGA6 and IGF-1 are negative regulators of apoptosis, E2F7 and MYBL2 contribute to the regulation of cell cycle (Tables 2 and 3). In addition, the pathways enriched for miR-29a overexpressing PSCs included IGF-1 signaling, Tp53 signaling, collagen pathway, integrin-laminin interactions, RAS/MAPK signaling and cytokine signaling as depicted in Table 3. Thus, the GO and pathway enrichment analyses indicate that miR-29a modulates effectors of signaling pathways associated with crucial mechanisms of ECM remodeling and tumor-stromal crosstalk, suggesting a potential role of the molecule in PSC-mediated regulation of PDAC tumor microenvironment (TME).
Table 2
Most relevant biological processes associated with downregulated genes in miR-29a overexpressing hPSCs
Biological Process
Gene Name
Ratio
p value
Positive regulation of cell proliferation
IGF1a; KIF14; IL1B; ESM 1; BCL2; KIF20B
6/490
0.024382
Cell division
CENPF; SPDL1; KIF14; SKA3; KIFC1; NEK2; SKA1; KIF18B; CENPE; CDCA5; KIF20B
11/346
4.69E-07
Regulation of G2/M transition of mitotic cell cycle
CENPF; KIF14; PLK4; NEK2; PLK1
5/80
3.2E-05
Negative regulator of extrinsic apoptotic pathway
ITGA6a; IGF1a
2/35
0.011085
Cell adhesion
LAMC1a; PCDH9a; PODXL; ITGA2; PCDH1; AJAP1
6/454
0.017486
Cell matrix adhesion
COL5A3a; ITGA6a; ADAMTS12; ITGA2
4/95
0.000932
Focal adhesion assembly
ITGA2; BCL2
2/27
0.006695
Positive regulation of fibroblast proliferation
IGF1a; E2F1
2/49
0.021029
Collagen fibril organization
COL5A3a; ADAMTS2a
2/46
0.018671
Extracellular matrix organization
COL5A3a; LAMC1a; ITGA6a; ITGA2; ABI3BP; PTX3
6/229
0.000624
Cell junction organization
ITGA6a;CLDN1a; LAMC1a; ITGA2
4/37
0.002963
Positive regulation of cell migration
CLDN1a; ITGA6a; IGF1a; PLAU; F2RL1; PODXL; LRRC15; IL1B
8/224
8.23E-06
Positive regulation of inflammatory response
ITGA2; IL1B
2/75
0.046007
Positive regulation of IL-6 secretion
F2RL1; IL1B
2/33
0.009895
a miR-29a direct targets
Table 3
Pathways enriched for downregulated genes in miR-29a overexpressing hPSCs
Pathway name
Genes
P value
Cell cycle
GINS1; PLK4; TOP2A; GINS2; BLM; CDCA5; PLK1; HJURP; CASC5; ESCO2; CENPA; AURKB; SKA1; CENPE; CENPF; EXO1; E2F1; E2F7a; NEK2; MYBL2a; SPDL1
R = 683; G = 21, p value = 2.07E-10
Tp53 pathway
BLM; EXO1; FANCD2; E2F1; SPDL1; E2F7a; AURKB
R = 259; G = 7; p value = 0.02074
Signaling by Ras mutants
NRASa; IQGAP3
R = 54; G = 2; p value=6.41E-04
IGF pathway
NRASa; LAMC1a; IGF1a; FSTL1a; PAPPA2
R = 127; G = 5; p value=0.032048
Laminin interactions
ITGA2; ITGA6a; LAMC1a
R = 31; G = 3; p value=0.003216579
Collagen binding
RC15; COL5A3a; ABI3BP; ITGA2; LRRC15
R = 53; G = 5; p value=0.00125
Collagen biosynthesis and metabolic pathway
COL5A3a; ADAMTS2a; ITGA2
R = 84; G = 3; p value=0.04578
R = the number of reference genes in the category; G = number of genes in the gene set for each category; a miR-29a direct targets

Validation analysis using qPCR and Western blots

Among the identified DEGs from the RNAseq, we selected all 19 down- and one upregulated miR-29a targets along with a subset of 24 additional DEGs to validate the RNAseq results using qRT-PCR. The expressions of 43 of the 44 tested genes well matched between the RNAseq and qPCR analyses (Table 4, Fig. 2a). Based on pathway analyses and available literature, IGF-1, COL5A3, CLDN1, E2F7, MYBL2, ITGA6 and ADAMTS2 were the most prominent miR-29a targets involved with one or more essential signaling mechanisms associated with TME regulation (Tables 2 and 3). Therefore, we next sought to find if miR-29a had a translational impact on these genes in PSCs. Our western blot analysis showed that protein levels of each of the seven selected targets were significantly diminished in miR-29a overexpressing PSCs (Fig. 2b). The most robust depletion was observed for ITGA6, followed by ADAMTS2 and IGF-1 respectively. All these three significantly downregulated target genes associate with ECM remodeling or fibrotic mechanisms. ITGA6 is a member of the integrin family that are heterodimer cell surface receptors comprising of α and β chains [26]. Alpha 6 containing integrins (α6/β4 and α6/β6) are the primary receptors for laminins, including laminin1 (LAMC1), a major ECM component [26]. Further, ECM in interaction with cellular integrins forms a scaffold, and plays essential role in cell proliferation, migration/invasion and survival [26]. ADAMTS2, belonging to the ADAM metallopeptidase with thrombospondin type 1 motif (ADAMTS) family, is responsible for processing of collagen type I, II, III and V precursors (pro-collagens) into mature collagen by excision of amino-propeptide, which is essential for generation of collagen monomers and assembly of mature collagen fibrils [27, 28]. Inhibition of ADAMTS2 has been shown to reduce stromal deposition and modulate TGF-β1 signaling [27, 29]. IGF-1 plays an essential role in fibrotic processes in different organs including pancreas, liver and lung [3032]. Recent reports demonstrate the association of IGF-1 in PSCs to promote stromal accumulation and basal growth rate in PDAC [33], as well as miR-29a-mediated regulation of the gene [34]. Interestingly, each of the seven tested targets have been shown to exhibit pro-tumorigenic effects. Together, the observations suggest an anti-fibrotic and tumor suppressive function of miR-29a in PSC mediated PDAC pathogenesis.
Table 4
qPCR validation of differentially expressed genes
Gene Symbol
RNAseq
qRT-PCR
logFC
p value
FDR
logFC
Downregulated
 IGF1a
−1.59
0.00
0.01
−1.48
 COL5A3a
−1.50
8.80E-07
0.00
−1.32
 CLDN1a
−1.49
0.00
0.01
−1.89
 E2F7a
−1.49
1.21E-06
0.00
−2.12
 MYBL2a
−1.35
1.42E-05
0.00
−1.92
 TET3a
−1.24
3.92E-05
0.00
−1.13
 PCDH9a
−1.2
3.02E-05
0.00
−1.18
 EMP1a
−1.19
4.09E-06
0.00
− 2.18
 ITGA6a
−1.18
0.00
0.012
−2.01
 XXYLT1a
−1.13
7.18E-05
0.00
−1.08
 BCL7Aa
−1.12
0.00
0.02
−1.80
 ADAMTS2a
−1.11
2.52E-05
0.00
−1.08
 DCLK3a
−1.10
0.00
0.02
−1.32
 LAMC1a
−1.09
9.16E-07
0.00
−1.32
 KIAA1549La
−1.07
1.43E-05
0.00
−1.12
 PRMT6a
−1.07
3.82E-05
0.00
−1.19
 KDELC1a
−1.03
6.87E-06
0.00
−1.83
 NRASa
−1.01
2.61E-05
0.00
−1.15
 FSTL1a
−1.0
1.16E-06
0.00
−2.3
 PPP1R14C
−3.54
0.00
0.02
−2.58
 ESM 1
−1.97
0.00
0.01
−1.41
 BCL2
−1.90
0.00
0.03
−1.30
 PLAU
−1.51
4.93E-07
0.00
−2.17
 IL1B
−1.26
9.42E-05
0.00
−2.88
 EXO1
−1.22
0.00
0.02
−1.62
 ITGA2
−1.10
4.23E-06
0.00
−2.17
 IQGAP3
−1.09
0.00
0.01
−1.20
 BLM
−1.06
0.00
0.01
−1.04
 E2F1
−1.03
0.00
0.02
−1.20
 AURKB
−1.02
5.01E-05
0.00
−1.92
Upregulated
 DPYSL3a
1.09
3.21E-06
0.00
1.01
 PYGM
3.58
0.00
0.01
3.64
 CXCL5
2.40
5.34E-07
0.00
1.98
 NEFL
1.98
0.00
0.02
1.26
 GNAO1
1.82
0.00
0.03
1.44
 TNFRSF10C
1.62
0.00
0.03
1.20
 HLA-DMA
1.51
0.00
0.03
2.06
 ITGA7
1.46
0.00
0.02
1.65
 FBXO32
1.41
1.06E-05
0.00
1.31
 PIK3AP1
1.31
8.41E-05
0.00
1.53
 HERC6
1.15
0.00
0.03
1.04
HIST1H1C
1.12
0.00
0.04
−1.18
 IGFBP3
1.06
5.82E-06
0.00
0.89
 HIST2H2BE
1.03
0.00
0.018
0.65
amiR-29a direct targets

