Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Aug 1.
Published in final edited form as: Toxicol Pathol. 2013 Aug 26;42(5):863–876. doi: 10.1177/0192623313501894

Spontaneous Mesotheliomas in F344/N Rats are Characterized by Dysregulation of Cellular Growth and Immune Function Pathways

Pamela E Blackshear 4, Arun R Pandiri 5, Thai-Vu T Ton 1, Natasha P Clayton 1, Keith R Shockley 2, Shyamal D Peddada 2, Kevin E Gerrish 3, Robert C Sills 1, Mark J Hoenerhoff 1
PMCID: PMC4967937  NIHMSID: NIHMS510379  PMID: 23980201

Abstract

Aged male Fischer 344/N rats are prone to developing spontaneous peritoneal mesotheliomas, which arise predominantly from the tunica vaginalis of the testes. A definitive cause for the predominance of this neoplasm in F344/N rats is unknown. Investigation of the molecular alterations that occur in spontaneous rat mesotheliomas may provide insight into their pathogenesis, as well enable a better understanding regarding the mechanisms underlying chemically induced mesothelioma in rodents. Mesothelial cell function represents a complex interplay of pathways related to host defense mechanisms and maintenance of cellular homeostasis. Global gene expression profiles of spontaneous mesotheliomas from vehicle control male F344/N rats from two-year National Toxicology Program carcinogenicity bioassays were analyzed to determine the molecular features of these tumors, and elucidate tumor-specific gene expression profiles. The resulting gene expression pattern showed that spontaneous mesotheliomas are associated with upregulation various growth factors, oncogenes, cytokines, pattern recognition response receptors (PRR) and pathogen associated molecular patterns (PAMP) receptors, and the production of reactive oxygen and nitrogen species, as well as downregulation of apoptosis pathways. Alterations in these pathways in turn trigger molecular responses that stimulate cell proliferation and promote tumor survival and progression.

Keywords: Mesothelioma, F344/N rat, National Toxicology Program, microarray, gene expression, mesothelial cell, Fred-PE cells

Introduction

Mesothelial cells form the lining of the heart, pleural and peritoneal cavities as well as visceral surfaces of organs. The mesothelial lining of the peritoneum in males extends into the scrotum to form the tunica vaginalis, which covers the testes and the epididymis (Hall et al. 1990). The mesothelial lining consists of a single layer of two morphologically distinct cell types, 1) flattened cells and 2) dome-shaped cuboidal to polygonal cells, overlying a very thin fibrovascular stroma. The flattened cells are held together by apical tight junctions, adherens junctions, zona occludens, and desmosomes (Antony 2003; Madison et al. 1979), and the dome-shaped cells surround pores that connect endothelial cells with the mesothelium, thus providing a direct role for the mesothelial cell in fluid and large molecule transport between the peritoneal cavity and lymphatic system (Hall et al. 1990). Both domed and flattened mesothelial cells bear microvilli, which are involved in the absorption of fluid and maintenance of lubrication of peritoneal and pleural cavities. Mesothelial cells are a dynamic cell population, derived from embryonic mesoderm, and provide innate immune mechanisms and multiple cellular functions that aid in maintaining homeostasis for the pleural and peritoneal cavities. Mesothelial cells are capable of pinocytocysis of small particles as well as phagocytosis of large particles. Their cytoplasm contains microfilaments, which are involved in cell movement and cytoskeletal rearrangement, as well as proper function of mitochondria, smooth and rough endoplasmic reticulum, and pinocytotic vesicles. Mesothelial cells have both epithelial and mesenchymal characteristics; they express a broad range of cytokeratins as well as vimentin, and when activated, they express desmin and smooth muscle actin (Afify et al. 2002; Kupryjanczyk and Karpinska 1998; Yung and Chan 2011). Additionally, they produce hyaluronic acid and express a variety of cell surface receptors including CD44 (Nasreen et al. 2002; Yung and Chan 2011), pathogen associated molecular pattern (PAMP) receptors (e.g. CD14), and pattern recognition receptors (PRR) such as the toll-like receptors (TLRs), receptors for advanced glycation end products (RAGE) (Jantz and Antony 2008), and mannose receptors.

Mesotheliomas occur rarely in rats and humans, and are characterized by the neoplastic transformation of pleural or peritoneal mesothelial cells. In humans, these tumors have a long latency period (Hall et al. 1990) and an insidious nature that makes the diagnosis of mesotheliomas challenging, prognosis poor, and treatment options limited. Mesotheliomas occur at a low incidence in aged male F344/N rats (3.2%, all routes of vehicle exposure, all vehicles, NTP historical controls) (National Toxicology Program 2011), and arise most commonly from the tunica vaginalis (Hall et al. 1990). In females, mesotheliomas arise much less frequently (0.17%, all routes of vehicle exposure, all vehicles) (National Toxicology Program 2011), and typically arise from various locations within the peritoneal cavity, including adjacent to the ovary (Hall et al. 1990). There are three primary histologic phenotypes of mesothelioma in the rat: epitheliomatous, sarcomatous, and mixed. Most mesotheliomas in the rat are epitheliomatous, characterized by papillary projections overlying a fibrovascular stalk. Sarcomatous tumors are associated with spindyloid morphology, and mixed tumors have components of both epitheliomatous and sarcomatous phenotypes. All three histologic phenotypes that occur in the rat are also reported in humans (Blobel et al. 1985; Mackay et al. 1987), with the epitheliomatous subtype the most common (Remon et al. 2012). Both peritoneal and pleural mesotheliomas occur in humans, but by far, pleural mesotheliomas are the most common, occurring in up to 80% of all cases (Sekido 2013). Malignant pleural mesothelioma (MPM) is a rare disease in humans with approximately 2000–3000 new cases in the United States diagnosed annually (Kaufman and Pass 2008). The incidence of MPM has steadily increased over the last 20 years, with 70–80% of cases implicating asbestos exposure as the dominant risk factor (Kaufman and Pass 2008; Robinson and Lake 2005). Men have a higher incidence of pleural mesothelioma than women, possibly due to higher occupational exposures of asbestos (Yang et al. 2008). The remaining 20–30% of cases are attributed to non-asbestos risk factors, such as germline mutations in BRCA1-Associated Protein 1 (BAP1) (Bott et al. 2011; Testa et al.), SV40 viral infection (Carbone et al. 2003), and exposure to radiation or chemical agents(Carbone and Bedrossian 2006; Yang et al. 2008).

Molecular alterations in human mesothelioma associated with asbestos exposure have been an area of intense interest, and a topic of several recent reviews and gene expression profile targeted publications; gene expression profiling and mutation analysis of these tumors in humans has revealed a variety of genetic alterations including mutations in tumor suppressor genes (TP53, WT1, NF2 and PTEN) or cell cycle genes (CDKN2A/2B and CDKN2C), and alterations in proteins associated with tumorigenesis (COL3, CCL2, LGALS) (Bott et al. 2011; Carbone and Bedrossian 2006; Gordon et al. 2005; Gueugnon et al. 2011; Romagnoli et al. 2009; Sekido 2013; Tomasetti et al. 2009; Tsujimura et al. 2012). These alterations in gene expression patterns appear to vary between different populations of people, as well as between the various morphological subtypes of human mesothelioma.

The objective of this study was to investigate the gene expression profiles of spontaneous mesotheliomas in the Fischer 344/N rat. Previous studies have characterized the gene expression profiles or molecular features of some chemically-induced mesotheliomas in F344/N rats or F344 crosses (Hu et al. 2010; Kim et al. 2006). However, global gene expression profiling of spontaneous mesotheliomas in rats has not previously been performed. Moreover, without a database of molecular changes or genetic signatures for spontaneously occurring mesotheliomas, separating changes due to chemical exposure from those inherent in mesothelioma pathogenesis is impossible. Characterization of spontaneous peritoneal mesotheliomas in rats may provide clues into the pathogenesis of chemically induced mesotheliomas, and may also provide useful information as to the utility of the F344/N rat as a hazard identification model for non-asbestos related mesothelioma in humans.

Materials and Methods

Sample collection and histopathology

Spontaneous mesotheliomas were collected from male vehicle control Fischer 344/N rats from three (3) two-year NTP carcinogenicity bioassay studies (Cobalt, Riddelliine, and Codeine). The incidence of malignant mesotheliomas in vehicle control male rats from these studies was 2/50 (4%), 4/50 (8%), and 1/50 (2%), for Cobalt, Riddelliine, and Codeine, respectively. Samples were sectioned in half and one half was fixed in 10% neutral buffered formalin (NBF) and the other half was flash frozen in liquid nitrogen. Five tumor samples were selected for molecular biology based on size from three animals (Riddelliine, peritoneum; Codeine, mesentery; Cobalt, tunica vaginalis, peritoneum, and mesentery) (Table 1), and the morphology of each sample was reviewed histologically to assure minimal necrotic tissue (<20%) and maximum tumor to normal tissue (>80% tumor tissue) per section. . The rarity of large spontaneous mesotheliomas available necessitated using multiple spontaneous tumors from one animal from the Cobalt study. Since isolation of normal mesothelium from rats would not yield adequate material for microarray, we used a non-transformed, immortalized mesothelial cell line (Fred-PE) which was grown and subcultured from the peritoneal cavity of untreated normal F344/N rats (a generous gift from Dr. Yongbaek Kim, NCSU), which has been used as a mesothelium control in previous studies in F344/N rats (Crosby et al. 2000; Kim et al. 2006).

Table 1.

Select Differentially Expressed Genes Dysregulated in Spontaneous Mesotheliomas in F344/N rats.

