Peptidylarginine deiminase 2 (PAD2/PADI2) has been implicated in various inflammatory diseases and, more recently, cancer. The goal of this study was to test the hypothesis that PAD2 promotes oncogenesis using a transgenic mouse model. We found that about 37% of transgenic mice overexpressing human FLAG-PAD2 downstream of the MMTV-LTR promoter develop spontaneous neoplastic skin lesions. Molecular and histopathologic analyses of the resulting lesions find that they contain increased levels of markers for invasion, inflammation, and epithelial-to-mesenchymal transition (EMT) and that a subset of the lesions progress to invasive squamous cell carcinoma (SCC). We then stably overexpressed FLAG-PAD2 in the human SCC cell line, A431, and found that the PAD2-overexpressing cells were more tumorigenic in vitro and also contained elevated levels of markers for inflammation and EMT. Collectively, these studies provide the first genetic evidence that PAD2 functions as an oncogene and suggest that PAD2 may promote tumor progression by enhancing inflammation within the tumor microenvironment. Cancer Res; 74(21); 6306–17. ©2014 AACR.

The peptidylarginine deiminase (PADI or more commonly referred to as PAD) family of posttranslational modification enzymes converts positively charged arginine residues on substrate proteins to neutrally charged citrulline. This activity, alternatively called deimination or citrullination, has been shown to have wide-ranging effects on target protein structure, function, and protein–protein interactions. Increasingly, the dysregulation of PAD activity is associated with a range of diseases, including rheumatoid arthritis, multiple sclerosis, ulcerative colitis, neural degeneration, chronic obstructive pulmonary disorder (COPD), and cancer (1–3). While the presumptive function of PAD activity in most diseases is linked to inflammation, the role that PADs play in cancer progression is still under investigation (4–7). Our recent studies suggest a role for PAD2 in the oncogenic progression of breast cancer (8–11) and provide preclinical evidence showing that the PAD inhibitor, Cl-amidine, could be used as a therapeutic agent for the treatment of tumors in vivo (11). To further investigate the involvement of PAD2 in the oncogenesis of epithelial tumors, we generated FVB/N mice expressing FLAG-PAD2 under the control of the mammary tumor virus (MMTV) promoter. As with previous studies (12, 13), we found that the MMTV promoter drives transgene expression in a range of tissues, including the mammary gland, salivary gland, ovaries, and skin. Results from our spontaneous tumor study show that about 37% of the PAD2 transgenic mice developed skin lesions within a period of 4 to 12 months after birth. These tumors expressed high levels of transgenic human PAD2 and display markers of increased invasiveness and epithelial-to-mesenchymal transition (EMT). Furthermore, a subset of these tumors displays the hallmarks of malignant progression to highly invasive squamous cell carcinomas (SCC). The findings identify a novel genetic mouse model of skin neoplasia and define a role for PAD2 in cancer progression.

Generation of MMTV-FLAG-PAD2 mice

To generate the MMTV-FLAG-PAD2 construct, human PAD2 cDNA was subcloned from pcDNA3.1-FLAG-PAD2 (14) into the EcoRI sites of the MMTV-SV40-Bssk plasmid (Addgene plasmid #1824), originally generated in the laboratory of Dr. Philip Leder at Harvard Medical School (Boston, MA; ref. 15). The linear MMTV-FLAG-PAD2 construct was purified and microinjected into the pronuclei of fertilized embryos from superovulated FVB/N (FVB/NJ, JAX) mice, and 2-cell stage embryos were transferred to pseudopregnant mothers. The microinjection and embryo transfer were performed by the Stem Cell and Transgenics Core at Cornell University's College of Veterinary Medicine (Ithaca, NY). Mice were genotyped for the presence of an integrated human PAD2 transgene by PCR with the primers hPAD2-cds-F/R, and mouse Pad4 was used as genomic control (see Supplementary Table S1). All mouse experiments were reviewed and approved by the Institutional Animal Care and Use Committees (IACUC) at Cornell University.

Stable FLAG-PAD2 expression in A431 cells

The human SCC A431 cell line was obtained from ATCC (CRL-1555) in 2013 and cultured according to manufacturer's directions for 2 weeks before the generation of stable lines. Authentication was performed by ATCC using short tandem repeat (STR) DNA fingerprinting (16). To generate A431 cells overexpressing FLAG-tagged PAD2, two separate plasmids were generated by subcloning FLAG-PAD2 into the pIRES2-EGFP vector (Clontech) and pLenti-PGK-GFP-Puro plasmid (Addgene #19070), followed by transfection and transduction, respectively (see Supplementary Methods).

Immunohistochemistry and immunofluorescence

Immunohistochemistry (IHC) and immunofluorescence experiments were carried out using a standard protocol as previously described (9). Primary antibodies are as follows: anti-PAD2 (12110-1-AP, ProteinTech), anti-FLAG-M2 (F1804, Sigma), anti-pan-Citrulline (ab6464, Abcam), anti-Ki67 (ab15580, Abcam), and anti-IL6 (ab6672, Abcam). Negative controls were either normal rabbit or mouse IgG.

Western blotting

Western blotting was carried out as previously described (9). Primary antibodies against PAD2 (12110-1-AP, ProteinTech) and FLAG-M2 (F1804, Sigma) were incubated overnight at 4°C. To confirm equal protein loading, membranes were stripped and reprobed with anti-β-actin (ab8227, Abcam).

RNA isolation, semiquantitative, and quantitative real-time PCR

RNA was purified using the Qiagen RNeasy Kit, including on-column DNase treatment, and reverse-transcribed using the Applied Biosystems High Capacity RNA-to-cDNA kit according to the manufacturer's protocol. See Supplementary Methods for a more detailed description of semiquantitative and quantitative real-time PCR (qPCR) and Supplementary Tables S1–S3 for primer sequences.

Assay for cellular malignancy and invasion

Collagen-coated inserts for 24-well plate wells (Falcon BD Fluoroblok; #351152) were used to conduct Transwell migration assays as previously described (17) and further detailed in the Supplementary Methods. Focus formation assays were carried out as previously described (18).

Statistical analysis

All experiments were independently repeated at least three times unless otherwise indicated. Values were expressed as the mean ± SD and analyzed using Student t test or ANOVA (with Tukey post hoc analysis) where three or more groups were compared with one another using continuous data. Incidence of spontaneous lesions in MMTV-FLAG-PAD2 mice was measured 4 to 12 months post-birth, along with age-matched FVB/N mice from the corresponding founder line. Data were analyzed using a χ2 test with Yates correction for continuity. Significance was set at an α of 0.05.