Network interactions of the downregulated miR-29a targets

To determine if the identified downregulated miR-29a direct target genes formed a network of interactions, we next analyzed the genes utilizing the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database. We included a few additional nodes to construct the network. We observed three distinct networks in the interactome, which consisted of insulin/IGF, RAS/MAPK and laminin signaling pathways (Fig. 3).
IGF-1, belonging to the IGF family members, is one of the key regulators of the insulin/IGF pathway. IGF-1 is a direct downregulated miR-29a target in our dataset, which interacts with other effectors of the pathway including IGF-1R, INSR, IGFBP4 IGFBP5 and FSTL1 (Fig. 3). Interestingly, one of the oncogenes PTPN1 in the pathway is also a predicted direct miR-29a target, however, our RNAseq data did not show differential expression for this gene with miR-29a overexpression, which could be an effect specific to the PSCs. Nonetheless, the insulin/IGF signaling is a key driver in tumor-stromal interactions, metastasis and PDAC progression [33]. IGF-1 secreted by activated PSCs and fibroblasts in PDAC stroma via IGF-1 receptor (IGF-1R) promote cancer cell migration, invasion and metastasis [33, 35]. In fact, the RAS/MAPK pathway identified in our study consisted of interactions of IGF-1 and IGF-1R with other genes in the pathway including NRAS, HRAS, KRAS, SOS1 and RAF1. It is well documented that the MAPK signaling cascade bridges the crosstalk between ECM-mediated extracellular signaling through growth factors and their receptors such as IGF-1/IGF-1R, and subsequent intracellular response to allow cancer cell proliferation and migration [36]. IGF-1 bound activated IGF-1R phosphorylates insulin receptor substrates (such as IRS1, IRS2 and Shc). The Src homology 2 (SH2) domains of these substrates are recognized by signaling molecules to activate the intracellular effectors such as RAS, RAF and SOS and the RAS/MAPK pathway [37, 38]. Interestingly, in our previous study, we observed significant downregulation of NRAS with miR-29a overexpression in PDAC cell lines [39]. In the current study, miR-29a overexpression also resulted in moderate downregulation of NRAS in PSCs (logFC = − 1.01), however the role of NRAS in PSCs is unknown. Nonetheless, it is apparent that miR-29a modulates extracellular IGF-1/IGF-1R signaling in PSCs, and intracellular NRAS expression in pancreatic cancer cells, which indicates a functional role of the molecule in tumor-stromal crosstalk via insulin/IRF -RAS/MAPK signaling mechanism in PDAC.
The identified interactome further consisted of three miR-29a targets namely ITGA6, LAMC1 and FSTL1 that associate with laminin interactions, which are salient to pancreatic ECM and desmoplasia [4042]. LAMC1 encodes for laminin γ1 chain isoform, which are essential non-collagenous ECM glycoproteins, integral to basement membrane assembly and crucial for intra- and extracellular communication to modulate cellular behavior [43]. Laminin interactions, including that of LAMC1, have been shown to promote oncogenesis via processes including cancer cell migration, differentiation and metastasis [4447]. Cytoplasmic laminin expression correlates with poor patient prognosis in pancreatic cancer [48] and has been shown as one of the most efficient ECM proteins to promote cell adhesion-mediated drug resistance [49]. Further, ECM-integrin interactions are found to be crucial for adhesion-mediated drug and resistance to chemotherapy [50, 51].

Discussion

In our previous studies, we observed significant loss of miR-29a in several PDAC cell lines [21, 39]. In addition, miR-29a was globally repressed in PDAC tumor tissues, as well as in a PSC- and epithelial cell- specific manner [21]. We further demonstrated that TGF-β1 via SMAD3 signaling negatively regulates miR-29a expression in PSCs and upregulates several ECM proteins including collagens, laminin and fibronectin [21]. In the current study, using RNAseq, we characterize the mechanism and pathway interactions by which miR-29a contributes to PSC-mediated regulation of ECM and tumor-stromal crosstalk. This will allow for a comprehensive understanding of the therapeutic applicability of the molecule in the context of PDAC stroma.
RNAseq analysis with miR-29a overexpressing PSCs and controls identified a number of DEGs, which included predicted direct and indirect targets of the molecule. Because miRNAs primarily regulate genes either by mRNA decay or translational repression, we focused on the direct targets that were downregulated with miR-29a overexpression. We validated the translational repression of the targets namely IGF-1, COL5A3, CLDN1, E2F7, MYBL2, which exhibited the highest fold changes in the RNAseq dataset, along with ITGA6 and ADAMTS2, which had functional relevance in stromal regulation. Our western blot analysis indicated the highest repression of ITGA6, ADAMTS2 and IGF-1 protein levels with miR-29a overexpression in PSCs (Fig. 2b). Among these identified direct targets, association of IGF-1 and COL5A3 with PSCs in PDAC has been reported previously [33, 52]. Network analysis with the targets identified three overlapping pathways related to IGF, RAS/MAPK signaling and laminin interactions. IGF-1 secreted by activated PSCs and CAFs via sonic hedgehog pathway activates IGF-1R in cancer cells triggering phosphorylation of insulin-receptor or Src substrates to promote PDAC metastasis via intracellular pathways such as RAS/MAPK [37, 53]. In addition, high IGF-1 with low IGFBP3 expressions associated with enhanced risks for PDAC [54]. Expectedly, patients with advance clinical stages (II and III) of PDAC had higher levels of IGF-1R and low IGFBP3, and exhibited poor prognosis [54]. Interestingly, the IGF-1R expressions in these patients associated with high stromal abundance, suggesting the regulation of tumor-stromal crosstalk via IGF/IGF-1R signaling [54]. Another identified miR-29a target CLDN1 is a tight junction protein that facilitates cell-ECM communication and EMT in various cancer types [5557]. The gene is shown to be a contributor in tumor-stroma crosstalk in pancreatic cancer [58]. Although the regulation of CLDN1 in PSCs has not been reported previously, studies have shown the gene to be under the regulation of IGF-1 signaling [59, 60]. Upregulation of collagens, including COL5A3, is a salient feature of fibrosis and malignant tumor stroma, including that in PDAC [52, 61, 62]. Collagens are abundantly expressed in PDAC ECM; and collagen V, by binding with α2β1 integrin receptors, stimulates migration, proliferation and metastasis in PDAC [63]. Interestingly, ADAMTS2, another identified miR-29a downregulated target, primarily functions to process collagens I, II, III and V precursors into mature molecules [27, 28]. The gene promotes fibrosis via activation of TGF-β signaling [64]. Evidently, miR-29a plays an anti-fibrotic role in PDAC by influencing ECM deposition via modulation of multiple targets in the collagen pathway. In addition to these genes that directly regulate tumor-microenvironment and desmoplasia, the top targets identified from our dataset consisted of the two additional genes E2F7 and MYBL2, which play essential roles in cell cycle regulation. E2F7 associates with poor patient outcome in several types of cancer including PDAC [6567] and has been shown essential for mouse embryonic survival [68]. Inhibition of E2F7 enhanced G1 phase percentage in prostate cancer reducing cellular proliferation [67]. Similarly, MYBL2 is a transcription factor which promotes cell proliferation and differentiation by fostering cell cycle entry into S and M phases, and is dysregulated in types of cancer [39, 69, 70]. A recent study demonstrated the regulatory role of MYBL2 in promoting PDAC desmoplasia and PSCs’ growth through sonic hedgehog and adrenomedullin via paracrine and autocrine signaling [71], however the role of the gene in PSCs has not been reported. A negative feedback regulatory mechanism between miR-29a and MYBL2 influencing the activation of PSCs is possible, but this requires future validation. Nonetheless, the identified set of miR-29a target genes exhibit a pro-fibrotic and tumorigenic function in PDAC desmoplasia and progression via multiple targeted pathways, although PSC-specific function of some of the identified target genes such as E2F7, CLDN1, MYBL2 and ADAMTS2 has not been studied previously. Together, the observations in the current study signify that overexpression of miR-29a may lead to inhibition of PSC-induced pro-fibrotic and desmoplastic effects by targeting these genes to impair signaling mechanisms such as sonic hedgehog, IGF, RAS/MAPK, collagen metabolism and laminin pathways, and perturbing their normal cellular responses to promote PDAC progression.
As mentioned above, IGF-1 signaling axis is a key mechanism that promotes PDAC tumor-stromal crosstalk and drug resistance. In our RNAseq dataset, we observed the most robust downregulation of the IGF-1 gene among all miR-29a targets. It is possible that in addition to IGF-1 alone, miR-29a regulates IGF-signaling via modulating multiple components of the pathway in PSCs, such as indirect regulation of genes including IGF-1R, INSR and direct targeting of some others. It is worthy to note that MYBL2 and E2F7 are miR-29a targets that are at the functional convergence of p53-IGF-1 pathways. Stromal p53 has been implicated as a key component that reprograms activated pancreatic and hepatic stellate cells to transform them into quiescent states [72, 73]. Depletion of p53 in stromal cells caused faster and more aggressive tumor development with enhanced invasion and metastasis of cancer cells, suggesting a paracrine mechanism of p53 in tumor progression [74, 75]. In addition, studies have reported the occurrence of inactivating p53 mutations in fibroblastic stromal cells and their association in promoting tumor progression and cancer cell metastasis in types of carcinogenesis [74], although the molecular mechanisms are still unclear. MYBL2 is a downstream effector of the p53 pathway [69]. With p53 mutations, MYBL2 repression is uncoupled allowing enhanced binding of the molecule with MuvB and FOXM1 leading to activation of mitotic genes [69, 76]. FOXM1 is an essential component of Akt signaling, which functions both in the context of tumor stroma and cancer cells to promote tumorigenesis [7780]. Interestingly, Akt pathway is under inverse regulation of IGF-1 signaling [79, 81, 82]. Similarly, E2F7 is a crucial transcription factor, which promotes E2F1-p53 dependent apoptosis and cell-cycle arrest [68, 83]. In our RNAseq data with miR-29a overexpressing PSCs, we found E2F1 as one of the indirect downregulated targets. In addition, E2F7 has also been shown to be activated by Akt signaling in carcinomas [8385]. Although the exact mechanisms of MYBL2 and E2F7 in PSCs is still to be understood, our results suggest that dysregulation of miR-29a in PSCs derepresses genes such as IGF-1, MYBL2 and E2F7, which may in turn disrupt stromal p53 regulation, promoting PSC-mediated tumor proliferation.
GO analysis showed that the direct and indirect miR-29a downregulated targets were enriched in crucial cellular and molecular functions associated with PDAC stromal remodeling and proliferation. The biological processes consisted of those related to cell cycle regulation, collagen formation, ECM organization and immune signaling (Table 2). Our study further identified inter-connected networks comprising of essential pathways in PDAC stromal regulation and desmoplasia (Table 3). Although a single miRNA is known to target hundreds of genes, resulting in their post- transcriptional repression, based on the functional network of the differentially expressed targets, the predominant phenotypic effect of a miRNA can be systematically analyzed in a context-specific manner. Our analysis using PSCs identifies a number of miR-29a target genes that are crucial players in PDAC stromal remodeling and tumor-stromal crosstalk, suggesting the importance of the molecule in their pathway regulations to modulate PDAC microenvironment and tumor progression.