Gene Name Gene Symbol Fold Change
Transforming Growth Factor
transforming growth factor, alpha Tgfa 12.45
transforming growth factor, beta 2 Tgfb2 −17.61
transforming growth factor, beta receptor 1 Tgfbr1 2.14
transforming growth factor, beta-induced, 68kDa Tgfbi 43.39
Platelet Derived Growth Factor
platelet derived growth factor D Pdgfd 1.88
platelet derived growth factor C Pdgfc −1.75
Vascular Endothelial Growth Factor
vascular endothelial growth factor A Vegfa −2.03
vascular endothelial growth factor B Vegfb −2.58
vascular endothelial growth factor C Vegfc −7.65
epidermal growth factor
epidermal growth factor receptor Egfr 1.53
Fibroblast Growth Factor
fibroblast growth factor 13 Fgf13 −10.61
fibroblast growth factor receptor 2 Fgfr2 −5.11
Insulin-like Growth Factor
insulin-like growth factor 1 (somatomedin C) Igf1 17.48
insulin-like growth factor 2 binding protein 1 Igf2bp1 −2.73
insulin-like growth factor 2 binding protein 2 Igf2bp2 −5.19
insulin-like growth factor 2 binding protein 3 Igf2bp3 −9.54
insulin-like growth factor binding protein 6 Igfbp6 10.20
insulin-like growth factor binding protein 3 Igfbp3 32.46
Cell cycle
cyclin-dependent kinase inhibitor 1A (p21, Cip1) Cdkn1a −4.53
cyclin-dependent kinase inhibitor 1B (p27, Kip1) Cdkn1b −5.69
cyclin D1 Ccnd1 −1.90
Oncogenes/Protooncogenes
v-maf musculoaponeurotic fibrosarcoma oncogene homolog B
(avian)
Mafb 25.86
v-myc myelocytomatosis viral related oncogene, neuroblastoma
derived (avian)
Mycn −29.75
FBJ murine osteosarcoma viral oncogene homolog Fos −9.62
jun B proto-oncogene Junb −4.69
v-yes-1 Yamaguchi sarcoma viral related oncogene homolog LYN (v-yes) 5.12
RAS pathway
RAS, dexamethasone-induced 1 Rasd1 39.86
Rho family GTPase 1 Rnd1 −13.89
Rho family GTPase 3 Rnd3 −13.90
Tumor suppressor genes
cyclin-dependent kinase inhibitor 2B (p15, inhibits CDK4) Cdkn2b −3.77
tumor protein p53 Tp53 −2.25
phosphatase and tensin homolog Pten −1.41
Wilms Tumor 1 Wt1 3.18
Ras association (RalGDS/AF-6) domain family member 1 Rassf1 −1.34
Adhesion Molecule, Integrins, catenins
epithelial cell adhesion molecule Epcam 22.65
cadherin 22, type 2 Cd22 128.56
catenin (cadherin-associated protein), beta 1, 88kDa Ctnnb1 −1.41
integrin, beta 2 (complement component 3 receptor 3 and 4
subunit)
Itgb2 22.11
Protein Kinase
protein kinase C, beta Prkcb 73.34
mitogen-activated protein kinase 12 Mapk12 5.33
mitogen-activated protein kinase-activated protein kinase 3 Mapkapk3 15.96
Reactive Oxygen Species (ROS)
dual oxidase 2 Duox2 104.54
glutathione peroxidase 2 (gastrointestinal) Gpx2 102.71
Collagen
collagen, type VI, alpha 2 Col6a2 66.60
collagen, type VI, alpha 1 Col6a1 64.48
Transporters and Solute Carriers
solute carrier family 7, member 9 Slc7A9 12.70
solute carrier family 7, member 7 Slc7A7 14.15
solute carrier family 28, member 2 Slc28A2 14.08
ATP-binding cassette, sub-family A (ABC1), member 4 Abca4 33.23
Mesothelial cell marker
cytokeratin 18 Krt18 162.94
cytokeratin 19 Krt19 511.14
thrombomodulin Thbd 28.22
desmin Des 24.19
Embryonic/cell developement (cell migration,
differentiation)
placenta-specific 8 Plac8 39.53
wingless-type MMTV integration site family, member 4 Wnt4 11.04
plasminogen activator, urokinase Plau 18.00
GATA binding protein 5 Gata5 15.36
Hormone receptors
progesterone receptor Pgr 8.10
estrogen receptor 1 Esr1 2.15
estrogen receptor 2 (ER beta) Esr2 −1.23
androgen receptor Ar 1.69
Growth Arrest, Apoptosis
growth arrest and DNA-damage-inducible, beta Gadd45b −11.25
growth arrest and DNA-damage-inducible, alpha Gadd45a −2.01
BCL2-associated X protein Bax −1.53
BCL2-related protein A1 Bcl2A1 12.93
Fas apoptotic inhibitory molecule 3 Faim3 9.96
Fas ligand (TNF superfamily, member 6) Faslg 2.17

Extraction and Quantification of RNA

Extraction of RNA was performed using the Invitrogen PureLink Mini Kit (Invitrogen cat no. 12183-081A). Frozen tissue samples were lysed and homogenized in TRIzol reagent (Invitrogen, Carlsbad, CA) using a rotary-stator homogenizer. Isolation of RNA was performed according to the mini kit protocol. On-column DNase treatment was performed using the Invitrogen PureLink DNase kit (Invitrogen, Carlsbad, CA) to purify RNA samples. RNA quantification and RNA integrity number were measured on a bioanalyzer (Agilent Technologies, Santa Clara, CA). Samples were aliquoted and stored at −80°C until used.

RNA Labeling, Microarray Hybridization, and Data Processing

Gene expression analysis was conducted using Affymetrix Rat Genome 230 2.0 GeneChip® arrays (Affymetrix, Santa Clara, CA). One hundred ng of total RNA was amplified as directed in the Affymetrix 3’ IVT Express kit protocol. 12.5µg of amplified biotin-aRNAs were fragmented and 10µg were hybridized to each array for 16 hours at 45°C in a rotating hybridization oven using the Affymetrix Eukaryotic Target Hybridization Controls and protocol. Array slides were stained with streptavidin/phycoerythrin utilizing a double-antibody staining procedure and then washed for antibody amplification according to the GeneChip Hybridization, Wash and Stain Kit and user manual. Arrays were scanned in an Affymetrix Scanner 3000 and data was obtained using the GeneChip® Command Console Software (AGCC; Version 1.1) using the MAS5 algorithm to generate .CHP files.

Probe intensity data from all arrays were entered into the R software environment (http://www.R-project.org) directly from .cel files using the R/affy package (Gautier et al. 2003). Data quality was assessed using image reconstruction, intensity histograms and boxplots. Normalization was performed across all 11 samples (6 Fred-PE and 5 spontaneous mesothelioma samples) using the robust multiarray average (RMA) method to form one expression measure for each gene on each array (Irizarry et al. 2003). The RMA method adjusts the background of perfect match (PM) probes, applies a quantile normalization of the corrected PM values, and calculates final expression measures using the Tukey median polish algorithm. Treatment groups (cultured mesothelial cells (Fred-PE), spontaneous mesothelioma) were compared using a t-statistic whose null distribution was derived using 10,000 bootstrap samples by resampling the residuals (Efron and Tibshirani 1993). The bootstrap methodology was implemented in the ORIOGEN software package (Peddada et al. 2005). We applied Benjamini-Hochberg procedure (Benjamini and Hochberg 1995) for multiple testing with a nominal FDR of 0.01 to determine differential gene expression.

Core analysis of differentially expressed genes was conducted to determine biologic functions, canonical pathways, and transcription factor activation and over-represented classifications of genes were determined from statistical outcomes by testing for association with gene product relationships from a curated database of biological networks (Ingenuity Pathways Analysis™ (IPA) version 9.0) (www.ingenuity.com). The Ingenuity Pathways Knowledge Base (IPKB) consists of data with known biological relationships between genes and gene products. The significantly differentially expressed genes (p<0.001) in the IPA core analysis were then grouped by pathways to account for upstream and downstream players as well as overlapping pathways. Upstream activation (formerly known as transcription factor activation in earlier versions of IPA) was based on IPA z scores >2.0 with no bias.

Quantitative real-time PCR (qPCR)

Quantitative gene expression levels were detected using real-time PCR with the ABI PRISM 7900HT Sequence Detection System (Applied Biosystems) and TaqMan MGB probes (FAM™ dye labeled). Primers and probes for all genes analyzed were purchased from Applied Biosystems Assays-on-Demand Gene Expression products. For amplification, diluted cDNA was combined with a reaction mixture containing TaqMan universal PCR Master Mix (Applied Biosystems, Catalog No. 4304437) according to manufacturer’s instructions. Samples were analyzed in duplicate, and a sample without RT was included with each plate to detect contamination by genomic DNA. Amplification was carried out as follows: (1) 50 °C, 2 min (for uracil-N-glycosylase incubation): (2) 95 °C, 10 min (denaturation): (3) 95 °C, 15 s, 60 °C, 30 s (denaturation/amplification) for 40 cycles. Fold increases or decreases in gene expression were determined by quantitation of cDNA from target samples relative to a calibrator sample (Fred PE passage 8). The 18S RNA gene was used as the endogenous control for normalization of initial RNA levels. To determine this normalized value, 2−(ΔΔCt) values were compared between target and calibrator samples, where the changes in crossing threshold (ΔCt) = CtTarget gene − Ct18S RNA, and ΔΔCt = ΔCtcontrol − ΔCttarget.