Generation of MMTV-FLAG-PAD2 transgenic mice

To assess the potential role of PAD2 in the oncogenesis of epithelial tissue, we generated a mouse model in which the human PAD2 gene is overexpressed under the control of the hormone-responsive MMTV-LTR promoter. The transgenic construct consists of an MMTV-LTR promoter placed upstream of the human FLAG-tagged PAD2 cDNA, followed by an SV40 splice/polyadenylation site (Fig. 1A). MMTV-FLAG-PAD2 mice were generated, and seven potential founders were tested for the presence of transgene by PCR. Four founders (4807, 4853, 4680, and 4863) were identified (indicated in red, Fig. 1B) that carried germline transmission of the FLAG-PAD2 transgene. We note that 4807 was not included in subsequent analyses due to poor breeding. Using semiquantitative RT-PCR, we confirmed the presence of FLAG-PAD2 transcript in the skin, salivary gland, mammary gland, and ovary of the transgenic mice (Fig. 1C). These four tissues were chosen for analysis because they have previously been shown to express high levels of MMTV-LTR–driven transgenes (13). qPCR analysis of the same tissues demonstrated that while PAD2 expression was highest in salivary and mammary glands, significant levels of transgenic PAD2 were also found in the skin of all three founders (P < 0.01; Fig. 1D). We note here that while transgenic expression of PAD2 was observed in the mammary glands of both virgin (Supplementary Fig. S1A) and multiparous mice (Supplementary Fig. S1B), we did not detect any gross abnormalities or any observable phenotype in the mammary glands of these mice. The potential mechanisms that may be blocking PAD2-mediated oncogenesis in the mammary gland are currently being investigated.

Figure 1.

MMTV-FLAG-PAD2 transgenic construct. A, schematic of the linearized MMTV-FLAG-hPAD2 transgene, containing FLAG-tagged human PAD2 cDNA cloned between the EcoRI sites of the MMTV-SV40-Bssk plasmid (15). The construct used for generation of FLAG-hPAD2-transgenic mice (herein FLAG-PAD2) consists of FLAG-PAD2 under the control of the hormone-responsive MMTV-LTR promoter enhancer with an SV40 splice-polyadenylation signal (SV40pA). B, PCR screening of DNA extracted from mouse tails for the presence of integrated human PAD2 transgene (TG: hPAD2). Four founders were identified (red); primers for mouse Pad4 were used as a WT (mPad4) control for amplification. Primer details can be found in Supplementary Table S1. C, semiquantitative RT-PCR was performed on tissues known to have high expression in MMTV-LTR transgenic mice: skin, salivary gland (SG), mammary gland (MG), and ovary. Relative mRNA levels were measured for the transgenic human PAD2, along with endogenous mouse Pad2. Mouse Gapdh was used as the loading control. D, qPCR for the human PAD2 transgene was performed across the same tissues, using WT (FVB/N) skin as the reference, with mouse Gapdh normalization (*, P < 0.01).

Figure 1.

MMTV-FLAG-PAD2 transgenic construct. A, schematic of the linearized MMTV-FLAG-hPAD2 transgene, containing FLAG-tagged human PAD2 cDNA cloned between the EcoRI sites of the MMTV-SV40-Bssk plasmid (15). The construct used for generation of FLAG-hPAD2-transgenic mice (herein FLAG-PAD2) consists of FLAG-PAD2 under the control of the hormone-responsive MMTV-LTR promoter enhancer with an SV40 splice-polyadenylation signal (SV40pA). B, PCR screening of DNA extracted from mouse tails for the presence of integrated human PAD2 transgene (TG: hPAD2). Four founders were identified (red); primers for mouse Pad4 were used as a WT (mPad4) control for amplification. Primer details can be found in Supplementary Table S1. C, semiquantitative RT-PCR was performed on tissues known to have high expression in MMTV-LTR transgenic mice: skin, salivary gland (SG), mammary gland (MG), and ovary. Relative mRNA levels were measured for the transgenic human PAD2, along with endogenous mouse Pad2. Mouse Gapdh was used as the loading control. D, qPCR for the human PAD2 transgene was performed across the same tissues, using WT (FVB/N) skin as the reference, with mouse Gapdh normalization (*, P < 0.01).

Close modal

PAD2 transgenic mice develop skin lesions with the potential to advance to invasive SCCs

Analysis of spontaneous neoplastic growth in experimental mice found that about 37% of the mice across all founder lines developed skin lesions between 4 and 12 months of age (Table 1). When compared with age-matched wild-type (WT) FVB/N mice, the increased rate of neoplastic growth in FLAG-PAD2 transgenic mice was highly significant (χ2 = 30.65; P < 0.001), suggesting that the PAD2 transgene is likely promoting oncogenesis. The percentage of males developing tumors was about 44%, whereas females developed tumors at a rate of about 56%, which was not significantly different across genders. These lesions occurred on both the dorsum and ventrum of transgenic mice and are characterized by gross abnormalities, such as alopecia, multifocal epidermal ulceration often covered in serocellular crust, dysplasia, and thickening of the adjacent epidermis (Fig. 2A).

Figure 2.

Transgenic FLAG-PAD2 expression in the epidermis of mice leads to the development of skin lesions. A, representative gross lesions showing the skin phenotype, which includes alopecia, multifocal epidermal ulceration often covered in a serocellular crust, and thickening of the adjacent epidermis. B, histologic evaluation of the skin sections by hematoxylin and eosin (H&E) reveals features consistent with invasive SCC. i, nests of neoplastic cells arising from the epidermis are shown invading the deep dermis and subcutis, with the epidermis overlying the neoplasm extensively ulcerated. i and ii, adjacent epidermal layers are hyperplastic, with the loss of adnexal structures and increased desmoplastic response. iii, tumor borders are poorly defined, with highly anaplastic tumor cells invading the deeper dermis and subcutis, often infiltrating and separating the skeletal myofibers. iv, carcinoma cells are frequently found budding off from the primary neoplasm. C, representative IHC analysis of FLAG-PAD2 protein expression in skin neoplasms from transgenic mice. i, lower magnification (×4) image showing PAD2 protein expression in the hyperplastic epidermis, neoplastic islands, and hair follicular epithelium. ii and iii, PAD2 protein expression in neoplastic cells surrounding concentric layers of keratin (arrow, keratin pearl). iv, representative image showing high levels of PAD2 protein expression in budding nest of carcinoma cells (broken arrow). D, comparison of citrulline levels in WT skin epithelium and lesion from FLAG-PAD2 transgenic mouse (TG). Rabbit IgG served as negative control. Magnification for images: B, i (×4), ii (×10), iii (×20), and iv (×40); C, i (×4), ii (×20), iii (×20), and iv (×40); D, all ×5 (inset, ×40).

Figure 2.