Conclusion

The current study is the first to use RNAseq platform for a comprehensive characterization of the PSC transcriptome under the regulation of miR-29a. In PDAC, activated PSCs foster cancer cell migration via desmoplastic reaction characterized by increased collagen, laminin and other ECM deposition resulting in fibrosis. Our data identified altered expressions of a number of novel genes under miR-29a regulation, including IGF-1, COL5A3, CLDN1, E2F7, MYBL2, ITGA6, ADAMTS2, and related pathways such as insulin-IRF, RAS/MAPK, laminin and collagen pathways in PSCs that are dysregulated or associate with PDAC tumor-stromal crosstalk and ECM remodeling. Given the functional relationship among the identified miR-29a targets in our PSCs dataset, it is likely that restoration of miR-29a in PSCs will dwindle or escalate the interconnected tumor-suppressive/pro-tumorigenic networks respectively in PDAC microenvironment, causing global regulation of the network functions to hinder the disease progression. Since our conclusions are primarily based on computational analysis, future investigations aimed to delineate the mechanistic relationship of miR-29a, its targets and related pathways in PSCs as well as cancer cells, would allow for a deeper comprehension of the associated pathological changes in tumor-stromal crosstalk in PDAC. This would be essential to assess the therapeutic modalities of miR-29a and its target networks in the disease. Nonetheless, our data in the current report identifies novel genes and networks under the regulation of miR-29a in PSCs, bolstering an anti-tumorigenic function of the molecule in the context of PDAC stroma. These findings suggest that targeted upregulation of miR-29a may hold great therapeutic value in efficacious PDAC treatment.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12885-020-07135-2.

Acknowledgements

We thank the Collaborative Core for Cancer Bioinformatics, where the RNAseq was performed, shared by IU Simon Cancer Center (P30CA082709) and Purdue University Center for Cancer Research (P30CA023168) with support from the Walther Cancer Foundation. We are also thankful to the Indiana University Precision Health Initiative for their support.
The cell line used in this study is purchased commercially from ScienCell Research Laboratories in compliance with ethical and regulatory guidelines.
Not applicable.