Immunohistochemistry

Protein expression assayed by immunohistochemistry was used to characterize the immunophenotype of spontaneous rat mesotheliomas, and to validate genes associated with mesothelioma development. Formalin fixed, paraffin embedded rat tissues were deparaffinized and rehydrated and counterstained with hematoxylin, dehydrated, cleared, and coverslipped in a similar manner for all three antibodies. Endogenous peroxidase was blocked with 3% hydrogen peroxide. Antigen retrieval was performed with heat and pressure, using citrate buffer (Biocare Medical, Concord, CA). The sections were incubated with 10% normal donkey serum or normal horse serum (Jackson Immunoresearch Laboratories, Inc. West Grove, PA) for 20 minutes based on the respective antibodies, followed by the Avidin-Biotin Blocking Kit (Vector Laboratories, Burlingame, CA). The sections were incubated with rabbit monoclonal Vimentin antibody (Clone EPR3776, Epitomics, Burlingame,CA) and rabbit IgG (negative control; Epitomics, Burlingame,CA) at 1:250 dilution or Keratin 18 (Clone C-04, Santa Cruz, CA) at a 1:100 dilution and a non-immune mouse serum (negative control) diluted to match protein concentration of Keratin 18 antibody (Jackson ImmunoResearch, West Grove PA) for 60 minutes. Sections were incubated with a donkey anti-rabbit secondary antibody (Jackson Immunoresearch Laboratories, Inc. West Grove, PA) or biotinylated horse anti mouse (Vector Laboratories, Burlingame, CA) for 30 minutes at 1:500 dilution. Label incubation was performed using Vector R.T.U. Vectastain Kit (Vector Laboratories, Burlingame, CA) for 30 minutes also. Antigen-antibody complex was visualized using DAB (Dako, Carpinteria, CA) for 6 minutes. EpCAM staining was performed on Discovery XT autostainer (Ventana Medical Systems, Tucson, AZ). Standard OmniMap Kit (Ventana Medical Systems, Tucson, AZ) reagents were used. Sections were incubated with rabbit monoclonal EpCAM antibody (Clone E144, Abcam, Inc, Cambridge, MA) at 1:350 dilution and normal rabbit serum (negative control; Jackson Immunoresearch Laboratories, Inc., West Grove, PA) for 32 minutes with no heat setting. The sections were incubated with Discovery XT polymer (Ventana Medical Systems, Tucson, AZ) for 16 minutes. Antigen-antibody complex was visualized using DAB (Dako, Carpinteria, CA) for 8 minutes.

Results

Histopathology of spontaneous mesotheliomas in F344 rats

Spontaneously arising mesotheliomas from vehicle control animals (Table 1) were classified as epitheliomatous, characterized by papillary projections of one or more layers of atypical mesothelial cells overlying a fibrovascular stalk (Figure 1). Some tumors had a more solid phenotype, forming sheets of cells interspersed with variable amounts of fibrovascular stroma. Mesotheliomas frequently had mixed to mononuclear inflammatory infiltrates within the fibrovascular stroma. The mixed inflammatory component was variable from sample to sample with either a prominence of eosinophils, mast cells, or neutrophils.

Figure 1.

Figure 1

Spontaneous mesothelioma, abdominal cavity, F344/N rat. Spontaneous mesotheliomas in F344/N rats exhibited an epitheliomatous phenotype, characterized by papillary projections of plump mesothelial cells overlying a fibrovascular stalk (A, H&E, 10X). Cells were polygonal to cuboidal, with scant to moderate faintly eosinophilic cytoplasm and a central to eccentric round to oval hyperchromatic nucleus and an occasional prominent nucleoli (B, H&E, 40X).

Transcriptomic profiling identifies significant differences in global gene expression profiles between spontaneous mesothelioma and control (Fred-PE) mesothelium

Of the 21,901 genes on the microarray, 15,714 genes were differentially expressed in rat spontaneous mesotheliomas compared to Fred-PE mesothelial cells. Of these genes, 10,294 were mapped in IPA and 5,420 were unrecognized by IPA and therefore designated as unmapped. Of the mapped genes, 7,953 were considered analysis ready according to IPA based on threshold of p < 0.001 (Supplementary Table 1). Principal component analysis (PCA) of differentially expressed genes showed clear separation of spontaneous mesotheliomas from Fred-PE cells in terms of global gene expression (Figure 2). Spontaneous mesotheliomas sampled from different anatomic locations within the same animal (Cobalt) as well as from different anatomic locations and different carcinogenicity studies clustered closely together, indicating that there was little variation in gene expression profiles between these samples based on PCA. However, there was some variability in the gene expression profiles between cell lines based on PCA analysis, which may be attributable to different cell passages or other in vitro cell culture effects. Regardless, hierarchical cluster analysis (HCA) showed distinct clustering of samples within groups based on statistically significant differentially up- and downregulation of genes across the genome (Figure 3). Ingenuity Pathways Analysis (IPA) core analysis was used to group the 7953 significantly differentially expressed genes (p < 0.001) into relevant biological functions. Biological functions representative of the top up- and downregulated genes included inflammatory response, cell growth and proliferation, embryonic development, cell death, cancer, carbohydrate metabolism, cell cycle and gene expression. Many of the significantly differentially expressed genes had overlapping biological functions in multiple categories. The microarray accession can be found in the GEO database (accession # GSE47581).

Figure 2.

Figure 2

Principal Component Analysis (PCA) comparing global gene profiles of cultured Fred-PE mesothelial cells (Meso-Normal) and spontaneous mesotheliomas from control F344/N rats (CTL). PCA analysis shows clear clustering of samples within groups, indicating significant similarities in global gene expression of samples within experimental groups, and clear separation of experimental groups in space, indicating significant differences between groups in terms of their global gene expression.

Figure 3.

Figure 3

Unsupervised hierarchal cluster analysis (HCA) comparing global gene expression profiles of Fred-PE mesothelial cells (Meso-Normal) and spontaneous mesotheliomas from control F344/N rats (CTL). HCA analysis indicates significant differential expression of upregulated (red) and downregulated (green) genes across the genome between Fred-PE mesothelial cells and spontaneous F344/N rat mesotheliomas.

Global gene expression profiling of spontaneous mesotheliomas in F344/N rats identifies dysregulation of genes associated with cell proliferation and tumorigenesis

Up-regulation of genes (Table 2) associated with cell proliferation, cell motility and cell migration, cell invasion, and cell survival included growth factors (Tgfα, Tgfβ1), transporter molecules (Abcg1), solute carrier molecules (Slc7a9, Slc7a7,Slc28a2) adhesion molecules (Epcam, Cdh22), bioactive lipids receptors (Sp1r1), and oncogenes (v-Mafb, Lyn (v-yes1 homolog)), Allograft inflammatory factor 1 (Aif1), pattern recognition receptors (Tlr8, Tlr7, Mannose receptor-1) and C-type lectins (Clec4a3, Clec4A, Clec7A, Clec10A), and embryonic genes (Plac8, Plau, Wnt4, Gata5). In addition there was the upregulation of biomarkers for mesothelial cells and mesothelial cell hyperplasia (Ck18, Vim, Wt, Des, Thbd). Genes associated with up- and downstream regulation of transcription factor pathways with a Z score of >2 included interferon alpha and gamma (Ifnα, Ifnγ), signal transducer and activator of transcription 1 (Stat1), and interleukin 10 (Il10). There was downregulation of critical human tumor suppressor genes (Pten, Tp53, Rassf1, and Cdkn2a/2b), cell cycle regulators (p21, p27) and genes associated with cell proliferation (Myc, Fos, Jun). In addition, there was upregulation of anti-apoptotic factors such as Bcl2a1 and downregulation of pro-apoptotic factors such as Bax. These alterations in gene expression represent dysregulation of cell growth and proliferation pathways in mesothelial cells leading to transformation and oncogenesis in F344/N rat spontaneous mesotheliomas.

Table 2.

Select Differentially Expressed Genes Associated with Immune Response in Spontaneous Mesotheliomas in F344/N rats.