Transgenic FLAG-PAD2 expression in the epidermis of mice leads to the development of skin lesions. A, representative gross lesions showing the skin phenotype, which includes alopecia, multifocal epidermal ulceration often covered in a serocellular crust, and thickening of the adjacent epidermis. B, histologic evaluation of the skin sections by hematoxylin and eosin (H&E) reveals features consistent with invasive SCC. i, nests of neoplastic cells arising from the epidermis are shown invading the deep dermis and subcutis, with the epidermis overlying the neoplasm extensively ulcerated. i and ii, adjacent epidermal layers are hyperplastic, with the loss of adnexal structures and increased desmoplastic response. iii, tumor borders are poorly defined, with highly anaplastic tumor cells invading the deeper dermis and subcutis, often infiltrating and separating the skeletal myofibers. iv, carcinoma cells are frequently found budding off from the primary neoplasm. C, representative IHC analysis of FLAG-PAD2 protein expression in skin neoplasms from transgenic mice. i, lower magnification (×4) image showing PAD2 protein expression in the hyperplastic epidermis, neoplastic islands, and hair follicular epithelium. ii and iii, PAD2 protein expression in neoplastic cells surrounding concentric layers of keratin (arrow, keratin pearl). iv, representative image showing high levels of PAD2 protein expression in budding nest of carcinoma cells (broken arrow). D, comparison of citrulline levels in WT skin epithelium and lesion from FLAG-PAD2 transgenic mouse (TG). Rabbit IgG served as negative control. Magnification for images: B, i (×4), ii (×10), iii (×20), and iv (×40); C, i (×4), ii (×20), iii (×20), and iv (×40); D, all ×5 (inset, ×40).

Close modal
Table 1.

Incidence of spontaneous skin lesions in MMTV-FLAG-PAD2 mice

MMTV-FLAG-PAD2WT FVB/Nb
Founder lineLesions/miceIncidence (%)Male/female (%)Lesions/miceIncidence (%)Male/female (%)
4853 4/20 20.0 25/75 1/18 5.6 0/100 
4860 20/46 43.5 55/45 2/43 4.7 50/50 
4863 9/24 37.5 33/67 0/33 0.0 0/0 
Total 33/90a 36.7c 45/55 3/94a 3.2 44/56 
MMTV-FLAG-PAD2WT FVB/Nb
Founder lineLesions/miceIncidence (%)Male/female (%)Lesions/miceIncidence (%)Male/female (%)
4853 4/20 20.0 25/75 1/18 5.6 0/100 
4860 20/46 43.5 55/45 2/43 4.7 50/50 
4863 9/24 37.5 33/67 0/33 0.0 0/0 
Total 33/90a 36.7c 45/55 3/94a 3.2 44/56 

NOTE: Transgenic (MMTV-FLAG-PAD2) and age-matched WT FVB/N mice were observed over a period of 4 to 12 months post-birth, and spontaneous lesions were characterized by gross abnormalities, such as alopecia, multifocal epidermal ulceration (covered in serocellular crust), dysplasia, and thickening of the adjacent epidermis. Data were analyzed using a χ2 test with Yates correction for continuity), χ2 = 30.65, and P ≤ 0.001.

aMice were observed over 1 year, with lesions appearing spontaneously between 4 and 12 months.

bWT FVB/N mice are from the corresponding founder lines and age-matched to transgenic mice.

cχ2 analysis (with Yates correction), P ≤ 0.001.

Histologic evaluation of skin lesions from PAD2-overexpressing mice by hematoxylin and eosin reveals that these sites contain highly neoplastic tissue, having features consistent with invasive SCC. We found nests of neoplastic cells that appeared to originate from the epidermis and to invade into the dermis and subcutis (Fig. 2B, i and ii); moreover, the epidermis overlying the neoplasm was extensively ulcerated and frequently adjacent to hyperplastic epidermal layers (Fig. 2B, i). In addition, we observed that concentric layers of keratin surrounded tumor cells forming keratin pearls, which are characteristic of SCC (Fig. 2B, ii). We found that tumor borders were poorly defined, often with highly anaplastic tumor cells invading the deeper dermis and subcutis, and that neoplastic cells frequently infiltrated and separated skeletal myofibers (Fig. 2B, iii). The nests and islands of neoplastic cells within the subcutis showed a high degree of anisokaryosis and anisocytosis, with the nuclei often containing one to two prominent nucleoli (Fig. 2B, iv). Clusters of neoplastic cells were found to be surrounded by loose collagenous stroma with a marked loss of adnexal structures, consistent with desmoplastic response (Fig. 2B, ii–iv). Finally, we observed that these lesions often contained carcinoma cells budding from the primary neoplasm (Fig. 2B, iv), again indicating an invasive component to these tumors.

Recombinant PAD2 is expressed in the skin and tumor tissue of PAD2 transgenic mice

To test whether the observed skin neoplasia in the transgenic mice was driven by overexpression of PAD2, we next investigated PAD2 expression levels in these mice by IHC. We probed these tissues with an anti-human PAD2 antibody and found that PAD2 protein was observed in the hyperplastic epidermis, neoplastic islands, and in the hair follicular epithelium (Fig. 2C, i). In addition, we observed high PAD2 expression in the neoplastic epithelium surrounding keratin pearls (Fig. 2C, ii and iii). PAD2 was also expressed in the invasive, budding nests of carcinoma cells, which appeared to invade stromal tissue adjacent to the primary neoplasm (Fig. 2C, iv). To test whether PAD2 overexpression correlated with increased deimination activity, we next probed sections from WT skin and transgenic PAD2 lesions with a pan-citrulline antibody that has been previously shown to specifically detect citrullinated proteins (19, 20). Results show that the skin lesions from MMTV-FLAG-PAD2 mice appear to have increased pan-citrulline staining when compared with WT skin (Fig. 2D), with the most intense levels of citrullination appearing to occur in the hyperplastic regions. In addition, we found strong nuclear anti-pan-citrulline staining in these lesions (Fig. 2D, inset bottom image), which is in line with our previous studies showing that PAD2-mediated histone deimination regulates gene expression (9, 10, 14, 21). Because the anti-human PAD2 antibody likely cross-reacts with endogenous mouse PAD2, we also stained these tissues with an anti-FLAG antibody to specifically stain the PAD2 fusion protein. We observed a similar expression pattern within the proliferating basal layers of hyperplastic/neoplastic epidermis, suggesting that much of the PAD2 signal originates from overexpression of the transgene (data not shown). We confirmed this prediction using indirect immunofluorescence to co-stain for FLAG and PAD2 expression in the skin lesions of transgenic mice (Fig. 3). Both PAD2 and FLAG were seen to colocalize to the neoplastic epithelial cells (Fig. 3A, i–iv). While PAD2 shows strong staining in multiple layers of the epidermis, we found that FLAG-PAD2 expression is slightly more restricted to the basal layer, which is known to be more stem cell–like and highly proliferative (22). Interestingly, we also see an apparent slight increase in the proliferative marker, Ki67, within the FLAG staining section of the SCC lesion (Fig. 3B, ii–iv). This observation supports the hypothesis that PAD2 overexpression in the skin promotes epidermal proliferation, which is further supported by the identification of a subset of lesions that are highly proliferative and characteristic of invasive SCC. We also note that control staining with rabbit IgG was negative (Supplementary Fig. S2) and that the anti-FLAG antibody did not stain epidermal tissue from WT mice (Fig. 3C, i–vi) or adjacent normal skin from transgenic mice (data not shown).