Competing interests

The authors declare no potential conflicts of interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.PubMed Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.PubMed
3.
Zurück zum Zitat Apte MV, Park S, Phillips PA, Santucci N, Goldstein D, Kumar RK, Ramm GA, Buchler M, Friess H, McCarroll JA, et al. Desmoplastic reaction in pancreatic cancer: role of pancreatic stellate cells. Pancreas. 2004;29(3):179–87.PubMed Apte MV, Park S, Phillips PA, Santucci N, Goldstein D, Kumar RK, Ramm GA, Buchler M, Friess H, McCarroll JA, et al. Desmoplastic reaction in pancreatic cancer: role of pancreatic stellate cells. Pancreas. 2004;29(3):179–87.PubMed
4.
Zurück zum Zitat Bachem MG, Schunemann M, Ramadani M, Siech M, Beger H, Buck A, Zhou S, Schmid-Kotsas A, Adler G. Pancreatic carcinoma cells induce fibrosis by stimulating proliferation and matrix synthesis of stellate cells. Gastroenterology. 2005;128(4):907–21.PubMed Bachem MG, Schunemann M, Ramadani M, Siech M, Beger H, Buck A, Zhou S, Schmid-Kotsas A, Adler G. Pancreatic carcinoma cells induce fibrosis by stimulating proliferation and matrix synthesis of stellate cells. Gastroenterology. 2005;128(4):907–21.PubMed
5.
Zurück zum Zitat Kota J, Hancock J, Kwon J, Korc M. Pancreatic cancer: Stroma and its current and emerging targeted therapies. Cancer Lett. 2017;391:38–49.PubMed Kota J, Hancock J, Kwon J, Korc M. Pancreatic cancer: Stroma and its current and emerging targeted therapies. Cancer Lett. 2017;391:38–49.PubMed
6.
Zurück zum Zitat Apte MV, Pirola RC, Wilson JS. Pancreatic stellate cells: a starring role in normal and diseased pancreas. Front Physiol. 2012;3:344.PubMedPubMedCentral Apte MV, Pirola RC, Wilson JS. Pancreatic stellate cells: a starring role in normal and diseased pancreas. Front Physiol. 2012;3:344.PubMedPubMedCentral
7.
Zurück zum Zitat Bachem MG, Schneider E, Gross H, Weidenbach H, Schmid RM, Menke A, Siech M, Beger H, Grunert A, Adler G. Identification, culture, and characterization of pancreatic stellate cells in rats and humans. Gastroenterology. 1998;115(2):421–32.PubMed Bachem MG, Schneider E, Gross H, Weidenbach H, Schmid RM, Menke A, Siech M, Beger H, Grunert A, Adler G. Identification, culture, and characterization of pancreatic stellate cells in rats and humans. Gastroenterology. 1998;115(2):421–32.PubMed
8.
Zurück zum Zitat Masamune A, Watanabe T, Kikuta K, Shimosegawa T. Roles of pancreatic stellate cells in pancreatic inflammation and fibrosis. Clin Gastroenterol Hepatol. 2009;7(11 Suppl):S48–54.PubMed Masamune A, Watanabe T, Kikuta K, Shimosegawa T. Roles of pancreatic stellate cells in pancreatic inflammation and fibrosis. Clin Gastroenterol Hepatol. 2009;7(11 Suppl):S48–54.PubMed
9.
Zurück zum Zitat Farran B, Nagaraju GP. The dynamic interactions between the stroma, pancreatic stellate cells and pancreatic tumor development: novel therapeutic targets. Cytokine Growth Factor Rev. 2019;48:11–23.PubMed Farran B, Nagaraju GP. The dynamic interactions between the stroma, pancreatic stellate cells and pancreatic tumor development: novel therapeutic targets. Cytokine Growth Factor Rev. 2019;48:11–23.PubMed
10.
Zurück zum Zitat Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281–97.PubMed Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281–97.PubMed
11.
Zurück zum Zitat Fabian MR, Sonenberg N, Filipowicz W. Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem. 2010;79:351–79.PubMed Fabian MR, Sonenberg N, Filipowicz W. Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem. 2010;79:351–79.PubMed
12.
13.
Zurück zum Zitat Couzin J. MicroRNAs make big impression in disease after disease. Science. 2008;319(5871):1782–4.PubMed Couzin J. MicroRNAs make big impression in disease after disease. Science. 2008;319(5871):1782–4.PubMed
14.
Zurück zum Zitat Rupaimoole R, Slack FJ. MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov. 2017;16(3):203–22.PubMed Rupaimoole R, Slack FJ. MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov. 2017;16(3):203–22.PubMed
15.
Zurück zum Zitat Kadera BE, Li L, Toste PA, Wu N, Adams C, Dawson DW, Donahue TR. MicroRNA-21 in pancreatic ductal adenocarcinoma tumor-associated fibroblasts promotes metastasis. PLoS One. 2013;8(8):e71978.PubMedPubMedCentral Kadera BE, Li L, Toste PA, Wu N, Adams C, Dawson DW, Donahue TR. MicroRNA-21 in pancreatic ductal adenocarcinoma tumor-associated fibroblasts promotes metastasis. PLoS One. 2013;8(8):e71978.PubMedPubMedCentral
16.
Zurück zum Zitat Ali S, Suresh R, Banerjee S, Bao B, Xu Z, Wilson J, Philip PA, Apte M, Sarkar FH. Contribution of microRNAs in understanding the pancreatic tumor microenvironment involving cancer associated stellate and fibroblast cells. Am J Cancer Res. 2015;5(3):1251–64.PubMedPubMedCentral Ali S, Suresh R, Banerjee S, Bao B, Xu Z, Wilson J, Philip PA, Apte M, Sarkar FH. Contribution of microRNAs in understanding the pancreatic tumor microenvironment involving cancer associated stellate and fibroblast cells. Am J Cancer Res. 2015;5(3):1251–64.PubMedPubMedCentral
17.
Zurück zum Zitat Kim JE, Kim BG, Jang Y, Kang S, Lee JH, Cho NH. The stromal loss of miR-4516 promotes the FOSL1-dependent proliferation and malignancy of triple negative breast cancer. Cancer Lett. 2020;469:256–65.PubMed Kim JE, Kim BG, Jang Y, Kang S, Lee JH, Cho NH. The stromal loss of miR-4516 promotes the FOSL1-dependent proliferation and malignancy of triple negative breast cancer. Cancer Lett. 2020;469:256–65.PubMed
18.
Zurück zum Zitat Qin X, Guo H, Wang X, Zhu X, Yan M, Wang X, Xu Q, Shi J, Lu E, Chen W, et al. Exosomal miR-196a derived from cancer-associated fibroblasts confers cisplatin resistance in head and neck cancer through targeting CDKN1B and ING5. Genome Biol. 2019;20(1):12.PubMedPubMedCentral Qin X, Guo H, Wang X, Zhu X, Yan M, Wang X, Xu Q, Shi J, Lu E, Chen W, et al. Exosomal miR-196a derived from cancer-associated fibroblasts confers cisplatin resistance in head and neck cancer through targeting CDKN1B and ING5. Genome Biol. 2019;20(1):12.PubMedPubMedCentral
19.
Zurück zum Zitat Taddei ML, Cavallini L, Ramazzotti M, Comito G, Pietrovito L, Morandi A, Giannoni E, Raugei G, Chiarugi P. Stromal-induced downregulation of miR-1247 promotes prostate cancer malignancy. J Cell Physiol. 2019;234(6):8274–85.PubMed Taddei ML, Cavallini L, Ramazzotti M, Comito G, Pietrovito L, Morandi A, Giannoni E, Raugei G, Chiarugi P. Stromal-induced downregulation of miR-1247 promotes prostate cancer malignancy. J Cell Physiol. 2019;234(6):8274–85.PubMed
20.
Zurück zum Zitat Eichelmann AK, Matuszcak C, Hummel R, Haier J. Role of miRNAs in cell signaling of cancer associated fibroblasts. Int J Biochem Cell Biol. 2018;101:94–102.PubMed Eichelmann AK, Matuszcak C, Hummel R, Haier J. Role of miRNAs in cell signaling of cancer associated fibroblasts. Int J Biochem Cell Biol. 2018;101:94–102.PubMed
21.
Zurück zum Zitat Kwon JJ, Nabinger SC, Vega Z, Sahu SS, Alluri RK, Abdul-Sater Z, Yu Z, Gore J, Nalepa G, Saxena R, et al. Pathophysiological role of microRNA-29 in pancreatic cancer stroma. Sci Rep. 2015;5:11450.PubMedPubMedCentral Kwon JJ, Nabinger SC, Vega Z, Sahu SS, Alluri RK, Abdul-Sater Z, Yu Z, Gore J, Nalepa G, Saxena R, et al. Pathophysiological role of microRNA-29 in pancreatic cancer stroma. Sci Rep. 2015;5:11450.PubMedPubMedCentral
23.
Zurück zum Zitat Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, Gingeras TR. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29(1):15–21.PubMed Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, Gingeras TR. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29(1):15–21.PubMed
24.
Zurück zum Zitat Liao Y, Smyth GK. Shi W: featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014;30(7):923–30.PubMed Liao Y, Smyth GK. Shi W: featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014;30(7):923–30.PubMed
25.
Zurück zum Zitat Jensen LJ, Kuhn M, Stark M, Chaffron S, Creevey C, Muller J, Doerks T, Julien P, Roth A, Simonovic M, et al. STRING 8--a global view on proteins and their functional interactions in 630 organisms. Nucleic Acids Res. 2009;37(Database issue):D412–6.PubMed Jensen LJ, Kuhn M, Stark M, Chaffron S, Creevey C, Muller J, Doerks T, Julien P, Roth A, Simonovic M, et al. STRING 8--a global view on proteins and their functional interactions in 630 organisms. Nucleic Acids Res. 2009;37(Database issue):D412–6.PubMed
26.
Zurück zum Zitat Hamidi H, Pietila M, Ivaska J. The complexity of integrins in cancer and new scopes for therapeutic targeting. Br J Cancer. 2016;115(9):1017–23.PubMedPubMedCentral Hamidi H, Pietila M, Ivaska J. The complexity of integrins in cancer and new scopes for therapeutic targeting. Br J Cancer. 2016;115(9):1017–23.PubMedPubMedCentral
27.
Zurück zum Zitat Kesteloot F, Desmouliere A, Leclercq I, Thiry M, Arrese JE, Prockop DJ, Lapiere CM, Nusgens BV, Colige A. ADAM metallopeptidase with thrombospondin type 1 motif 2 inactivation reduces the extent and stability of carbon tetrachloride-induced hepatic fibrosis in mice. Hepatology. 2007;46(5):1620–31.PubMed Kesteloot F, Desmouliere A, Leclercq I, Thiry M, Arrese JE, Prockop DJ, Lapiere CM, Nusgens BV, Colige A. ADAM metallopeptidase with thrombospondin type 1 motif 2 inactivation reduces the extent and stability of carbon tetrachloride-induced hepatic fibrosis in mice. Hepatology. 2007;46(5):1620–31.PubMed