Gene Name Gene Symbol Fold Change
Chemokines
chemokine (C-C motif) ligand 11 Ccl11 875.96
chemokine (C-X-C motif) ligand 10 Cxcl10 9.45
chemokine (C-C motif) ligand 5 Ccl5 20.40
chemokine (C-C motif) ligand 6 Ccl6 79.68
chemokine (C-X-C motif) ligand 11 Cxcl11 7.27
chemokine (C-X-C motif) ligand 13 Cxcl13 167.52
chemokine (C-X-C motif) ligand 2 Cxcl2 −133.25
Cytokines & Cytokine Receptors
interleukin 1, beta Il1b 3.21
interleukin 4 receptor Il4r 20.95
interleukin 18 (interferon-gamma-inducing factor) IL18 41.81
interleukin 34 Il34 8.49
interleukin 6 receptor Il6r 7.20
interleukin 7 receptor Il7r 8.49
tumor necrosis factor receptor superfamily, member 11a, NFKB activator Tnfrsf11a 1.90
tumor necrosis factor receptor superfamily, member 11b Tnfrsf11b 45.94
interleukin 24 Il24 27.76
interleukin 10 Il10 24.21
Interleukin 1 receptor antagonist Il1rn 10.35
interleukin 23A Il23a −5.99
CD40 molecule, TNF receptor superfamily member 5 Cd40 10.29
Jak/Stat Pathway
signal transducer and activator of transcription 1, 91kDa Stat1 7.03
signal transducer and activator of transcription 2, 113kDa Stat2 4.31
signal transducer and activator of transcription 3 (acute-phase response factor) Stat3 1.77
Janus kinase 2 Jak2 6.86
Janus kinase 1 JAK1 1.60
Histocompatibility Markers
major histocompatibility complex, class II, DR alpha Hla-dra 425.51
major histocompatibility complex, class II, DQ alpha 1 Hla-dqa1 172.26
major histocompatibility complex, class II, DQ beta 1 Hla-dqb1 166.25
CD74 molecule, major histocompatibility complex, class II invariant chain Cd74 564.33
Complement
complement factor H Cfh 306.57
complement component 1, q subcomponent, B chain C1qb 115.07
complement component 1, q subcomponent, A chain C1qa 132.21
serpin peptidase inhibitor, clade G (C1 inhibitor), member 1 Serping1 303.39
Pattern Recognition Receptors
toll-like receptor 1 Tlr1 3.04
toll-like receptor 2 Tlr2 8.58
toll-like receptor 8 Tlr8 23.83
toll-like receptor 7 Tlr7 10.98
mannose receptor, C type 1 Mrc1 73.69
Interferon Pathway
interferon, alpha 16 Ifna16 −1.28
interferon gamma receptor 1 Ifngr1 2.33
interferon regulatory factor 8 Irf8 11.31
interferon regulatory factor 7 Irf7 7.42
interferon regulatory factor 9 Irf9 4.02
interferon regulatory factor 5 Irf5 2.27
interferon regulatory factor 3 Irf3 1.43
interferon induced transmembrane protein 1 Ifitm1 21.44
Inflammatory mediators/enzymes/miscellaneous
allograft inflammatory factor 1 AIF1 189.47
prostaglandin D2 synthase 21kDa Ptgds 288.68
prostaglandin-endoperoxide synthase 1 (prostaglandin G/H synthase and
cyclooxygenase)
Ptgs1 27.33
prostaglandin-endoperoxide synthase 2 (prostaglandin G/H synthase and
cyclooxygenase)
Ptgs2 −101.38
lysozyme 2 Lyz1/lyz2 269.89
mast cell protease 8 Mcpt8 249.84
hemoglobin, alpha 1 Hba1/hba2 241.31
tryptophan 2,3-dioxygenase Tdo2 212.85
ubiquitin D Ubd 191.62
carbonic anhydrase IV Ca4 159.31
DEAD (Asp-Glu-Ala-Asp) box polypeptide 60 Ddx60 149.65
cytochrome b-245, beta polypeptide Cybb 134.29
phospholipase A2, group IIA Pla2g2a 96.46
lymphatic vessel endothelial hyaluronan receptor 1 Lyve1 81.91
Activated macrophage products
chitinase 3-like 1 Chi3l1 149.52
SPARC-like 1 (hevin) Sparcl1 71.34
complement component 1, q subcomponent, B chain C1qb 115.07
complement component 1, q subcomponent, A chain C1qa 132.21
Cell surface markers/receptors
sphingosine-1-phosphate receptor 1 S1pr1 24.39
Fc fragment of IgG, low affinity IIb, receptor (CD32) Fcgr2b 198.66
Fc fragment of IgE, high affinity I, receptor for; alpha polypeptide Fcer1a 38.51
Fc fragment of IgE, high affinity I, receptor for; gamma polypeptide Fcer1g 117.78
Fc fragment of IgG, low affinity IIIa, receptor (CD16a) Fcgr3a 57.05
Stabilin 1 Stab1 60.71
Fc fragment of IgG, high affinity Ia, receptor (CD64) Fcgr1a 30.53
Fc fragment of IgG, low affinity IIa, receptor (CD32) Fcgr2a 51.26
CD 163 molecule Cd163 49.51
CD 68 molecule Cd68 32.09
CD 53 molecule Cd53 130.44
C-type lectin domain family 4, member a3 Clec4a3 123.54
C-type lectin domain family 10, member A Clec10a 46.75
C-type lectin domain family 4, member A Clec4a 36.02
C-type lectin domain family 7, member A Clec7a 28.97
CD36 molecule (thrombospondin receptor) Cd36 148.28

Alterations in inflammatory and immune cell function are overrepresented in global gene expression profiles of spontaneous F344/N rat mesotheliomas

The top up- and downupregulated genes from the 7953 analyzed were associated with inflammatory/immune response (Table 3), and included cytokines (Il18, Il10, Il24) and chemokines (Ccl11, Cxcl13, Ccl5), inflammatory pathways/products of inflammatory cells (arachidonic acid metabolism, Mcpt8, Lyz1/2, Chi3l1), major histocompatibility complexes (Hla-dra, Hla-dqa1, Hla-dqb1), the complement pathway (Cfh, C1qb, Serping1), integrins and adhesion molecules (Cdh22, Itgb2), and oxidative stress/response to reactive oxygen species (ROS) (Duox2, Gpx2). Many of the genes with greater than 5 fold change in expression were inflammatory mediators associated with macrophage activation and maturation, lymphocyte (T and B cell), leukocyte, and NK cell activation and recruitment, and immune evasion (Il18, Il24, Il10, Cd40, Col6a1, Cd68, Cd163, Cd53, Cd74, Stab1). Select differentially expressed genes were validated by quantitative RT-PCR (qPCR) (Table 4). Genes were selected based on known involvement with cancer progression, embryonic development, cell survival/decreased apoptosis, mesothelial cell function, immune response or for their role as a tumor biomarker. In all cases, RNA expression of selected genes matched the same directional change observed in the microarray.

Table 3.

qPCR validation of select up- and downregulated genes in spontaneous mesotheliomas in F344/N rats.

Entrez Gene Name Gene Symbol qRT-PCR Microarray
GATA binding protein 5 Gata5 604.09 14.2
Interleukin-10 Il10 697.23 24.21
Interleukin-18 Il18 159.52 41.81
Glutathione peroxidase 2 Gpx2 484.44 102.71
Cytokeratin 18 Krt18 1079.19 162.94
Tumor necrosis factor receptor 11b Tnfr11b 729.76 45.94
Plasminogen activator, urokinase Plau 278.72 17.9
Wingless-type MMTV integration site family, member 4 Wnt4 7.84 11.19
CD40 molecule, TNF receptor superfamily member 5 Cd40 843.49 10.18
Epithelial adhesion molecule Epcam 481.96 22.65
Protein kinase C, beta Prkcb 274.06 73.34
Transforming Growth Factor alpha Tgf-a 12.38 12.45
Cyclin dependent kinase 1a (p21) Cdkn1a −2.88 −4.53
Fatty acid binding protein 4 Fabp4 2705.77 68.98
Placenta-specific 8 Plac8 16744.15 39.53
RAS, dexamethasone-induced 1 Rasd1 86.8 39.86
Tumor protein p53 Tp53 −1.11 −2.25

Table 4.

qPCR validation of select up- and downregulated genes in spontaneous mesotheliomas in F344/N rats.

Entrez Gene Name Gene Symbol qRT-PCR Microarray
GATA binding protein 5 Gata5 604.09 14.2
Interleukin-10 Il10 697.23 24.21
Interleukin-18 Il18 159.52 41.81
Glutathione peroxidase 2 Gpx2 484.44 102.71
Cytokeratin 18 Krt18 1079.19 162.94
Tumor necrosis factor receptor 11b Tnfr11b 729.76 45.94
Plasminogen activator, urokinase Plau 278.72 17.9
Wingless-type MMTV integration site family, member 4 Wnt4 7.84 11.19
CD40 molecule, TNF receptor superfamily member 5 Cd40 843.49 10.18
Epithelial adhesion molecule Epcam 481.96 22.65
Protein kinase C, beta Prkcb 274.06 73.34
Transforming Growth Factor alpha Tgf-a 12.38 12.45
Cyclin dependent kinase 1a (p21) Cdkn1a −2.88 −4.53
Fatty acid binding protein 4 Fabp4 2705.77 68.98
Placenta-specific 8 Plac8 16744.15 39.53
RAS, dexamethasone-induced 1 Rasd1 86.8 39.86
Tumor protein p53 Tp53 −1.11 −2.25

Downstream protein expression of spontaneous F344/N rat mesotheliomas confirm a biphasic phenotype and share protein markers with human mesothelioma

Spontaneous mesotheliomas in the current study were evaluated immunohistochemically with antibodies to cytokeratin 18 (CK18) and vimentin (VIM) (Figure 4). Rat spontaneous mesotheliomas strongly expressed both vimentin (VIM) and cytokeratin 18 (CK18) (Table 1), and protein expression correlated positively with directional fold change in gene expression on the microarray. Expression of CK18 was restricted to the cell membrane in mesotheliomas, similar to expression in positive controls (rat salivary gland) compared to negative controls. Vimentin was co-expressed in the cytoplasm of mesothelioma cells, and expression was similar to positive controls (rat kidney). Mesotheliomas expressed epithelial cell adhesion molecule (EpCAM) (Table 1), a protein involved in the maintenance of tight junctions that is overexpressed in a variety of human cancers including mesothelioma. However, while expression of EpCAM was restricted to the cell membrane in positive controls (rat gastrointestinal tract mucosa), in spontaneous rat mesotheliomas, there was translocation of protein to the cytoplasm and nucleus, suggesting dysregulation of this protein (Figure 4).

Figure 4.

Figure 4

Immunohistochemical validation of mesothelioma markers in spontaneous mesotheliomas from control F344/N rats. Neoplastic cells in spontaneous mesotheliomas (A, C, E, 10X; B, D, F, 40X) showed strong membrane immunoreactivity with anti-cytokeratin 18 antibody (A–B), strong cytoplasmic immunoreactivity with anti-vimentin antibody (C–D), and strong cytoplasmic and nuclear immunoreactivity for antibodies to epithelial cell adhesion molecule (E–F) (hematoxylin counterstain).