Figure 3.

Confocal immunofluoresence analysis of PAD2, FLAG, and Ki67 expression in neoplastic skin lesions of the FLAG-PAD2 transgenic mouse. A, representative image showing co-staining (iv) of FLAG (red, ii) and PAD2 (green, iii) in the neoplastic epithelial cells of an SCC from a transgenic mouse. Nuclei were stained with 4′,6-diamidino-2-phenylindole (DAPI; blue, i). Endogenous PAD2 shows strong positive staining in two to three layers of epidermal cells, whereas FLAG-PAD2 is predominantly localized to the basal cell layer. Scattered tumor cell islands underlying the epidermis show high levels of PAD2 expression. B, representative image showing co-staining (iv) of the proliferative marker FLAG (red, ii) and Ki67 (green, iii) in neoplastic epithelial cells of SCC. Tumor islands that are strongly positive for FLAG-PAD2 expression levels contain an increased number of Ki67-positive cells (iv). Nuclei were stained with DAPI (blue, i). Normal skin from WT mice (FVB/N) is absent of transgene expression and has low levels of the proliferation marker, Ki67. C, representative image showing co-staining (vi) of FLAG (red, iii) and Ki67 (green, iv) in normal skin sections from a WT mouse. Bright-field image (ii and v). Both FLAG and PAD2 protein expression are absent, concomitant with reduced Ki67 levels. Nuclei were stained with DAPI (blue, i). Magnification in images A–C, ×40. Scale bar, 50 μm.

Figure 3.

Confocal immunofluoresence analysis of PAD2, FLAG, and Ki67 expression in neoplastic skin lesions of the FLAG-PAD2 transgenic mouse. A, representative image showing co-staining (iv) of FLAG (red, ii) and PAD2 (green, iii) in the neoplastic epithelial cells of an SCC from a transgenic mouse. Nuclei were stained with 4′,6-diamidino-2-phenylindole (DAPI; blue, i). Endogenous PAD2 shows strong positive staining in two to three layers of epidermal cells, whereas FLAG-PAD2 is predominantly localized to the basal cell layer. Scattered tumor cell islands underlying the epidermis show high levels of PAD2 expression. B, representative image showing co-staining (iv) of the proliferative marker FLAG (red, ii) and Ki67 (green, iii) in neoplastic epithelial cells of SCC. Tumor islands that are strongly positive for FLAG-PAD2 expression levels contain an increased number of Ki67-positive cells (iv). Nuclei were stained with DAPI (blue, i). Normal skin from WT mice (FVB/N) is absent of transgene expression and has low levels of the proliferation marker, Ki67. C, representative image showing co-staining (vi) of FLAG (red, iii) and Ki67 (green, iv) in normal skin sections from a WT mouse. Bright-field image (ii and v). Both FLAG and PAD2 protein expression are absent, concomitant with reduced Ki67 levels. Nuclei were stained with DAPI (blue, i). Magnification in images A–C, ×40. Scale bar, 50 μm.

Close modal

Enhanced inflammation marker expression is observed in the skin of transgenic mice when compared with WT skin and with the mammary gland of transgenic animals

Previous reports have implicated PAD2 in a wide range of inflammation-associated diseases (23–25), including our recent work where we reported that PAD2 appears to promote cancer progression in vitro and in vivo using a xenograft model of breast cancer (11). Therefore, in an effort to identify potential pathways involved in the progression of MMTV-FLAG-PAD2 skin lesions, we next investigated the expression of a subset of inflammation and invasion-associated genes in the transgenic PAD2 lesions. Interestingly, the lesions containing high levels of transgenic PAD2 (Fig. 4A–C, lesions 2 and 4) also expressed high levels of Ki67, Il6, and the functional murine orthologs for human IL8; Cxcl1 (KC; ref. 26), Cxcl2 (MIP-2; ref. 27), and Cxcl5 (LIX; ref. 28; P < 0.001; Fig. 4D). These lesions also expressed high levels of the inflammatory mediator, Cox2 (Fig. 4E), which has previously been shown to induce Il6 expression (29). In addition, these tumors also displayed elevated levels of the EMT markers, vimentin (Vim) and the E-cadherin repressor, Snail (Snai1, or more commonly referred to as Snail), along with decreased levels of E-cadherin (Cdh1, or hereafter Ecad; P < 0.01) and Gsk3β (Fig. 4E), which has been previously shown to be downregulated in SCCs of the tongue (30) and mouse models of skin tumorigenesis (31).

Figure 4.

Transgene expression in the skin lesions of MMTV-FLAG-PAD2 mice. A, semiquantitative RT-PCR of four representative skin lesions from transgenic mice (lesions 1–4). Relative mRNA levels were measured for the transgenic human PAD2, along with endogenous mouse Pad2. Mouse Gapdh was used as the loading control. B, qPCR (TaqMan) for the human PAD2 transgene was performed across the same lesions, using normal WT skin as the reference, along with mouse Gapdh normalization (*, P < 0.01). C, Western blot analysis of skin from WT mice, along with transgenic lesions (2 and 4). Whole-cell lysates were probed for FLAG expression, with a predicted protein size of about 75 kDa. β-Actin was used as a loading control. D and E, skin lesions from MMTV-FLAG-PAD2 transgenic mice express markers of inflammation (D; *, P < 0.001) and EMT (E; *, P < 0.01). Relative mRNA levels for the representative genes (see Supplementary Table S2) were determined by qPCR (SYBR) using normal WT skin as the reference, along with β-actin (Actb) normalization. F, representative IHC image of anti-Il6 staining in WT skin (bottom, left), FLAG-PAD2 transgenic (TG) mammary gland (bottom, middle), and TG skin lesion (bottom, right). Rabbit IgG served as negative control. Magnification, ×40. Scale bar, 20 μm.

Figure 4.

Transgene expression in the skin lesions of MMTV-FLAG-PAD2 mice. A, semiquantitative RT-PCR of four representative skin lesions from transgenic mice (lesions 1–4). Relative mRNA levels were measured for the transgenic human PAD2, along with endogenous mouse Pad2. Mouse Gapdh was used as the loading control. B, qPCR (TaqMan) for the human PAD2 transgene was performed across the same lesions, using normal WT skin as the reference, along with mouse Gapdh normalization (*, P < 0.01). C, Western blot analysis of skin from WT mice, along with transgenic lesions (2 and 4). Whole-cell lysates were probed for FLAG expression, with a predicted protein size of about 75 kDa. β-Actin was used as a loading control. D and E, skin lesions from MMTV-FLAG-PAD2 transgenic mice express markers of inflammation (D; *, P < 0.001) and EMT (E; *, P < 0.01). Relative mRNA levels for the representative genes (see Supplementary Table S2) were determined by qPCR (SYBR) using normal WT skin as the reference, along with β-actin (Actb) normalization. F, representative IHC image of anti-Il6 staining in WT skin (bottom, left), FLAG-PAD2 transgenic (TG) mammary gland (bottom, middle), and TG skin lesion (bottom, right). Rabbit IgG served as negative control. Magnification, ×40. Scale bar, 20 μm.