28.
Zurück zum Zitat Wang X, Chen W, Zhang J, Khan A, Li L, Huang F, Qiu Z, Wang L, Chen X. Critical role of ADAMTS2 (a Disintegrin and metalloproteinase with Thrombospondin motifs 2) in cardiac hypertrophy induced by pressure overload. Hypertension. 2017;69(6):1060–9.PubMed Wang X, Chen W, Zhang J, Khan A, Li L, Huang F, Qiu Z, Wang L, Chen X. Critical role of ADAMTS2 (a Disintegrin and metalloproteinase with Thrombospondin motifs 2) in cardiac hypertrophy induced by pressure overload. Hypertension. 2017;69(6):1060–9.PubMed
29.
Zurück zum Zitat Bekhouche M, Leduc C, Dupont L, Janssen L, Delolme F, Vadon-Le Goff S, Smargiasso N, Baiwir D, Mazzucchelli G, Zanella-Cleon I, et al. Determination of the substrate repertoire of ADAMTS2, 3, and 14 significantly broadens their functions and identifies extracellular matrix organization and TGF-beta signaling as primary targets. FASEB J. 2016;30(5):1741–56.PubMed Bekhouche M, Leduc C, Dupont L, Janssen L, Delolme F, Vadon-Le Goff S, Smargiasso N, Baiwir D, Mazzucchelli G, Zanella-Cleon I, et al. Determination of the substrate repertoire of ADAMTS2, 3, and 14 significantly broadens their functions and identifies extracellular matrix organization and TGF-beta signaling as primary targets. FASEB J. 2016;30(5):1741–56.PubMed
30.
Zurück zum Zitat Hung CF, Rohani MG, Lee SS, Chen P, Schnapp LM. Role of IGF-1 pathway in lung fibroblast activation. Respir Res. 2013;14:102.PubMedPubMedCentral Hung CF, Rohani MG, Lee SS, Chen P, Schnapp LM. Role of IGF-1 pathway in lung fibroblast activation. Respir Res. 2013;14:102.PubMedPubMedCentral
31.
Zurück zum Zitat Adamek A, Kasprzak A. Insulin-Like Growth Factor (IGF) System in Liver Diseases. Int J Mol Sci. 2018;19(5):1308.PubMedCentral Adamek A, Kasprzak A. Insulin-Like Growth Factor (IGF) System in Liver Diseases. Int J Mol Sci. 2018;19(5):1308.PubMedCentral
32.
Zurück zum Zitat Svegliati-Baroni G, Ridolfi F, Di Sario A, Casini A, Marucci L, Gaggiotti G, Orlandoni P, Macarri G, Perego L, Benedetti A, et al. Insulin and insulin-like growth factor-1 stimulate proliferation and type I collagen accumulation by human hepatic stellate cells: differential effects on signal transduction pathways. Hepatology. 1999;29(6):1743–51.PubMed Svegliati-Baroni G, Ridolfi F, Di Sario A, Casini A, Marucci L, Gaggiotti G, Orlandoni P, Macarri G, Perego L, Benedetti A, et al. Insulin and insulin-like growth factor-1 stimulate proliferation and type I collagen accumulation by human hepatic stellate cells: differential effects on signal transduction pathways. Hepatology. 1999;29(6):1743–51.PubMed
33.
Zurück zum Zitat Mutgan AC, Besikcioglu HE, Wang S, Friess H, Ceyhan GO, Demir IE. Insulin/IGF-driven cancer cell-stroma crosstalk as a novel therapeutic target in pancreatic cancer. Mol Cancer. 2018;17(1):66.PubMedPubMedCentral Mutgan AC, Besikcioglu HE, Wang S, Friess H, Ceyhan GO, Demir IE. Insulin/IGF-driven cancer cell-stroma crosstalk as a novel therapeutic target in pancreatic cancer. Mol Cancer. 2018;17(1):66.PubMedPubMedCentral
34.
Zurück zum Zitat Li ZH, Xiong QY, Xu L, Duan P, Yang QO, Zhou P. Tu JH: miR-29a regulated ER-positive breast cancer cell growth and invasion and is involved in the insulin signaling pathway. Oncotarget. 2017;8(20):32566–75.PubMedPubMedCentral Li ZH, Xiong QY, Xu L, Duan P, Yang QO, Zhou P. Tu JH: miR-29a regulated ER-positive breast cancer cell growth and invasion and is involved in the insulin signaling pathway. Oncotarget. 2017;8(20):32566–75.PubMedPubMedCentral
35.
Zurück zum Zitat Yang J, Waldron RT, Su HY, Moro A, Chang HH, Eibl G, Ferreri K, Kandeel FR, Lugea A, Li L, et al. Insulin promotes proliferation and fibrosing responses in activated pancreatic stellate cells. Am J Physiol Gastrointest Liver Physiol. 2016;311(4):G675–87.PubMedPubMedCentral Yang J, Waldron RT, Su HY, Moro A, Chang HH, Eibl G, Ferreri K, Kandeel FR, Lugea A, Li L, et al. Insulin promotes proliferation and fibrosing responses in activated pancreatic stellate cells. Am J Physiol Gastrointest Liver Physiol. 2016;311(4):G675–87.PubMedPubMedCentral
36.
Zurück zum Zitat Hastings JF, Skhinas JN, Fey D, Croucher DR, Cox TR. The extracellular matrix as a key regulator of intracellular signalling networks. Br J Pharmacol. 2019;176(1):82–92.PubMed Hastings JF, Skhinas JN, Fey D, Croucher DR, Cox TR. The extracellular matrix as a key regulator of intracellular signalling networks. Br J Pharmacol. 2019;176(1):82–92.PubMed
37.
Zurück zum Zitat Hakuno F, Takahashi SI. IGF1 receptor signaling pathways. J Mol Endocrinol. 2018;61(1):T69–86.PubMed Hakuno F, Takahashi SI. IGF1 receptor signaling pathways. J Mol Endocrinol. 2018;61(1):T69–86.PubMed
38.
Zurück zum Zitat Braicu C, Buse M, Busuioc C, Drula R, Gulei D, Raduly L, Rusu A, Irimie A, Atanasov AG, Slaby O, et al. A comprehensive review on MAPK: a promising therapeutic target in cancer. Cancers (Basel). 2019;11(10):1618. Braicu C, Buse M, Busuioc C, Drula R, Gulei D, Raduly L, Rusu A, Irimie A, Atanasov AG, Slaby O, et al. A comprehensive review on MAPK: a promising therapeutic target in cancer. Cancers (Basel). 2019;11(10):1618.
39.
Zurück zum Zitat Dey S, Kwon JJ, Liu S, Hodge GA, Taleb S, Zimmers TA, Wan J, Kota J. miR-29a is repressed by MYC in pancreatic Cancer and its restoration drives tumor-suppressive effects via Downregulation of LOXL2. Mol Cancer Res. 2020;18(2):311–23.PubMed Dey S, Kwon JJ, Liu S, Hodge GA, Taleb S, Zimmers TA, Wan J, Kota J. miR-29a is repressed by MYC in pancreatic Cancer and its restoration drives tumor-suppressive effects via Downregulation of LOXL2. Mol Cancer Res. 2020;18(2):311–23.PubMed
40.
Zurück zum Zitat Pandol S, Edderkaoui M, Gukovsky I, Lugea A, Gukovskaya A. Desmoplasia of pancreatic ductal adenocarcinoma. Clin Gastroenterol Hepatol. 2009;7(11 Suppl):S44–7.PubMedPubMedCentral Pandol S, Edderkaoui M, Gukovsky I, Lugea A, Gukovskaya A. Desmoplasia of pancreatic ductal adenocarcinoma. Clin Gastroenterol Hepatol. 2009;7(11 Suppl):S44–7.PubMedPubMedCentral
41.
Zurück zum Zitat Viloria K, Munasinghe A, Asher S, Bogyere R, Jones L, Hill NJ. A holistic approach to dissecting SPARC family protein complexity reveals FSTL-1 as an inhibitor of pancreatic cancer cell growth. Sci Rep. 2016;6:37839.PubMedPubMedCentral Viloria K, Munasinghe A, Asher S, Bogyere R, Jones L, Hill NJ. A holistic approach to dissecting SPARC family protein complexity reveals FSTL-1 as an inhibitor of pancreatic cancer cell growth. Sci Rep. 2016;6:37839.PubMedPubMedCentral
42.
Zurück zum Zitat Jang I, Beningo KA. Integrins, CAFs and Mechanical Forces in the Progression of Cancer. Cancers (Basel). 2019;11(5):721. Jang I, Beningo KA. Integrins, CAFs and Mechanical Forces in the Progression of Cancer. Cancers (Basel). 2019;11(5):721.
43.
Zurück zum Zitat Aumailley M. The laminin family. Cell Adhes Migr. 2013;7(1):48–55. Aumailley M. The laminin family. Cell Adhes Migr. 2013;7(1):48–55.
44.
Zurück zum Zitat Givant-Horwitz V, Davidson B, Reich R. Laminin-induced signaling in tumor cells. Cancer Lett. 2005;223(1):1–10.PubMed Givant-Horwitz V, Davidson B, Reich R. Laminin-induced signaling in tumor cells. Cancer Lett. 2005;223(1):1–10.PubMed
45.
Zurück zum Zitat Nishikawa R, Goto Y, Kojima S, Enokida H, Chiyomaru T, Kinoshita T, Sakamoto S, Fuse M, Nakagawa M, Naya Y, et al. Tumor-suppressive microRNA-29s inhibit cancer cell migration and invasion via targeting LAMC1 in prostate cancer. Int J Oncol. 2014;45(1):401–10.PubMed Nishikawa R, Goto Y, Kojima S, Enokida H, Chiyomaru T, Kinoshita T, Sakamoto S, Fuse M, Nakagawa M, Naya Y, et al. Tumor-suppressive microRNA-29s inhibit cancer cell migration and invasion via targeting LAMC1 in prostate cancer. Int J Oncol. 2014;45(1):401–10.PubMed
46.
Zurück zum Zitat Kashima H, Wu RC, Wang Y, Sinno AK, Miyamoto T, Shiozawa T, Wang TL, Fader AN, Shih Ie M. Laminin C1 expression by uterine carcinoma cells is associated with tumor progression. Gynecol Oncol. 2015;139(2):338–44.PubMedPubMedCentral Kashima H, Wu RC, Wang Y, Sinno AK, Miyamoto T, Shiozawa T, Wang TL, Fader AN, Shih Ie M. Laminin C1 expression by uterine carcinoma cells is associated with tumor progression. Gynecol Oncol. 2015;139(2):338–44.PubMedPubMedCentral
47.
Zurück zum Zitat Zhang Y, Xi S, Chen J, Zhou D, Gao H, Zhou Z, Xu L, Chen M. Overexpression of LAMC1 predicts poor prognosis and enhances tumor cell invasion and migration in hepatocellular carcinoma. J Cancer. 2017;8(15):2992–3000.PubMedPubMedCentral Zhang Y, Xi S, Chen J, Zhou D, Gao H, Zhou Z, Xu L, Chen M. Overexpression of LAMC1 predicts poor prognosis and enhances tumor cell invasion and migration in hepatocellular carcinoma. J Cancer. 2017;8(15):2992–3000.PubMedPubMedCentral
48.
Zurück zum Zitat Takahashi S, Hasebe T, Oda T, Sasaki S, Kinoshita T, Konishi M, Ochiai T, Ochiai A. Cytoplasmic expression of laminin gamma2 chain correlates with postoperative hepatic metastasis and poor prognosis in patients with pancreatic ductal adenocarcinoma. Cancer. 2002;94(6):1894–901.PubMed Takahashi S, Hasebe T, Oda T, Sasaki S, Kinoshita T, Konishi M, Ochiai T, Ochiai A. Cytoplasmic expression of laminin gamma2 chain correlates with postoperative hepatic metastasis and poor prognosis in patients with pancreatic ductal adenocarcinoma. Cancer. 2002;94(6):1894–901.PubMed