Discussion

Our study has characterized the genomic profile of spontaneously occurring peritoneal mesotheliomas in male Fisher 344/N rats. In their roles as a primary barrier to pathogens and maintenance of peritoneal homeostasis, mesothelial cells have a wide range of initiation and response mechanisms associated with cellular growth, cytokine and growth factor release, regulation of pattern recognition receptors, and pathogen-associated molecular patterns (Izzi et al. 2012; Wu et al. 2010; Yung and Chan 2007). Carcinogenesis is a very complex process involving dysregulation of multiple cell growth and proliferation pathways, metabolic perturbations, inflammation and immune dysfunction, and hormonal factors, and consistent with this, we have shown that the genomic profiles observed in spontaneous F344/N rat mesotheliomas are similarly complex, involving dysregulation of numerous intersecting and interrelated pathways related to, cell growth and proliferation, contributions by inflammatory mediators, and interactions with reactive oxygen species and free radicals, all well-known and contributory mechanisms in carcinogenesis.

Oncogenes, Tumor Suppressor Genes, and Apoptosis Mediators

Alterations in tumor suppressor genes and oncogenes are key genetic factors related to tumor development and progression. Spontaneous F344/N rat mesotheliomas showed downregulation of several tumor suppressor genes (Tp53, Pten, Gadd45A, Cdkn2b, Cdnk1a, Cdnk1b) and overexpression of oncogenes (v-MafB, v-Yes). MAFB is overexpressed in human mesothelioma (Gordon et al. 2005), and c-YES is a mediator of cell proliferation in human malignant mesothelioma cells (Sato et al. 2012). In addition, a variety of tumor suppressor genes (CKDN2A, TP53, PTEN, NF2, BAP1) and their downstream targets are similarly affected in human mesothelioma (Bueno et al. 2010; Gordon et al. 2005; Gueugnon et al. 2011; Tsujimura et al. 2012). While promoters of cell cycle progression and cellular proliferation such as FOS, MYC, JUN and CCND1 are typically upregulated in asbestos associated human mesothelioma, these genes were downregulated in spontaneous rat mesothelioma in this study. Lastly, spontaneous rat mesotheliomas were associated with an anti-apoptotic phenotype characterized by the overexpression of anti-apoptotic Bcl mediators (Bcl2a1), downregulation pro-apoptotic Bcl members (Bax) and Fadd and Fas mediators, and upregulation of Fas apoptotic inhibitory molecule 3 (Faim3). Alterations in tumor suppressor genes, oncogenes, and apoptosis mediators have a profound effect on the expression of downstream cellular growth factors and the inflammatory response, two other significantly represented categories of altered gene expression in this study.

Growth Factor Pathways

Dysregulation of cellular growth is an integral part of carcinogenesis. In spontaneous F344/N rat mesotheliomas, there was upregulation of mediators in a number of growth factor pathways, including the transforming growth factor-alpha and -beta (Tgfα/β), insulin-like growth factor (Igf), p38 mitogen-associated protein kinase (p38 Mapk) and nuclear factor kappa-B (Nfκb) pathways. TGFβ was originally discovered as a mediator involved in tumor cell survival (Abbas and Lichtman 2003), regulates insulin-like growth factor-1 (IGF1) and inhibits cell adhesion and enhances cell-collagen interactions associated with invasion and metastasis. TGFα is associated with positive regulation of cell proliferation, differentiation and development, and negatively regulates apoptosis. Upregulation of TGFα/β is associated with a variety of cancers, including breast, liver, kidney, lung, pancreatic, and hematologic cancers. In some cancers, TGFβ plays conflicting roles in transformation and tumor progression; for example, it has been shown to act as a tumor suppressor in early stages of breast cancer development, but acts to promote invasion and metastasis late in the course of disease (Akhurst and Derynck 2001; Tang et al. 2003). TGFα and β are also involved in a large number of physiologic processes, and interface with other growth factor pathways such as the MAP-kinase (MAPK) and NFκB signaling pathways. In addition, TGFβ1 is produced by and regulates cells of the immune system (Letterio and Roberts 1998; Yang et al. 2010), representing another overlapping component of the interlacing pathways associated with mesotheliomas in this study. Several members of the insulin like growth factor (Igf) pathway were upregulated in spontaneous mesotheliomas (Igf1, Igfbp1, Igfbp2–7), similar to a variety of human cancers including mesothelioma, where they are associated with high-grade or poorly differentiated tumors (Gullu et al. 2012; Jean et al. 2012; Whitson and Kratzke 2006). The p38 mitogen activated protein kinase (p38 Mapk) pathway is regulated by a wide variety of molecules and conditions including growth factors, cytokines, cytosolic proteins and environmental stresses (Cuadrado and Nebreda 2010). Several mediators within the p38 Mapk pathway (p38 Mapkα/β/γ, Stat1, Tgfβr1, Tgfβ1, Tnfrs11b, Tnfrs14, Tnfrs25) were up regulated in spontaneous mesotheliomas. Mediators in this pathway are involved in cell survival and proliferation, and there is moderate crosstalk between other growth pathways such as the NFkB, WNT, JNK and AKT pathways (Cuadrado and Nebreda 2010). Moreover, p38 Mapk is activated in rat mesothelial cells exposed to asbestos (Swain et al. 2004), and regulates cell proliferation in rat pleural mesothelioma cell lines (Zhong et al. 2011). The various p38 Mapk isoforms act across many pathways, and may be activated by environmental stress (oxidative stress, UV irradiation, hypoxia, ischemia) and inflammatory cytokines such as IL1 and TNFα (Cargnello and Roux 2011).

Epithelial cell adhesion molecule (Epcam) is a trans membrane p-glycoprotein reported to have multiple roles including cell signaling, migration, proliferation, and differentiation (Patriarca et al. 2012). It is a well-known epithelial tumor marker that is overexpressed in breast, bladder, colorectal, pancreas, prostate and ovarian cancer, and epitheliomatous mesotheliomas in humans (Ryan et al. 1997; Shield et al. 1994). Alterations in EpCAM interfere with cadherin-mediated cell to cell adhesion (Litvinov et al. 1997), and cytoplasmic and nuclear translocation of its intercellular domain (EpICD) is associated with stimulation of cell proliferation (Maetzel et al. 2009). In addition to up regulation of Epcam at the gene expression level, there was cytoplasmic and nuclear translocation of EpCAM protein in spontaneous rat mesotheliomas in this study (Figure 4); the EpCAM antibody used in this study recognizes the C terminus on EpICD (Patriarca et al. 2012), confirming the nuclear translocation of the intercellular domain and implicating EpCAM as mediator of cell proliferation and mesotheliomagenesis in both F344/N rats and humans. It is well known that mesothelial cells have a biphasic nature, expressing a broad spectrum of low and high molecular weight epithelial keratins and vimentins (Mullink et al. 1986; Whitaker et al. 1980). As such, spontaneous mesotheliomas in this study expressed both vimentin and CK18 by immunohistochemistry.

Inflammatory Pathways and Immune Dysfunction

Inflammation and immune dysregulation are mechanisms central to many neoplastic processes, including the development of mesotheliomas, and are considered the seventh hallmark of cancer (Colotta et al. 2009; Hanahan and Weinberg 2011). In fact, a key component of early stage asbestos-related mesothelioma in humans is a non-specific inflammatory response (Boutin and Rey 1993). Given the nature of the disease in humans, a chronic inflammatory response is not surprising. What is interesting however, is the predominance of genomic changes associated with altered immune function and inflammation in spontaneous rat mesotheliomas, which arise in the absence of any initiating chemical or physical agent. This response could be attributed to chronic inflammation, cancer-related inflammation, dysregulation of mesothelial cell function, and/or intrinsic genetic events. Mesothelial cell proliferation may be stimulated through the induction of proinflammatory pathways by activated macrophages or mesothelial cells, with the subsequent production of proinflammatory cytokines (TNF, IL-1) and growth factors (IL6, GM-CSF), upregulation of prostaglandins, nitrous oxide (NO), angiogenic factors, and adhesion molecules. Research on the etiology of mesotheliomas in humans (asbestos) and animal models (multiwalled carbon nanotubes, MWCNT) suggest that a major pathophysiologic factor in the development of these tumors is frustrated phagocytosis leading to biopersistence of the initiating agent, which results in chronic oxidative stress and inflammation (Walker et al. 1992; Yang et al. 2008). Similarly, rat spontaneous mesotheliomas in this study were associated with upregulation of a variety of cytokines and cytokine receptors associated with chronic inflammation (Il6r, Tnfrs11b, Tnfrs14, Tnfrs25, and Il1b), including Il10, which promotes cell survival and abrogation of immune surveillance. It is known that asbestos in fact can cause alterations in normal cells that result in decreased tumor immunity (Matsuzaki et al. 2012). Whether these are contributing factors in the induction and pathogenesis of this tumor in rats, or merely secondary to the presence of the neoplasm and the tissue damage associated with its invasion is uncertain. Certainly, the chronic tissue damage and inflammation associated with asbestos exposure in human mesotheliomagenesis is due to the persistence of the asbestos fiber and its interaction with cellular components within the lung parenchyma, ultimately leading to generation of reactive oxygen species and secondary DNA damage (Matsuzaki et al. 2012). In terms of non-asbestos etiologies in human mesothelioma, many of these are also associated with inflammatory processes in the lung (SV40 viral infection, exposure to radiation or chemical agents (Carbone and Bedrossian 2006; Yang et al. 2008); whereas others, such as germline mutations in BRCA1-Associated Protein 1 (BAP1) (Bott et al. 2011; Testa et al.), arise without such associated tissue damage and inflammation. Similarly, mesothelioma in vehicle control F344/N rats arises spontaneously without an inciting cause related to tissue damage or inflammation, and may represent an inherent genetic susceptibility that could be influenced by chemical exposure.