Close modal

Our finding that PAD2 overexpression in the skin, but not the mammary gland, leads to neoplastic growth was somewhat surprising, and we speculated that the lack of tumor growth in the mammary gland may have been due to a reduced ability of FLAG-PAD2 to promote inflammatory and/or EMT marker expression in this tissue. To test this hypothesis, we next examined inflammatory/EMT marker expression in skin and mammary gland from WT and transgenic mice. Results show that, in large part, while PAD2 overexpression enhanced inflammatory marker expression in the skin, similar levels of induction were not observed in the mammary gland (Supplementary Fig. S3). These observations supported the hypothesis that PAD2 overexpression selectively promotes inflammatory marker expression and tumorigenesis in the skin but not the mammary gland. To test this hypothesis, we probed tissue sections with a well-validated anti-IL6 antibody. Results show that protein levels of Il6 appeared to be higher in the transgenic skin lesions than in the transgenic mammary gland and in WT skin (Fig. 4F). Taken together, these findings suggest that, in the skin, PAD2 overexpression may enhance tumor progression by selectively promoting inflammation and by altering the invasive properties of epithelial cells. We also evaluated the effect of PAD2 overexpression on inflammatory and EMT marker expression in the salivary gland and found that EMT marker expression appeared to be dampened, whereas inflammatory marker gene expression was not noticeably affected when compared with skin (Supplementary Fig. S3). Interestingly, there does not appear to be a direct PAD2 gene dosing effect with respect to inflammatory mediator/EMT expression in the different tumors and we predict that this discrepancy may be due to opposing and/or synergistic interactions between PAD2 and other PAD family members within individual tumors. In support of this prediction, we found that expression levels of PAD1–4 varied dramatically between tumors (Supplementary Fig. S4B and S4C). While speculative, it is possible that the ratio of PAD isozyme expression levels could modulate inflammatory/EMT expression levels and tumor growth and/or invasiveness to varying degrees.

Overexpression of PAD2 in human SCC A431 cells increases invasiveness and malignancy

Outcomes from our transgenic MMTV-FLAG-PAD2 mouse study show that ectopic expression of PAD2 is sufficient to drive tumorigenesis in epithelial cells and that this expression correlates with an increase in the expression of markers of inflammation and invasion. To test whether PAD2 might play a similar role in humans, we next investigated whether PAD2 overexpression in the human skin cancer cell line, A431, might lead to a similar phenotype. This cell line functions as a model system for human SCC and was derived from a vulvular epidermoid carcinoma (32). To investigate the oncogenic potential of PAD2 in these cells, we first transiently expressed FLAG-PAD2 using mammalian vectors (pcDNA3.1 and pIRES2) and found that these cell lines expressed high levels of the FLAG-PAD2 protein (Fig. 5A and B). In addition, we found that PAD2 overexpression suppressed E-cadherin and increased vimentin protein expression when compared with A431 cells that had been transfected with an empty vector (Fig. 5A). We next tested whether PAD2 overexpression altered the expression of inflammation and EMT markers and found that, similar to what was seen in transgenic mice, IL6 and IL8 gene levels were significantly increased in the PAD2-overexpressing cells, along with the inflammatory mediators COX2 and NFκB/RELA. Moreover, in addition to increasing the proliferation marker Ki67, we found that transient PAD2 overexpression also increased SNAIL and SLUG levels while suppressing E-cadherin levels (P < 0.05; Fig. 5C). Finally, we tested the effects of PAD2 overexpression on the expression of two proliferative oncogenes H-Ras (33, 34) and c-Myc (35), which are known to be upregulated in skin carcinomas. Results show that levels of these oncogenes were unchanged in the PAD2-overexpressing cells, suggesting that the observed effects of PAD2 on oncogenesis are more direct in nature.

Figure 5.

Transient overexpression of FLAG-PAD2 increases markers of inflammation and EMT in the human SCC A431 cell line. A, human SCC cells (A431) were transiently transfected with FLAG-PAD2 (pcDNA3.1-FP2 or pIRES2-FP2) or empty vector (pcDNA3.1-empty). Western blot analysis of protein expression for EMT markers, E-cadherin and vimentin, along with PAD2, was performed on transfected A431 cells. β-Actin was used as a loading control. B, PAD2 levels (*, P < 0.001) for the FLAG-PAD2–transfected A431 cells, relative to empty vector control (ACTB normalized), were analyzed by qPCR (SYBR). C, additional genes known to be involved in EMT and inflammation were also analyzed (*, P < 0.05). See Supplementary Table S3 for primer sequences.

Figure 5.

Transient overexpression of FLAG-PAD2 increases markers of inflammation and EMT in the human SCC A431 cell line. A, human SCC cells (A431) were transiently transfected with FLAG-PAD2 (pcDNA3.1-FP2 or pIRES2-FP2) or empty vector (pcDNA3.1-empty). Western blot analysis of protein expression for EMT markers, E-cadherin and vimentin, along with PAD2, was performed on transfected A431 cells. β-Actin was used as a loading control. B, PAD2 levels (*, P < 0.001) for the FLAG-PAD2–transfected A431 cells, relative to empty vector control (ACTB normalized), were analyzed by qPCR (SYBR). C, additional genes known to be involved in EMT and inflammation were also analyzed (*, P < 0.05). See Supplementary Table S3 for primer sequences.

Close modal

Given the data from our transiently transfected A431 cells, along with our data from the transgenic mice, we decided to investigate whether stable overexpression of FLAG-PAD2 might have an effect on the cellular malignancy and/or invasiveness of A431 cells. Using both lentiviral transduction of FLAG-PAD2 and traditional transfection, we created two stable PAD2-overexpressing lines: (i) pLenti-FLAG-PAD2 (pLenti-FP2), which expresses low levels of FLAG-PAD2 and (ii) pIRES2-FLAG-PAD2 (pIRES-FP2), which expresses high levels of FLAG-PAD2 (Fig. 6A and B). Next, we tested the invasive properties of these cell lines by measuring their ability to migrate through a collagen matrix. For both cell lines, we see a significant increase in cellular migration after 24 hours (P < 0.01), with the pIRES2-FP2 cell line also showing a significant increase in migration after only 4 hours (P < 0.01; Fig. 6C). These results suggest that PAD2 dosage might correlate with invasion. Finally, we wanted to examine the stable A431-pIRES2-FP2 cells for any increase in cellular malignancy. Assaying for focus formation, we show a significant increase in the FLAG-PAD2–overexpressing A431 cells compared with the empty vector control (P < 0.001; Fig. 6D). In addition, we find that the morphology of these cells also displays an elongated, fibroblast-like shape, indicative of cells that have undergone EMT (Fig. 6E).