49.
Zurück zum Zitat Miyamoto H, Murakami T, Tsuchida K, Sugino H, Miyake H, Tashiro S. Tumor-stroma interaction of human pancreatic cancer: acquired resistance to anticancer drugs and proliferation regulation is dependent on extracellular matrix proteins. Pancreas. 2004;28(1):38–44.PubMed Miyamoto H, Murakami T, Tsuchida K, Sugino H, Miyake H, Tashiro S. Tumor-stroma interaction of human pancreatic cancer: acquired resistance to anticancer drugs and proliferation regulation is dependent on extracellular matrix proteins. Pancreas. 2004;28(1):38–44.PubMed
50.
Zurück zum Zitat Damiano JS, Cress AE, Hazlehurst LA, Shtil AA, Dalton WS. Cell adhesion mediated drug resistance (CAM-DR): role of integrins and resistance to apoptosis in human myeloma cell lines. Blood. 1999;93(5):1658–67.PubMed Damiano JS, Cress AE, Hazlehurst LA, Shtil AA, Dalton WS. Cell adhesion mediated drug resistance (CAM-DR): role of integrins and resistance to apoptosis in human myeloma cell lines. Blood. 1999;93(5):1658–67.PubMed
51.
Zurück zum Zitat Zhang H, Ozaki I, Mizuta T, Matsuhashi S, Yoshimura T, Hisatomi A, Tadano J, Sakai T, Yamamoto K. Beta 1-integrin protects hepatoma cells from chemotherapy induced apoptosis via a mitogen-activated protein kinase dependent pathway. Cancer. 2002;95(4):896–906.PubMed Zhang H, Ozaki I, Mizuta T, Matsuhashi S, Yoshimura T, Hisatomi A, Tadano J, Sakai T, Yamamoto K. Beta 1-integrin protects hepatoma cells from chemotherapy induced apoptosis via a mitogen-activated protein kinase dependent pathway. Cancer. 2002;95(4):896–906.PubMed
52.
Zurück zum Zitat Amrutkar M, Aasrum M, Verbeke CS, Gladhaug IP. Secretion of fibronectin by human pancreatic stellate cells promotes chemoresistance to gemcitabine in pancreatic cancer cells. BMC Cancer. 2019;19(1):596.PubMedPubMedCentral Amrutkar M, Aasrum M, Verbeke CS, Gladhaug IP. Secretion of fibronectin by human pancreatic stellate cells promotes chemoresistance to gemcitabine in pancreatic cancer cells. BMC Cancer. 2019;19(1):596.PubMedPubMedCentral
53.
Zurück zum Zitat Rucki AA, Foley K, Zhang P, Xiao Q, Kleponis J, Wu AA, Sharma R, Mo G, Liu A, Van Eyk J, et al. Heterogeneous stromal signaling within the tumor microenvironment controls the metastasis of pancreatic Cancer. Cancer Res. 2017;77(1):41–52.PubMed Rucki AA, Foley K, Zhang P, Xiao Q, Kleponis J, Wu AA, Sharma R, Mo G, Liu A, Van Eyk J, et al. Heterogeneous stromal signaling within the tumor microenvironment controls the metastasis of pancreatic Cancer. Cancer Res. 2017;77(1):41–52.PubMed
54.
Zurück zum Zitat Rohrmann S, Grote VA, Becker S, Rinaldi S, Tjonneland A, Roswall N, Gronbaek H, Overvad K, Boutron-Ruault MC, Clavel-Chapelon F, et al. Concentrations of IGF-I and IGFBP-3 and pancreatic cancer risk in the European prospective investigation into Cancer and nutrition. Br J Cancer. 2012;106(5):1004–10.PubMedPubMedCentral Rohrmann S, Grote VA, Becker S, Rinaldi S, Tjonneland A, Roswall N, Gronbaek H, Overvad K, Boutron-Ruault MC, Clavel-Chapelon F, et al. Concentrations of IGF-I and IGFBP-3 and pancreatic cancer risk in the European prospective investigation into Cancer and nutrition. Br J Cancer. 2012;106(5):1004–10.PubMedPubMedCentral
55.
Zurück zum Zitat Sobel G, Szabo I, Paska C, Kiss A, Kovalszky I, Kadar A, Paulin F, Schaff Z. Changes of cell adhesion and extracellular matrix (ECM) components in cervical intraepithelial neoplasia. Pathol Oncol Res. 2005;11(1):26–31.PubMed Sobel G, Szabo I, Paska C, Kiss A, Kovalszky I, Kadar A, Paulin F, Schaff Z. Changes of cell adhesion and extracellular matrix (ECM) components in cervical intraepithelial neoplasia. Pathol Oncol Res. 2005;11(1):26–31.PubMed
56.
Zurück zum Zitat Huang J, Zhang L, He C, Qu Y, Li J, Zhang J, Du T, Chen X, Yu Y, Liu B, et al. Claudin-1 enhances tumor proliferation and metastasis by regulating cell anoikis in gastric cancer. Oncotarget. 2015;6(3):1652–65.PubMed Huang J, Zhang L, He C, Qu Y, Li J, Zhang J, Du T, Chen X, Yu Y, Liu B, et al. Claudin-1 enhances tumor proliferation and metastasis by regulating cell anoikis in gastric cancer. Oncotarget. 2015;6(3):1652–65.PubMed
57.
Zurück zum Zitat Wu X, Xiao J, Zhao C, Zhao C, Han Z, Wang F, Yang Y, Jiang Y, Fang F. Claudin1 promotes the proliferation, invasion and migration of nasopharyngeal carcinoma cells by upregulating the expression and nuclear entry of beta-catenin. Exp Ther Med. 2018;16(4):3445–51.PubMedPubMedCentral Wu X, Xiao J, Zhao C, Zhao C, Han Z, Wang F, Yang Y, Jiang Y, Fang F. Claudin1 promotes the proliferation, invasion and migration of nasopharyngeal carcinoma cells by upregulating the expression and nuclear entry of beta-catenin. Exp Ther Med. 2018;16(4):3445–51.PubMedPubMedCentral
58.
Zurück zum Zitat Xue R, Hua L, Xu W, Gao Y, Pang Y, Hao J. Derivation and validation of the potential Core genes in pancreatic Cancer for tumor-Stroma crosstalk. Biomed Res Int. 2018;2018:4283673.PubMedPubMedCentral Xue R, Hua L, Xu W, Gao Y, Pang Y, Hao J. Derivation and validation of the potential Core genes in pancreatic Cancer for tumor-Stroma crosstalk. Biomed Res Int. 2018;2018:4283673.PubMedPubMedCentral
59.
Zurück zum Zitat Hatakeyama N, Kojima T, Iba K, Murata M, Thi MM, Spray DC, Osanai M, Chiba H, Ishiai S, Yamashita T, et al. IGF-I regulates tight-junction protein claudin-1 during differentiation of osteoblast-like MC3T3-E1 cells via a MAP-kinase pathway. Cell Tissue Res. 2008;334(2):243–54.PubMedPubMedCentral Hatakeyama N, Kojima T, Iba K, Murata M, Thi MM, Spray DC, Osanai M, Chiba H, Ishiai S, Yamashita T, et al. IGF-I regulates tight-junction protein claudin-1 during differentiation of osteoblast-like MC3T3-E1 cells via a MAP-kinase pathway. Cell Tissue Res. 2008;334(2):243–54.PubMedPubMedCentral
60.
Zurück zum Zitat Lara-Diaz VJ, Castilla-Cortazar I, Martin-Estal I, Garcia-Magarino M, Aguirre GA, Puche JE, de la Garza RG, Morales LA, Munoz U. IGF-1 modulates gene expression of proteins involved in inflammation, cytoskeleton, and liver architecture. J Physiol Biochem. 2017;73(2):245–58.PubMedPubMedCentral Lara-Diaz VJ, Castilla-Cortazar I, Martin-Estal I, Garcia-Magarino M, Aguirre GA, Puche JE, de la Garza RG, Morales LA, Munoz U. IGF-1 modulates gene expression of proteins involved in inflammation, cytoskeleton, and liver architecture. J Physiol Biochem. 2017;73(2):245–58.PubMedPubMedCentral
61.
Zurück zum Zitat Roderburg C, Luedde M, Vargas Cardenas D, Vucur M, Mollnow T, Zimmermann HW, Koch A, Hellerbrand C, Weiskirchen R, Frey N, et al. miR-133a mediates TGF-beta-dependent derepression of collagen synthesis in hepatic stellate cells during liver fibrosis. J Hepatol. 2013;58(4):736–42.PubMed Roderburg C, Luedde M, Vargas Cardenas D, Vucur M, Mollnow T, Zimmermann HW, Koch A, Hellerbrand C, Weiskirchen R, Frey N, et al. miR-133a mediates TGF-beta-dependent derepression of collagen synthesis in hepatic stellate cells during liver fibrosis. J Hepatol. 2013;58(4):736–42.PubMed
62.
Zurück zum Zitat Huang G, Ge G, Izzi V. Greenspan DS: alpha3 chains of type V collagen regulate breast tumour growth via glypican-1. Nat Commun. 2017;8:14351.PubMedPubMedCentral Huang G, Ge G, Izzi V. Greenspan DS: alpha3 chains of type V collagen regulate breast tumour growth via glypican-1. Nat Commun. 2017;8:14351.PubMedPubMedCentral
63.
Zurück zum Zitat Weniger M, Honselmann KC, Liss AS. The extracellular matrix and pancreatic cancer: a complex relationship. Cancers (Basel). 2018;10(9):316. Weniger M, Honselmann KC, Liss AS. The extracellular matrix and pancreatic cancer: a complex relationship. Cancers (Basel). 2018;10(9):316.
64.
Zurück zum Zitat Saneyasu T, Akhtar R, Sakai T. Molecular cues guiding matrix stiffness in liver fibrosis. Biomed Res Int. 2016;2016:2646212.PubMedPubMedCentral Saneyasu T, Akhtar R, Sakai T. Molecular cues guiding matrix stiffness in liver fibrosis. Biomed Res Int. 2016;2016:2646212.PubMedPubMedCentral
65.
Zurück zum Zitat Hazar-Rethinam M, de Long LM, Gannon OM, Boros S, Vargas AC, Dzienis M, Mukhopadhyay P, Saenz-Ponce N, Dantzic DD, Simpson F, et al. RacGAP1 is a novel downstream effector of E2F7-dependent resistance to doxorubicin and is prognostic for overall survival in squamous cell carcinoma. Mol Cancer Ther. 2015;14(8):1939–50.PubMed Hazar-Rethinam M, de Long LM, Gannon OM, Boros S, Vargas AC, Dzienis M, Mukhopadhyay P, Saenz-Ponce N, Dantzic DD, Simpson F, et al. RacGAP1 is a novel downstream effector of E2F7-dependent resistance to doxorubicin and is prognostic for overall survival in squamous cell carcinoma. Mol Cancer Ther. 2015;14(8):1939–50.PubMed
66.
Zurück zum Zitat Lomberk G, Blum Y, Nicolle R, Nair A, Gaonkar KS, Marisa L, Mathison A, Sun Z, Yan H, Elarouci N, et al. Distinct epigenetic landscapes underlie the pathobiology of pancreatic cancer subtypes. Nat Commun. 2018;9(1):1978.PubMedPubMedCentral Lomberk G, Blum Y, Nicolle R, Nair A, Gaonkar KS, Marisa L, Mathison A, Sun Z, Yan H, Elarouci N, et al. Distinct epigenetic landscapes underlie the pathobiology of pancreatic cancer subtypes. Nat Commun. 2018;9(1):1978.PubMedPubMedCentral