Many of the receptors associated with innate immunity that are constitutively expressed in mesothelial cells were upregulated in spontaneous mesotheliomas in this study. Pleural and peritoneal mesothelial cells express a wide range of pattern recognition receptors (PRR), such as the toll-like receptors (TLRs), which are triggered by pathogen activated molecular patterns (PAMPs), including components of bacteria, fungi, or viruses, and are therefore used in defense systems employed by the mesothelium (Takeuchi and Akira 2010). Toll-like receptors are pattern recognition receptors that trigger activation of the NFkB pathway and/or MAPK activation, and increased production of proinflammatory cytokines and interferons (Takeuchi and Akira 2010). The genomic profile of spontaneous rat mesotheliomas is highlighted by the prominent upregulation of pattern recognition receptors (TLR1, 2, 7, 8, Mrc1) that trigger activation of the TLR-NFkB and interferon pathways, with subsequent release of inflammatory cytokines and other inflammatory mediators. Endogenous TLR ligands also have an increasing role in tumorigenesis through promotion of cancer cell survival and proliferation, immune system evasion, and enhancemenet of metastasis and chemoresistance (Yu et al. 2012). Endogenous molecules that are released from mesothelial cells following tissue injury can act as danger associated molecular pattern receptors (DAMPs) and activate TLRs (Yu et al. 2012). Several mediators that act as DAMPs (Mrc1, S100a8/9, hyaluronan, Lyve1, Stab1), were upregulated in spontaneous rat mesotheliomas; these mediators can promote tumorigenesis and invasion through the activation of various TLRs (Turovskaya et al. 2008; Voelcker et al. 2008; Yu et al. 2012). Spontaneous rat mesotheliomas were also associated with upregulation of interferon pathway genes, associated with immunomodulation (If135, Psmb8, Ifitm1, Tap1, Aif1, Irf1/7/9). Overexpression of some of these immunomodulatory genes are also associated with cell proliferation, inhibition of apoptosis, and tumor growth (Liu et al. 2008), and upregulation of the interferon receptor pathway results in stimulation of a variety of other cell growth signaling networks including the Jak/Stat pathway, glucocorticoid pathway, and immune function and cytokine expression. This suggests a significant inflammatory component to the process of spontaneous mesothelioma development in these animals.

Evidence supporting the hypothesis that the immune system plays an important role in modulating cancer has been increasing over the last decade. Indeed, in addition to their role as activators of the immune response, protumorigenic activity has been described for upregulated TLRs in cancer cells or tumor associated macrophages (Yu et al. 2012). Moreover, TLR stimulation has been associated with tumor promotion and invasion in rodents and humans (Ochi et al. 2012; Xie et al. 2010). Mediators expressed by activated and/or tumor associated macrophages have been shown to have tumor promoting qualities (Traves et al. 2012). Tumor associated macrophages (TAMs) are present in variety of cancer types and are associated with tumor progression, tumor cell invasion, metastasis, and angiogenesis (Pollard 2004). Cell surface markers associated with TAMs in rat spontaneous mesotheliomas in the current study included Cd163, Cd68, Cd53, and C type lectins (Cd93), many of which play roles in cell-cell and cell-matrix interactions (Bos et al. 2012), cell adhesion, migration, and inflammation (Greenlee-Wacker et al. 2012), pro-survival signaling through the AKT/PIK3 pathway (Yunta and Lazo 2003), or are expressed by tumor cells and TAMs in a variety of human cancers (Maniecki et al. 2012; Shabo and Svanvik 2011;Ch’ng et al. 2013; Feng et al. 2012; Jia et al. 2008). The upregulation of these cell surface markers in spontaneous rat mesotheliomas may be due to overexpression by mesothelioma cell themselves, or due to the presence of activated or tumor-associated macrophages within these tumors.

In this study, a complex interaction of dysregulated mesothelial cell processes including growth, differentiation, and immune system function was observed. It is clear that mesothelioma in humans (Yung and Chan 2007) and rats (Xu et al. 2012) is associated with risk factors of chronic inflammation; however, in spontaneous mesothelioma, the prominent inflammatory phenotype is unexpected, and it is unclear as to whether the prominent immune cell response was a contributing factor for the development of mesothelioma, or merely a response to the presence of the neoplasms in these animals.

Conclusion

This study shows that there is dysregulation of numerous genes in overlapping pathways including those associated with cellular proliferation and survival, inflammation and immune system dysregulation, inhibition of apoptosis, and dysregulation of oncogenes, tumor suppressor and embryonic genes in the process of spontaneous mesotheliomagenesis. Importantly, this dataset now provides a point of reference to better understand the events that occur in chemically induced mesothelioma in the F344/N rat, and how these events may be relevant to human hazard characterization.

Supplementary Material

Sup1

Acknowledgments

We acknowledge Norris Flagler, Eli Ney, and Beth Mahler for their imaging expertise and assistance with figures. We would like to thank the CMPB Histology and Immunohistochemistry Core Laboratories, and the NIEHS Microarray Core for their technical expertise.

Author’s note: This work was supported by the National Institutes of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), and The Division of the National Toxicology Program (DNTP). This article may be the work product of an employee or group of employees of the National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), however, the statements, opinions or conclusions contained therein do not necessarily represent the statements, opinions or conclusions of the NIEHS, NIH, or the United States government.