Figure 6.

A431 cells stably overexpressing PAD2 are more tumorigenic in vitro. A431 cells were stably transfected with pIRES2-FLAG-PAD2 (pIRES2-FP2) or infected with lentivirus expressing FLAG-PAD2-EGFP (pLenti-FP2). Control vectors only express the EGFP gene (pIRES2-GFP and pLenti-GFP). A, Western blot analysis of PAD2 protein expression levels. β-Actin was used as a loading control. B, PAD2 expression in FLAG-PAD2–transfected A431 cells was analyzed by qPCR (SYBR). GFP-only vector was used as a control (ACTB normalized). Expression levels were analyzed using the 2 −ΔΔC(t) method, and data expressed as the mean ± SD from three independent experiments (*, P < 0.001). C, Transwell migration assay through a collagen matrix—cells were seeded and allowed to migrate through a collagen matrix. After 4 or 24 hours, migrating cells were counted under a microscope at ×40 magnification (*, P < 0.01). D and E, A431 skin cancer cells overexpressing FLAG-PAD2 show increased malignancy and EMT morphology. D, focus formation assay. Cells stably overexpressing FLAG-PAD2 (A431-FP2) or EGFP control (A431-GFP) were grown for 4 days in a 6-well plate, fixed with 4% paraformaldehyde, and stained with crystal violet for subsequent focus formation analysis. Absorbance levels were measured at 600 nm (*, P < 0.001). E, representative morphology of A431 cells overexpressing FLAG-PAD2.

Figure 6.

A431 cells stably overexpressing PAD2 are more tumorigenic in vitro. A431 cells were stably transfected with pIRES2-FLAG-PAD2 (pIRES2-FP2) or infected with lentivirus expressing FLAG-PAD2-EGFP (pLenti-FP2). Control vectors only express the EGFP gene (pIRES2-GFP and pLenti-GFP). A, Western blot analysis of PAD2 protein expression levels. β-Actin was used as a loading control. B, PAD2 expression in FLAG-PAD2–transfected A431 cells was analyzed by qPCR (SYBR). GFP-only vector was used as a control (ACTB normalized). Expression levels were analyzed using the 2 −ΔΔC(t) method, and data expressed as the mean ± SD from three independent experiments (*, P < 0.001). C, Transwell migration assay through a collagen matrix—cells were seeded and allowed to migrate through a collagen matrix. After 4 or 24 hours, migrating cells were counted under a microscope at ×40 magnification (*, P < 0.01). D and E, A431 skin cancer cells overexpressing FLAG-PAD2 show increased malignancy and EMT morphology. D, focus formation assay. Cells stably overexpressing FLAG-PAD2 (A431-FP2) or EGFP control (A431-GFP) were grown for 4 days in a 6-well plate, fixed with 4% paraformaldehyde, and stained with crystal violet for subsequent focus formation analysis. Absorbance levels were measured at 600 nm (*, P < 0.001). E, representative morphology of A431 cells overexpressing FLAG-PAD2.

Close modal

In the present study, we demonstrate that transgenic overexpression of PAD2 in the epidermis promotes carcinoma formation in the skin. In addition, we found that these lesions have the potential to advance to invasive SCC. These in vivo findings were supported by our in vitro studies, which showed that stable overexpression of PAD2 in the human SCC line, A431, resulted in increased cellular invasiveness and malignancy. At a more mechanistic level, we also demonstrate that PAD2 may promote carcinogenesis by enhancing the production of factors that promote inflammation and an EMT phenotype.

A number of mouse models, including models of advanced SCC, have documented a role for inflammation in cancer progression (36). In humans, the links between inflammation and cancer are now so strong that inflammation is considered to be the seventh hallmark of cancer (36, 37). Two key mediators of inflammation-driven cancers are the cytokines, IL6 and IL8, which promote cancer progression by stimulating tumor growth and angiogenesis (38–48). Interestingly, a number of previous studies have documented a role for PAD2-mediated deimination in regulating inflammatory activities during disease progression (23, 24). In addition, PAD activity has also been found to regulate tissue inflammation via direct modification of IL8 (25). Given these observations, we hypothesized that PAD2 may promote tumorigenesis by regulating the expression of IL6 and IL8. We tested this hypothesis and found that Il6 and Il8 ortholog expression is, in fact, increased in the skin lesions of PAD2-overexpressing mice and that IL6 and IL8 expression is elevated in PAD2-overexpressing cells.

In this study, we also show that PAD2 overexpression in transgenic mice, in addition to human cell lines, increased markers of invasiveness and EMT. An early critical molecular feature of EMT is the downregulation of E-cadherin (Cdh1), a cell adhesion molecule present in the plasma membrane of most normal epithelial cells. As cells progresses from an epithelial-like to mesenchymal-like state, E-cadherin downregulation is accompanied by the increased expression of Snail (Snai1) and the intermediate filament vimentin (Vim) (reviewed by Kaluri and colleagues; ref. 49). In our study, we found that PAD2 overexpression in mice induced an EMT-like phenotype in the resulting tumor cells as evidenced by the reduction in E-cadherin expression, along with an increase in Snail and vimentin expression. This molecular characterization of an EMT transition matches well with the invasive histology of the PAD2-overexpressing lesions.

A number of previous studies have shown that inflammatory cytokines, such as IL8, appear to promote cancer progression by inducing an EMT in tumor cells (41). Thus, it is possible that PAD2 overexpression promotes cell invasiveness and an EMT phenotype by upregulating cytokine production in tumor cells. Interestingly, however, we recently reported that another PAD, PAD4, plays an important role in regulating an EMT in breast cancer cells via regulation of GSK3β–TGFβ signaling (50). In that study, we found that PAD4 targets GSK3β for citrullination and that PAD4 depletion dramatically reduces levels of nuclear GSK3β, promotes EMT, and increases tumor invasiveness. In the current study, we found that overexpression of PAD2 suppressed Gsk3β expression, promoted an EMT phenotype and tumor growth. Together, these results raise the possibility that PAD2 and PAD4 may oppose or counterbalance each other in the maintenance of epithelial identity and in oncogenesis.