67.
Zurück zum Zitat Wang Y. The effect of E2F7 expression in prostate cancer on apoptosis and cell cycle of prostate cancer cells. J Clin Oncol. 2019;37(15_suppl):e16568. Wang Y. The effect of E2F7 expression in prostate cancer on apoptosis and cell cycle of prostate cancer cells. J Clin Oncol. 2019;37(15_suppl):e16568.
68.
Zurück zum Zitat Carvajal LA, Hamard PJ, Tonnessen C, Manfredi JJ. E2F7, a novel target, is up-regulated by p53 and mediates DNA damage-dependent transcriptional repression. Genes Dev. 2012;26(14):1533–45.PubMedPubMedCentral Carvajal LA, Hamard PJ, Tonnessen C, Manfredi JJ. E2F7, a novel target, is up-regulated by p53 and mediates DNA damage-dependent transcriptional repression. Genes Dev. 2012;26(14):1533–45.PubMedPubMedCentral
69.
Zurück zum Zitat Musa J, Aynaud MM, Mirabeau O, Delattre O, Grunewald TG. MYBL2 (B-Myb): a central regulator of cell proliferation, cell survival and differentiation involved in tumorigenesis. Cell Death Dis. 2017;8(6):e2895.PubMedPubMedCentral Musa J, Aynaud MM, Mirabeau O, Delattre O, Grunewald TG. MYBL2 (B-Myb): a central regulator of cell proliferation, cell survival and differentiation involved in tumorigenesis. Cell Death Dis. 2017;8(6):e2895.PubMedPubMedCentral
70.
Zurück zum Zitat Qin H, Li Y, Zhang H, Wang F, He H, Bai X, Li S. Prognostic implications and oncogenic roles of MYBL2 protein expression in esophageal squamous-cell carcinoma. Onco Targets Ther. 2019;12:1917–27.PubMedPubMedCentral Qin H, Li Y, Zhang H, Wang F, He H, Bai X, Li S. Prognostic implications and oncogenic roles of MYBL2 protein expression in esophageal squamous-cell carcinoma. Onco Targets Ther. 2019;12:1917–27.PubMedPubMedCentral
71.
Zurück zum Zitat Bhardwaj A, Srivastava SK, Singh S, Tyagi N, Arora S, Carter JE, Khushman M, Singh AP. MYB promotes Desmoplasia in pancreatic Cancer through direct transcriptional up-regulation and cooperative action of sonic hedgehog and Adrenomedullin. J Biol Chem. 2016;291(31):16263–70.PubMedPubMedCentral Bhardwaj A, Srivastava SK, Singh S, Tyagi N, Arora S, Carter JE, Khushman M, Singh AP. MYB promotes Desmoplasia in pancreatic Cancer through direct transcriptional up-regulation and cooperative action of sonic hedgehog and Adrenomedullin. J Biol Chem. 2016;291(31):16263–70.PubMedPubMedCentral
72.
Zurück zum Zitat Saison-Ridinger M, DelGiorno KE, Zhang T, Kraus A, French R, Jaquish D, Tsui C, Erikson G, Spike BT, Shokhirev MN, et al. Reprogramming pancreatic stellate cells via p53 activation: a putative target for pancreatic cancer therapy. PLoS One. 2017;12(12):e0189051.PubMedPubMedCentral Saison-Ridinger M, DelGiorno KE, Zhang T, Kraus A, French R, Jaquish D, Tsui C, Erikson G, Spike BT, Shokhirev MN, et al. Reprogramming pancreatic stellate cells via p53 activation: a putative target for pancreatic cancer therapy. PLoS One. 2017;12(12):e0189051.PubMedPubMedCentral
73.
Zurück zum Zitat Huang YH, Chen MH, Guo QL, Chen ZX, Chen QD, Wang XZ. Interleukin-10 induces senescence of activated hepatic stellate cells via STAT3-p53 pathway to attenuate liver fibrosis. Cell Signal. 2020;66:109445.PubMed Huang YH, Chen MH, Guo QL, Chen ZX, Chen QD, Wang XZ. Interleukin-10 induces senescence of activated hepatic stellate cells via STAT3-p53 pathway to attenuate liver fibrosis. Cell Signal. 2020;66:109445.PubMed
74.
Zurück zum Zitat Kiaris H, Chatzistamou I, Trimis G, Frangou-Plemmenou M, Pafiti-Kondi A, Kalofoutis A. Evidence for nonautonomous effect of p53 tumor suppressor in carcinogenesis. Cancer Res. 2005;65(5):1627–30.PubMed Kiaris H, Chatzistamou I, Trimis G, Frangou-Plemmenou M, Pafiti-Kondi A, Kalofoutis A. Evidence for nonautonomous effect of p53 tumor suppressor in carcinogenesis. Cancer Res. 2005;65(5):1627–30.PubMed
75.
Zurück zum Zitat Kang SY, Halvorsen OJ, Gravdal K, Bhattacharya N, Lee JM, Liu NW, Johnston BT, Johnston AB, Haukaas SA, Aamodt K, et al. Prosaposin inhibits tumor metastasis via paracrine and endocrine stimulation of stromal p53 and Tsp-1. Proc Natl Acad Sci U S A. 2009;106(29):12115–20.PubMedPubMedCentral Kang SY, Halvorsen OJ, Gravdal K, Bhattacharya N, Lee JM, Liu NW, Johnston BT, Johnston AB, Haukaas SA, Aamodt K, et al. Prosaposin inhibits tumor metastasis via paracrine and endocrine stimulation of stromal p53 and Tsp-1. Proc Natl Acad Sci U S A. 2009;106(29):12115–20.PubMedPubMedCentral
76.
Zurück zum Zitat Sadasivam S, Duan S, DeCaprio JA. The MuvB complex sequentially recruits B-Myb and FoxM1 to promote mitotic gene expression. Genes Dev. 2012;26(5):474–89.PubMedPubMedCentral Sadasivam S, Duan S, DeCaprio JA. The MuvB complex sequentially recruits B-Myb and FoxM1 to promote mitotic gene expression. Genes Dev. 2012;26(5):474–89.PubMedPubMedCentral
77.
Zurück zum Zitat Chin YR, Toker A. Function of Akt/PKB signaling to cell motility, invasion and the tumor stroma in cancer. Cell Signal. 2009;21(4):470–6.PubMed Chin YR, Toker A. Function of Akt/PKB signaling to cell motility, invasion and the tumor stroma in cancer. Cell Signal. 2009;21(4):470–6.PubMed
78.
Zurück zum Zitat Tape CJ, Ling S, Dimitriadi M, McMahon KM, Worboys JD, Leong HS, Norrie IC, Miller CJ, Poulogiannis G, Lauffenburger DA, et al. Oncogenic KRAS regulates tumor cell signaling via stromal reciprocation. Cell. 2016;165(7):1818.PubMedPubMedCentral Tape CJ, Ling S, Dimitriadi M, McMahon KM, Worboys JD, Leong HS, Norrie IC, Miller CJ, Poulogiannis G, Lauffenburger DA, et al. Oncogenic KRAS regulates tumor cell signaling via stromal reciprocation. Cell. 2016;165(7):1818.PubMedPubMedCentral
79.
Zurück zum Zitat Zhang X, Lv QL, Huang YT, Zhang LH, Zhou HH. Akt/FoxM1 signaling pathway-mediated upregulation of MYBL2 promotes progression of human glioma. J Exp Clin Cancer Res. 2017;36(1):105.PubMedPubMedCentral Zhang X, Lv QL, Huang YT, Zhang LH, Zhou HH. Akt/FoxM1 signaling pathway-mediated upregulation of MYBL2 promotes progression of human glioma. J Exp Clin Cancer Res. 2017;36(1):105.PubMedPubMedCentral
80.
Zurück zum Zitat Ahmed F. Integrated network analysis reveals FOXM1 and MYBL2 as key regulators of cell proliferation in non-small cell lung Cancer. Front Oncol. 2019;9:1011.PubMedPubMedCentral Ahmed F. Integrated network analysis reveals FOXM1 and MYBL2 as key regulators of cell proliferation in non-small cell lung Cancer. Front Oncol. 2019;9:1011.PubMedPubMedCentral
81.
Zurück zum Zitat Latres E, Amini AR, Amini AA, Griffiths J, Martin FJ, Wei Y, Lin HC, Yancopoulos GD, Glass DJ. Insulin-like growth factor-1 (IGF-1) inversely regulates atrophy-induced genes via the phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR) pathway. J Biol Chem. 2005;280(4):2737–44.PubMed Latres E, Amini AR, Amini AA, Griffiths J, Martin FJ, Wei Y, Lin HC, Yancopoulos GD, Glass DJ. Insulin-like growth factor-1 (IGF-1) inversely regulates atrophy-induced genes via the phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR) pathway. J Biol Chem. 2005;280(4):2737–44.PubMed
82.
Zurück zum Zitat Werner H, Sarfstein R, LeRoith D, Bruchim I. Insulin-like growth factor 1 signaling Axis meets p53 genome protection pathways. Front Oncol. 2016;6:159.PubMedPubMedCentral Werner H, Sarfstein R, LeRoith D, Bruchim I. Insulin-like growth factor 1 signaling Axis meets p53 genome protection pathways. Front Oncol. 2016;6:159.PubMedPubMedCentral
83.
Zurück zum Zitat Ma YS, Lv ZW, Yu F, Chang ZY, Cong XL, Zhong XM, Lu GX, Zhu J, Fu D. MicroRNA-302a/d inhibits the self-renewal capability and cell cycle entry of liver cancer stem cells by targeting the E2F7/AKT axis. J Exp Clin Cancer Res. 2018;37(1):252.PubMedPubMedCentral Ma YS, Lv ZW, Yu F, Chang ZY, Cong XL, Zhong XM, Lu GX, Zhu J, Fu D. MicroRNA-302a/d inhibits the self-renewal capability and cell cycle entry of liver cancer stem cells by targeting the E2F7/AKT axis. J Exp Clin Cancer Res. 2018;37(1):252.PubMedPubMedCentral
84.
Zurück zum Zitat Wang C, Li S, Xu J, Niu W. Li S: microRNA-935 is reduced in non-small cell lung cancer tissue, is linked to poor outcome, and acts on signal transduction mediator E2F7 and the AKT pathway. Br J Biomed Sci. 2019;76(1):17–23.PubMed Wang C, Li S, Xu J, Niu W. Li S: microRNA-935 is reduced in non-small cell lung cancer tissue, is linked to poor outcome, and acts on signal transduction mediator E2F7 and the AKT pathway. Br J Biomed Sci. 2019;76(1):17–23.PubMed
85.
Zurück zum Zitat Zhou H, Guo R, Wang C. Long non-coding RNA NEAT1 accelerates cell progression in cervical cancer by regulating the miR-889-3p/E2F7 axis through the activation of the PI3K/AKT pathway. RSC Adv. 2019;9:34627–35. Zhou H, Guo R, Wang C. Long non-coding RNA NEAT1 accelerates cell progression in cervical cancer by regulating the miR-889-3p/E2F7 axis through the activation of the PI3K/AKT pathway. RSC Adv. 2019;9:34627–35.
Metadaten
Titel
Global targetome analysis reveals critical role of miR-29a in pancreatic stellate cell mediated regulation of PDAC tumor microenvironment
verfasst von
Shatovisha Dey
Sheng Liu
Tricia D. Factora
Solaema Taleb
Primavera Riverahernandez
Lata Udari
Xiaoling Zhong
Jun Wan
Janaiah Kota
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2020
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-020-07135-2