References

  1. Abbas AK, Lichtman AH. Cellular and Molecular Immunology. Philadelphia: Saunders; 2003. [Google Scholar]
  2. Afify AM, Al-Khafaji BM, Paulino AF, Davila RM. Diagnostic use of muscle markers in the cytologic evaluation of serous fluids. Appl.Immunohistochem.Mol.Morphol. 2002;10:178–182. doi: 10.1097/00129039-200206000-00014. [DOI] [PubMed] [Google Scholar]
  3. Akhurst RJ, Derynck R. TGF-beta signaling in cancer--a double-edged sword. Trends Cell Biol. 2001;11:S44–S51. doi: 10.1016/s0962-8924(01)02130-4. [DOI] [PubMed] [Google Scholar]
  4. Antony VB. Immunological mechanisms in pleural disease. European Respiratory Journal. 2003;21:539–544. doi: 10.1183/09031936.03.00403902. [DOI] [PubMed] [Google Scholar]
  5. Benjamini Y, Hochberg Y. Controlling the false discovery rate:apractical and powerful approach to multiple testing. 1995:289–300. [Google Scholar]
  6. Blobel GA, Moll R, Franke WW, Kayser KW, Gould VE. The intermediate filament cytoskeleton of malignant mesotheliomas and its diagnostic significance. American Journal of Pathology. 1985;121:235–247. [PMC free article] [PubMed] [Google Scholar]
  7. Bos SD, Lakenberg N, van der BR, Houwing-Duistermaat JJ, Kloppenburg M, de Craen AJ, Beekman M, Meulenbelt I, Slagboom PE. A genome-wide linkage scan reveals CD53 as an important regulator of innate TNF-alpha levels. European Journal of Human Genetics. 2012;18:953–959. doi: 10.1038/ejhg.2010.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bott M, Brevet M, Taylor BS, Shimizu S, Ito T, Wang L, Creaney J, Lake RA, Zakowski MF, Reva B, Sander C, Delsite R, Powell S, Zhou Q, Shen R, Olshen A, Rusch V, Ladanyi M. The nuclear deubiquitinase BAP1 is commonly inactivated by somatic mutations and 3p21.1 losses in malignant pleural mesothelioma. Nat Genet. 2011;43:668–672. doi: 10.1038/ng.855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Boutin C, Rey F. Thoracoscopy in pleural malignant mesothelioma: a prospective study of 188 consecutive patients. Part 1: Diagnosis. Cancer. 1993;72:389–393. doi: 10.1002/1097-0142(19930715)72:2<389::aid-cncr2820720213>3.0.co;2-v. [DOI] [PubMed] [Google Scholar]
  10. Bueno R, De Rienzo A, Dong L, Gordon GJ, Hercus CF, Richards WG, Jensen RV, Anwar A, Maulik G, Chirieac LR, Ho KF, Taillon BE, Turcotte CL, Hercus RG, Gullans SR, Sugarbaker DJ. Second generation sequencing of the mesothelioma tumor genome. PLoS One. 2010;5:e10612. doi: 10.1371/journal.pone.0010612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Carbone M, Bedrossian CW. The pathogenesis of mesothelioma. Seminars in Diagnostic Pathology. 2006;23:56–60. doi: 10.1053/j.semdp.2006.08.002. [DOI] [PubMed] [Google Scholar]
  12. Carbone M, Pass HI, Miele L, Bocchetta M. New developments about the association of SV40 with human mesothelioma. Oncogene. 2003;22:5173–5180. doi: 10.1038/sj.onc.1206552. [DOI] [PubMed] [Google Scholar]
  13. Cargnello M, Roux PP. Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases. Microbiol Mol Biol Rev. 2011;75:50–83. doi: 10.1128/MMBR.00031-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Ch’ng ES, Tuan Sharif SE, Jaafar H. In human invasive breast ductal carcinoma, tumor stromal macrophages and tumor nest macrophages have distinct relationships with clinicopathological parameters and tumor angiogenesis. Virchows Arch. 2013;462:257–267. doi: 10.1007/s00428-012-1362-4. [DOI] [PubMed] [Google Scholar]
  15. Colotta F, Allavena P, Sica A, Garlanda C, Mantovani A. Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis. 2009;30:1073–1081. doi: 10.1093/carcin/bgp127. [DOI] [PubMed] [Google Scholar]
  16. Crosby LM, Hyder KS, DeAngelo AB, Kepler TB, Gaskill B, Benavides GR, Yoon L, Morgan KT. Morphologic analysis correlates with gene expression changes in cultured F344 rat mesothelial cells. Toxicology and Applied Pharmacology. 2000;169:205–221. doi: 10.1006/taap.2000.9049. [DOI] [PubMed] [Google Scholar]
  17. Cuadrado A, Nebreda AR. Mechanisms and functions of p38 MAPK signalling. Biochem J. 2010;429:403–417. doi: 10.1042/BJ20100323. [DOI] [PubMed] [Google Scholar]
  18. Efron B, Tibshirani R. An Introduction to the bootstrap. 1993. [Google Scholar]
  19. Feng F, Wu Y, Zhang S, Liu Y, Qin L, Yan Z, Wu W. Macrophages facilitate coal tar pitch extract-induced tumorigenic transformation of human bronchial epithelial cells mediated by NF-kappaB. PLoS One. 2012;7:e51690. doi: 10.1371/journal.pone.0051690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Gautier L, Bolstad BM, Irizarry RA. Affy-Analysis od Affymetrix GeneCHip probe level data. Bioinformatics. 2003;20:307–315. doi: 10.1093/bioinformatics/btg405. [DOI] [PubMed] [Google Scholar]
  21. Gordon GJ, Rockwell GN, Jensen RV, Rheinwald JG, Glickman JN, Aronson JP, Pottorf BJ, Nitz MD, Richards WG, Sugarbaker DJ, Bueno R. Identification of novel candidate oncogenes and tumor suppressors in malignant pleural mesothelioma using large-scale transcriptional profiling. American Journal of Pathology. 2005;166:1827–1840. doi: 10.1016/S0002-9440(10)62492-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Greenlee-Wacker MC, Galvan MD, Bohlson SS. CD93: recent advances and implications in disease. Curr Drug Targets. 2012;13:411–420. doi: 10.2174/138945012799424651. [DOI] [PubMed] [Google Scholar]
  23. Gueugnon F, Leclercq S, Blanquart C, Sagan C, Cellerin L, Padieu M, Perigaud C, Scherpereel A, Gregoire M. Identification of novel markers for the diagnosis of malignant pleural mesothelioma. Am J Pathol. 2011;178:1033–1042. doi: 10.1016/j.ajpath.2010.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Gullu G, Karabulut S, Akkiprik M. Functional roles and clinical values of insulin-like growth factor-binding protein-5 in different types of cancers. Chin J Cancer. 2012;31:266–280. doi: 10.5732/cjc.011.10405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Hall WC, Boorman G, Eustis S, Elwell MR, Montgomery CA, MacKenzie WF. Pathology of the Fischer Rat. San Diego, CA: Academic Press; 1990. Peritoneum, Retroperitoneum, Mesentery, and Abdominal Cavity. [Google Scholar]
  26. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
  27. Hu Q, Akatsuka S, Yamashita Y, Ohara H, Nagai H, Okazaki Y, Takahashi T, Toyokuni S. Homozygous deletion of CDKN2A/2B is a hallmark of iron-induced high-grade rat mesothelioma. Lab Invest. 2010;90:360–373. doi: 10.1038/labinvest.2009.140. [DOI] [PubMed] [Google Scholar]
  28. Irizarry RA, Bolstad BM, Collin F, Cope LM, Hobbs B, Speed TP. Summaries of Affymetrix GeneChip probe level data. Nucleic Acids Research. 2003;31:e15. doi: 10.1093/nar/gng015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Izzi V, Masuelli L, Tresoldi I, Foti C, Modesti A, Bei R. Immunity and malignant mesothelioma: from mesothelial cell damage to tumor development and immune response-based therapies. Cancer Lett. 2012;322:18–34. doi: 10.1016/j.canlet.2012.02.034. [DOI] [PubMed] [Google Scholar]
  30. Jantz MA, Antony VB. Pathophysiology of the pleura. Respiration. 2008;75:121–133. doi: 10.1159/000113629. [DOI] [PubMed] [Google Scholar]
  31. Jean D, Daubriac J, Le Pimpec-Barthes F, Galateau-Salle F, Jaurand MC. Molecular changes in mesothelioma with an impact on prognosis and treatment. Arch Pathol Lab Med. 2012;136:277–293. doi: 10.5858/arpa.2011-0215-RA. [DOI] [PubMed] [Google Scholar]
  32. Jia J, Bai Y, Fu K, Sun ZJ, Chen XM, Zhao YF. Expression of allograft inflammatory factor-1 and CD68 in haemangioma: implication in the progression of haemangioma. British Journal of Dermatology. 2008;159:811–819. doi: 10.1111/j.1365-2133.2008.08744.x. [DOI] [PubMed] [Google Scholar]
  33. Kaufman AJ, Pass HI. Current concepts in malignant pleural mesothelioma. Expert.Rev.Anticancer Ther. 2008;8:293–303. doi: 10.1586/14737140.8.2.293. [DOI] [PubMed] [Google Scholar]
  34. Kim Y, Ton TV, DeAngelo AB, Morgan K, Devereux TR, Anna C, Collins JB, Paules RS, Crosby LM, Sills RC. Major carcinogenic pathways identified by gene expression analysis of peritoneal mesotheliomas following chemical treatment in F344 rats. Toxicology and Applied Pharmacology. 2006;214:144–151. doi: 10.1016/j.taap.2005.12.009. [DOI] [PubMed] [Google Scholar]
  35. Kupryjanczyk J, Karpinska G. Desmin expression in reactive mesothelium: a potential aid in evaluation of gynecologic specimens. International Journal of Gynecological Pathology. 1998;17:123–128. [PubMed] [Google Scholar]
  36. Letterio JJ, Roberts AB. Regulation of immune responses by TGF-beta. Annual Review of Immunology. 1998;16:137–161. doi: 10.1146/annurev.immunol.16.1.137. 137–161. [DOI] [PubMed] [Google Scholar]
  37. Litvinov SV, Balzar M, Winter MJ, Bakker HA, Briaire-de BI, Prins F, Fleuren GJ, Warnaar SO. Epithelial cell adhesion molecule (Ep-CAM) modulates cell-cell interactions mediated by classic cadherins. Journal of Cell Biology. 1997;139:1337–1348. doi: 10.1083/jcb.139.5.1337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Liu S, Tan WY, Chen QR, Chen XP, Fu K, Zhao YY, Chen ZW. Daintain/AIF-1 promotes breast cancer proliferation via activation of the NF-kappaB/cyclin D1 pathway and facilitates tumor growth. Cancer Sci. 2008;99:952–957. doi: 10.1111/j.1349-7006.2008.00787.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Mackay AM, Tracy RP, Craighead JE. Intermediate filament proteins in asbestos-induced mesotheliomas of the rat. Cancer Res. 1987;47:5461–5468. [PubMed] [Google Scholar]
  40. Madison LD, Bergstrom-Porter B, Torres AR, Shelton E. Regulation of surface topography of mouse peritoneal cells. Formation of microvilli and vesiculated pits on omental mesothelial cells by serum and other proteins. Journal of Cell Biology. 1979;82:783–797. doi: 10.1083/jcb.82.3.783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Maetzel D, Denzel S, Mack B, Canis M, Went P, Benk M, Kieu C, Papior P, Baeuerle PA, Munz M, Gires O. Nuclear signalling by tumour-associated antigen EpCAM. Nat.Cell Biol. 2009;11:162–171. doi: 10.1038/ncb1824. [DOI] [PubMed] [Google Scholar]