A surprising outcome of our project was that neoplastic growth appeared to be limited to the skin of PAD2 transgenic mice and was not observed in the mammary glands. While the ultimate explanation for this outcome is likely manifold, we found that the PAD2-mediated induction of inflammatory mediator expression was suppressed in the mammary glands of transgenic mice when compared with both the normal skin and the hyperplastic lesions from transgenic mice. These results suggest that, as opposed to the skin, a dampening mechanism may exist within the mammary gland that prevents FLAG-PAD2 from inducing inflammatory cytokine expression, thereby suppressing tumorigenesis. A potential explanation for this dampening effect may be derived from our finding that FLAG-PAD2 overexpression in vivo appears to significantly modulate the expression of other PAD family members in both normal and tumor tissue (Supplementary Fig. S4). Therefore, while purely speculative, it is possible that the relatively low levels of FLAG-PAD2 that we observed in the skin promotes the expression of a specific ratio of PAD family members that is conducive to inflammation and tumorigenesis, whereas the relatively higher level of FLAG-PAD2 in the mammary gland alters this ratio, leading to reduced inflammatory mediator expression and an absence of neoplastic growth. Regarding the potential mechanisms by which PAD2 overexpression regulates gene activity, we have recently found that PAD2 functions as a transcription factor co-factor and modulates target gene expression via histone deimination at transcription factor–binding sites (9, 10, 14, 21). Therefore, we are currently testing the hypothesis that PAD2 regulates the expression of target genes via similar mechanisms.

In conclusion, this study demonstrates that overexpression of human PAD2 in mice and in human cell lines promotes carcinogenesis. Furthermore, we show that a subset of tumors in mice display hallmarks of malignant progression from skin lesions to highly invasive SCCs. In addition, we demonstrate that PAD2 overexpression in vivo and in vitro enhances the expression of inflammatory cytokines and mediators of the EMT. Collectively, these studies provide functional and mechanistic evidence establishing PAD2 as a potential novel oncogene in the initiation and progression of epidermal carcinomas.

No potential conflicts of interest were disclosed.

Conception and design: J.L. McElwee, S. Mohanan, S. Horibata, S.A. Coonrod

Development of methodology: J.L. McElwee, S. Mohanan, S. Horibata, L.J. Anguish, S.A. Coonrod

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): J.L. McElwee, S. Mohanan, S. Horibata, K.L. Sams, L.J. Anguish, D. McLean, S.A. Coonrod

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): J.L. McElwee, S. Mohanan, S. Horibata, L.J. Anguish, D. McLean, J.J. Wakshlag, S.A. Coonrod

Writing, review, and/or revision of the manuscript: J.L. McElwee, S. Mohanan, J.J. Wakshlag, S.A. Coonrod

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): S. Horibata, K.L. Sams

Study supervision: J.L. McElwee

Other (technical and veterinary assistance in vivo and in vitro experiments): I. Cvitaš