Weitere Artikel der Ausgabe 1/2020

BMC Cancer 1/2020 Zur Ausgabe

„Überwältigende“ Evidenz für Tripeltherapie beim metastasierten Prostata-Ca.

22.05.2024 Prostatakarzinom Nachrichten

Patienten mit metastasiertem hormonsensitivem Prostatakarzinom sollten nicht mehr mit einer alleinigen Androgendeprivationstherapie (ADT) behandelt werden, mahnt ein US-Team nach Sichtung der aktuellen Datenlage. Mit einer Tripeltherapie haben die Betroffenen offenbar die besten Überlebenschancen.

So sicher sind Tattoos: Neue Daten zur Risikobewertung

22.05.2024 Melanom Nachrichten

Das größte medizinische Problem bei Tattoos bleiben allergische Reaktionen. Melanome werden dadurch offensichtlich nicht gefördert, die Farbpigmente könnten aber andere Tumoren begünstigen.

CAR-M-Zellen: Warten auf das große Fressen

22.05.2024 Onkologische Immuntherapie Nachrichten

Auch myeloide Immunzellen lassen sich mit chimären Antigenrezeptoren gegen Tumoren ausstatten. Solche CAR-Fresszell-Therapien werden jetzt für solide Tumoren entwickelt. Künftig soll dieser Prozess nicht mehr ex vivo, sondern per mRNA im Körper der Betroffenen erfolgen.

Blutdrucksenkung könnte Uterusmyome verhindern

Frauen mit unbehandelter oder neu auftretender Hypertonie haben ein deutlich erhöhtes Risiko für Uterusmyome. Eine Therapie mit Antihypertensiva geht hingegen mit einer verringerten Inzidenz der gutartigen Tumoren einher.

Update Onkologie

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