  42. Maniecki MB, Etzerodt A, Ulhoi BP, Steiniche T, Borre M, Dyrskjot L, Orntoft TF, Moestrup SK, Moller HJ. Tumor-promoting macrophages induce the expression of the macrophage-specific receptor CD163 in malignant cells. Int J Cancer. 2012;131:2320–2331. doi: 10.1002/ijc.27506. [DOI] [PubMed] [Google Scholar]
  43. Matsuzaki H, Maeda M, Lee S, Nishimura Y, Kumagai-Takei N, Hayashi H, Yamamoto S, Hatayama T, Kojima Y, Tabata R, Kishimoto T, Hiratsuka J, Otsuki T. Asbestos-induced cellular and molecular alteration of immunocompetent cells and their relationship with chronic inflammation and carcinogenesis. J Biomed Biotechnol. 2012;2012:492608. doi: 10.1155/2012/492608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Mullink H, Henzen-Logmans SC, Alons-van Kordelaar JJ, Tadema TM, Meijer CJ. Simultaneous immunoenzyme staining of vimentin and cytokeratins with monoclonal antibodies as an aid in the differential diagnosis of malignant mesothelioma from pulmonary adenocarcinoma. Virchows Arch B Cell Pathol Incl Mol Pathol. 1986;52:55–65. doi: 10.1007/BF02889950. [DOI] [PubMed] [Google Scholar]
  45. Nasreen N, Mohammed KA, Hardwick J, Van Horn RD, Sanders K, Kathuria H, Loghmani F, Antony VB. Low molecular weight hyaluronan induces malignant mesothelioma cell (MMC) proliferation and haptotaxis: role of CD44 receptor in MMC proliferation and haptotaxis. Oncology Research. 2002;13:71–78. [PubMed] [Google Scholar]
  46. National Toxicology Program, N. NTP Historical Controls Report: All Routes and Vehicles, F344/N Rats. 2011 May;:9. [Google Scholar]
  47. Ochi A, Graffeo CS, Zambirinis CP, Rehman A, Hackman M, Fallon N, Barilla RM, Henning JR, Jamal M, Rao R, Greco S, Deutsch M, Medina-Zea MV, Bin Saeed U, Ego-Osuala MO, Hajdu C, Miller G. Toll-like receptor 7 regulates pancreatic carcinogenesis in mice and humans. J Clin Invest. 2012;122:4118–4129. doi: 10.1172/JCI63606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Patriarca C, Macchi RM, Marschner AK, Mellstedt H. Epithelial cell adhesion molecule expression (CD326) in cancer: a short review. Cancer Treat Rev. 2012;38:68–75. doi: 10.1016/j.ctrv.2011.04.002. [DOI] [PubMed] [Google Scholar]
  49. Peddada S, Harris S, Zajd J, Harvey E. ORIOGEN: order restricted inference for ordered gene expression data. Bioinformatics. 2005;21:3933–3934. doi: 10.1093/bioinformatics/bti637. [DOI] [PubMed] [Google Scholar]
  50. Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nat.Rev.Cancer. 2004;4:71–78. doi: 10.1038/nrc1256. [DOI] [PubMed] [Google Scholar]
  51. Remon J, Lianes P, Martinez S, Velasco M, Querol R, Zanui M. Malignant mesothelioma: New insights into a rare disease. Cancer Treat Rev. 2012 doi: 10.1016/j.ctrv.2012.12.005. [DOI] [PubMed] [Google Scholar]
  52. Robinson BW, Lake RA. Advances in malignant mesothelioma. New England Journal of Medicine. 2005;353:1591–1603. doi: 10.1056/NEJMra050152. [DOI] [PubMed] [Google Scholar]
  53. Romagnoli S, Fasoli E, Vaira V, Falleni M, Pellegrini C, Catania A, Roncalli M, Marchetti A, Santambrogio L, Coggi G, Bosari S. Identification of potential therapeutic targets in malignant mesothelioma using cell-cycle gene expression analysis. American Journal of Pathology. 2009;174:762–770. doi: 10.2353/ajpath.2009.080721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Ryan PJ, Oates JL, Crocker J, Stableforth DE. Distinction between pleural mesothelioma and pulmonary adenocarcinoma using MOC31 in an asbestos sprayer. Respiratory Medicine. 1997;91:57–60. doi: 10.1016/s0954-6111(97)90137-2. [DOI] [PubMed] [Google Scholar]
  55. Sato A, Sekine M, Virgona N, Ota M, Yano T. Yes is a central mediator of cell growth in malignant mesothelioma cells. Oncology Reports. 2012;28:1889–1893. doi: 10.3892/or.2012.2010. [DOI] [PubMed] [Google Scholar]
  56. Sekido Y. Molecular pathogenesis of malignant mesothelioma. Carcinogenesis. 2013 doi: 10.1093/carcin/bgt166. [DOI] [PubMed] [Google Scholar]
  57. Shabo I, Svanvik J. Expression of macrophage antigens by tumor cells. Adv Exp Med Biol. 2011;714:141–150. doi: 10.1007/978-94-007-0782-5_7. [DOI] [PubMed] [Google Scholar]
  58. Shield PW, Callan JJ, Devine PL. Markers for metastatic adenocarcinoma in serous effusion specimens. Diagnostic Cytopathology. 1994;11:237–245. doi: 10.1002/dc.2840110309. [DOI] [PubMed] [Google Scholar]
  59. Swain WA, O’Byrne KJ, Faux SP. Activation of p38 MAP kinase by asbestos in rat mesothelial cells is mediated by oxidative stress. Am.J.Physiol Lung Cell Mol.Physiol. 2004;286:L859–L865. doi: 10.1152/ajplung.00162.2003. [DOI] [PubMed] [Google Scholar]
  60. Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140:805–820. doi: 10.1016/j.cell.2010.01.022. [DOI] [PubMed] [Google Scholar]
  61. Tang B, Vu M, Booker T, Santner SJ, Miller FR, Anver MR, Wakefield LM. TGF-beta switches from tumor suppressor to prometastatic factor in a model of breast cancer progression. J Clin Invest. 2003;112:1116–1124. doi: 10.1172/JCI18899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Testa JR, Cheung M, Pei J, Below JE, Tan Y, Sementino E, Cox NJ, Dogan AU, Pass HI, Trusa S, Hesdorffer M, Nasu M, Powers A, Rivera Z, Comertpay S, Tanji M, Gaudino G, Yang H, Carbone M. Germline BAP1 mutations predispose to malignant mesothelioma. Nat Genet. 43:1022–1025. doi: 10.1038/ng.912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Tomasetti M, Amati M, Santarelli L, Alleva R, Neuzil J. Malignant mesothelioma: biology, diagnosis and therapeutic approaches. Curr.Mol.Pharmacol. 2009;2:190–206. doi: 10.2174/1874467210902020190. [DOI] [PubMed] [Google Scholar]
  64. Traves PG, Luque A, Hortelano S. Macrophages, inflammation, and tumor suppressors: ARF, a new player in the game. Mediators Inflamm. 2012;2012:568783. doi: 10.1155/2012/568783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Tsujimura T, Torii I, Sato A, Song M, Fukuoka K, Hasegawa S, Nakano T. Pathological and molecular biological approaches to early mesothelioma. Int J Clin Oncol. 2012;17:40–47. doi: 10.1007/s10147-011-0369-1. [DOI] [PubMed] [Google Scholar]
  66. Turovskaya O, Foell D, Sinha P, Vogl T, Newlin R, Nayak J, Nguyen M, Olsson A, Nawroth PP, Bierhaus A, Varki N, Kronenberg M, Freeze HH, Srikrishna G. RAGE, carboxylated glycans and S100A8/A9 play essential roles in colitis-associated carcinogenesis. Carcinogenesis. 2008;29:2035–2043. doi: 10.1093/carcin/bgn188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Voelcker V, Gebhardt C, Averbeck M, Saalbach A, Wolf V, Weih F, Sleeman J, Anderegg U, Simon J. Hyaluronan fragments induce cytokine and metalloprotease upregulation in human melanoma cells in part by signalling via TLR4. Experimental Dermatology. 2008;17:100–107. doi: 10.1111/j.1600-0625.2007.00638.x. [DOI] [PubMed] [Google Scholar]
  68. Walker C, Everitt J, Barrett JC. Possible cellular and molecular mechanisms for asbestos carcinogenicity. American Journal of Industrial Medicine. 1992;21:253–273. doi: 10.1002/ajim.4700210214. [DOI] [PubMed] [Google Scholar]
  69. Whitaker D, Papadimitriou JM, Walters MN. The mesothelium: a histochemical study of resting mesothelial cells. Journal of Pathology. 1980;132:273–284. doi: 10.1002/path.1711320309. [DOI] [PubMed] [Google Scholar]
  70. Whitson BA, Kratzke RA. Molecular pathways in malignant pleural mesothelioma. Cancer Lett. 2006;239:183–189. doi: 10.1016/j.canlet.2005.08.010. [DOI] [PubMed] [Google Scholar]
  71. Wu J, Yang X, Zhang YF, Wang YN, Liu M, Dong XQ, Fan JJ, Yu XQ. Glucose-based peritoneal dialysis fluids downregulate toll-like receptors and trigger hyporesponsiveness to pathogen-associated molecular patterns in human peritoneal mesothelial cells. Clin Vaccine Immunol. 2010;17:757–763. doi: 10.1128/CVI.00453-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Xie W, Huang Y, Guo A, Wu W. Bacteria peptidoglycan promoted breast cancer cell invasiveness and adhesiveness by targeting toll-like receptor 2 in the cancer cells. PLoS One. 2010;5:e10850. doi: 10.1371/journal.pone.0010850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Xu J, Futakuchi M, Shimizu H, Alexander DB, Yanagihara K, Fukamachi K, Suzui M, Kanno J, Hirose A, Ogata A, Sakamoto Y, Nakae D, Omori T, Tsuda H. Multi-walled carbon nanotubes translocate into the pleural cavity and induce visceral mesothelial proliferation in rats. Cancer Sci. 2012;103:2045–2050. doi: 10.1111/cas.12005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Yang H, Testa JR, Carbone M. Mesothelioma epidemiology, carcinogenesis, and pathogenesis. Curr.Treat.Options.Oncol. 2008;9:147–157. doi: 10.1007/s11864-008-0067-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Yang L, Pang Y, Moses HL. TGF-beta and immune cells: an important regulatory axis in the tumor microenvironment and progression. Trends Immunol. 2010;31:220–227. doi: 10.1016/j.it.2010.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Yu L, Wang L, Chen S. Exogenous or endogenous Toll-like receptor ligands: which is the MVP in tumorigenesis? Cell Mol Life Sci. 2012;69:935–949. doi: 10.1007/s00018-011-0864-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Yung S, Chan TM. Mesothelial cells. Peritoneal Dialysis International. 2007;27(Suppl 2):S110–S115. S110–S115. [PubMed] [Google Scholar]
  78. Yung S, Chan TM. Pathophysiology of the peritoneal membrane during peritoneal dialysis: the role of hyaluronan. J Biomed Biotechnol. 2011;2011:180594. doi: 10.1155/2011/180594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Yunta M, Lazo PA. Apoptosis protection and survival signal by the CD53 tetraspanin antigen. Oncogene. 2003;22:1219–1224. doi: 10.1038/sj.onc.1206183. [DOI] [PubMed] [Google Scholar]
  80. Zhong J, Lardinois D, Szilard J, Tamm M, Roth M. Rat mesothelioma cell proliferation requires p38delta mitogen activated protein kinase and C/EBP-alpha. Lung Cancer. 2011;73:166–170. doi: 10.1016/j.lungcan.2010.12.003. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Sup1

RESOURCES