This work was supported in part by funding through the DOD Era of Hope Award W871XWH-07-1-0372 to S.A. Coonrod and through an NIH Graduate Fellowship (grant T32HD057854) to J.L. McElwee.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Chumanevich
AA
,
Causey
CP
,
Knuckley
BA
,
Jones
JE
,
Poudyal
D
,
Chumanevich
AP
, et al
Suppression of colitis in mice by Cl-amidine: a novel peptidylarginine deiminase inhibitor
.
Am J Physiol Gastrointest Liver Physiol
2011
;
300
:
G929
38
.
2.
Lange
S
,
Gögel
S
,
Leung
K-Y
,
Vernay
B
,
Nicholas
AP
,
Causey
CP
, et al
Protein deiminases: new players in the developmentally regulated loss of neural regenerative ability
.
Dev Biol
2011
;
355
:
205
14
.
3.
Willis
VC
,
Gizinski
AM
,
Banda
NK
,
Causey
CP
,
Knuckley
B
,
Cordova
KN
, et al
N-alpha-benzoyl-N5-(2-chloro-1-iminoethyl)-L-ornithine amide, a protein arginine deiminase inhibitor, reduces the severity of murine collagen-induced arthritis
.
J Immunol
2011
;
186
:
4396
404
.
4.
Yao
H
,
Li
P
,
Venters
BJ
,
Zheng
S
,
Thompson
PR
,
Pugh
BF
, et al
Histone Arg modifications and p53 regulate the expression of OKL38, a mediator of apoptosis
.
J Biol Chem
2008
;
283
:
20060
8
.
5.
Li
P
,
Wang
D
,
Yao
H
,
Doret
P
,
Hao
G
,
Shen
Q
, et al
Coordination of PAD4 and HDAC2 in the regulation of p53-target gene expression
.
Oncogene
2010
;
29
:
3153
62
.
6.
Tanikawa
C
,
Ueda
K
,
Nakagawa
H
,
Yoshida
N
,
Nakamura
Y
,
Matsuda
K
. 
Regulation of protein Citrullination through p53/PADI4 network in DNA damage response
.
Cancer Res
2009
;
69
:
8761
9
.
7.
Mohanan
S
,
Cherrington
BD
,
Horibata
S
,
McElwee
JL
,
Thompson
PR
,
Coonrod
SA
. 
Potential role of peptidylarginine deiminase enzymes and protein citrullination in cancer pathogenesis
.
Biochem Res Int
2012
;
2012
:
895343
.
8.
Cherrington
BD
,
Mohanan
S
,
Diep
AN
,
Fleiss
R
,
Sudilovsky
D
,
Anguish
LJ
, et al
Comparative analysis of peptidylarginine deiminase-2 expression in canine, feline and human mammary tumours
.
J Comp Pathol
2012
;
147
:
139
46
.
9.
Cherrington
BD
,
Morency
E
,
Struble
AM
,
Coonrod SA
,
Wakshlag
JJ
. 
Potential role for peptidylarginine deiminase 2 (PAD2) in citrullination of canine mammary epithelial cell histones
.
PLoS One
2010
;
5
:
e11768
.
10.
Cherrington
BD
,
Zhang
X
,
McElwee
JL
,
Morency
E
,
Anguish
LJ
,
Coonrod
SA
. 
Potential role for PAD2 in gene regulation in breast cancer cells
.
PLoS One
2012
;
7
:
e41242
.
11.
McElwee
JL
,
Mohanan
S
,
Griffith
OL
,
Breuer
HC
,
Anguish
LJ
,
Cherrington
BD
, et al
Identification of PADI2 as a potential breast cancer biomarker and therapeutic target
.
BMC Cancer
2012
;
12
:
500
.
12.
Hennighausen
L
,
Wall
RJ
,
Tillmann
U
,
Li
M
,
Furth
PA
. 
Conditional gene expression in secretory tissues and skin of transgenic mice using the MMTV-LTR and the tetracycline responsive system
.
J Cell Biochem
1995
;
59
:
463
72
.
13.
Wagner
KU
,
McAllister
K
,
Ward
T
,
Davis
B
,
Wiseman
R
,
Hennighausen
L
. 
Spatial and temporal expression of the Cre gene under the control of the MMTV-LTR in different lines of transgenic mice
.
Transgenic Res
2001
;
10
:
545
53
.
14.
Zhang
X
,
Bolt
M
,
Guertin
MJ
,
Chen
W
,
Zhang
S
,
Cherrington
BD
, et al
Peptidylarginine deiminase 2-catalyzed histone H3 arginine 26 citrullination facilitates estrogen receptor alpha target gene activation
.
Proc Natl Acad Sci U S A
2012
;
109
:
13331
6
.
15.
Ornitz
DM
,
Moreadith
RW
,
Leder
P
. 
Binary system for regulating transgene expression in mice: targeting int-2 gene expression with yeast GAL4/UAS control elements
.
Proc Natl Acad Sci U S A
1991
;
88
:
698
702
.
16.
Buehring
GC
,
Eby
EA
,
Eby
MJ
. 
Cell line cross-contamination: how aware are mammalian cell culturists of the problem and how to monitor it?
In Vitro Cell Dev Biol Anim
2004
;
40
:
211
5
.
17.
Marshall
J
. 
Transwell((R)) invasion assays
.
Methods Mol Biol
2011
;
769
:
97
110
.
18.
Eichhorn
PJ
,
Gili
M
,
Scaltriti
M
,
Serra
V
,
Guzman
M
,
Nijkamp
W
, et al
Phosphatidylinositol 3-kinase hyperactivation results in lapatinib resistance that is reversed by the mTOR/phosphatidylinositol 3-kinase inhibitor NVP-BEZ235
.
Cancer Res
2008
;
68
:
9221
30
.
19.
Mohamed
BM
,
Verma
NK
,
Davies
AM
,
McGowan
A
,
Crosbie-Staunton
K
,
Prina-Mello
A
, et al
Citrullination of proteins: a common post-translational modification pathway induced by different nanoparticles in vitro and in vivo
.
Nanomedicine
2012
;
7
:
1181
95
.
20.
Seo
DO
,
Lee
S
,
Rivier
CL
. 
Comparison between the influence of shocks and endotoxemia on the activation of brain cells that contain nitric oxide
.
Brain Res
2004
;
998
:
1
12
.
21.
Zhang
X
,
Gamble
MJ
,
Stadler
S
,
Cherrington
BD
,
Causey
CP
,
Thompson
PR
, et al
Genome-Wide analysis reveals PADI4 cooperates with Elk-1 to activate c-Fos expression in breast cancer cells
.
PLoS Genet
2011
;
7
:
e1002112
.
22.
Boehnke
K
,
Falkowska-Hansen
B
,
Stark
HJ
,
Boukamp
P
. 
Stem cells of the human epidermis and their niche: composition and function in epidermal regeneration and carcinogenesis
.
Carcinogenesis
2012
;
33
:
1247
58
.
23.
Mohanan
S
,
Horibata
S
,
McElwee
JL
,
Dannenberg
AJ
,
Coonrod
SA
. 
Identification of macrophage extracellular trap-like structures in mammary gland adipose tissue: a preliminary study
.
Front Immunol
2013
;
4
:
67
.
24.
Wang
Y
,
Li
M
,
Stadler
S
,
Correll
S
,
Li
P
,
Wang
D
, et al
Histone hypercitrullination mediates chromatin decondensation and neutrophil extracellular trap formation
.
J Cell Biol
2009
;
184
:
205
13
.
25.
Proost
P
,
Loos
T
,
Mortier
A
,
Schutyser
E
,
Gouwy
M
,
Noppen
S
, et al
Citrullination of CXCL8 by peptidylarginine deiminase alters receptor usage, prevents proteolysis, and dampens tissue inflammation
.
J Exp Med
2008
;
205
:
2085
97
.
26.
Bozic
CR
,
Gerard
NP
,
von Uexkull-Guldenband
C
,
Kolakowski
LF
 Jr
,
Conklyn
MJ
,
Breslow
R
, et al
The murine interleukin 8 type B receptor homologue and its ligands. Expression and biological characterization
.
J Biol Chem
1994
;
269
:
29355
8
.
27.
Lee
J
,
Cacalano
G
,
Camerato
T
,
Toy
K
,
Moore
MW
,
Wood
WI
. 
Chemokine binding and activities mediated by the mouse IL-8 receptor
.
J Immunol
1995
;
155
:
2158
64
.
28.
Smith
JB
,
Herschman
HR
. 
Glucocorticoid-attenuated response genes encode intercellular mediators, including a new C-X-C chemokine
.
J Biol Chem
1995
;
270
:
16756
65
.
29.
Dalwadi
H
,
Krysan
K
,
Heuze-Vourc'h
N
,
Dohadwala
M
,
Elashoff
D
,
Sharma
S
, et al
Cyclooxygenase-2-dependent activation of signal transducer and activator of transcription 3 by interleukin-6 in non-small cell lung cancer
.
Clin Cancer Res
2005
;
11
:
7674
82
.
30.
Goto
H
,
Kawano
K
,
Kobayashi
I
,
Sakai
H
,
Yanagisawa
S
. 
Expression of cyclin D1 and GSK-3beta and their predictive value of prognosis in squamous cell carcinomas of the tongue
.
Oral Oncol
2002
;
38
:
549
56
.
31.
Leis
H
,
Segrelles
C
,
Ruiz
S
,
Santos
M
,
Paramio
JM
. 
Expression, localization, and activity of glycogen synthase kinase 3beta during mouse skin tumorigenesis
.
Mol Carcinog
2002
;
35
:
180
5
.
32.
Giard
DJ
,
Aaronson
SA
,
Todaro
GJ
,
Arnstein
P
,
Kersey
JH
,
Dosik
H
, et al
In vitro cultivation of human tumors: establishment of cell lines derived from a series of solid tumors
.
J Natl Cancer Inst
1973
;
51
:
1417
23
.
33.
Pierceall
WE
,
Goldberg
LH
,
Tainsky
MA
,
Mukhopadhyay
T
,
Ananthaswamy
HN
. 
Ras gene mutation and amplification in human nonmelanoma skin cancers
.
Mol Carcinog
1991
;
4
:
196
202
.
34.
Spencer
JM
,
Kahn
SM
,
Jiang
W
,
DeLeo
VA
,
Weinstein
IB
. 
Activated ras genes occur in human actinic keratoses, premalignant precursors to squamous cell carcinomas
.
Arch Dermatol
1995
;
131
:
796
800
.
35.
Pelisson
I
,
Soler
C
,
Pechoux
C
,
Chignol
MC
,
Viac
J
,
Euvrard
S
, et al
c-myc and c-Ha-ras cellular oncogenes and human papillomaviruses in benign and malignant cutaneous lesions
.
J Dermatol Sci
1992
;
3
:
56
67
.
36.
Gasparoto
TH
,
de Oliveira
CE
,
de Freitas
LT
,
Pinheiro
CR
,
Ramos
RN
,
da Silva
AL
, et al
Inflammatory events during murine squamous cell carcinoma development
.
J Inflamm (Lond)
2012
;
9
:
46
.
37.
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
81
.
38.
Ancrile
B
,
Lim
KH
,
Counter
CM
. 
Oncogenic Ras-induced secretion of IL6 is required for tumorigenesis
.
Genes Dev
2007
;
21
:
1714
9
.
39.
Sparmann
A
,
Bar-Sagi
D
. 
Ras-induced interleukin-8 expression plays a critical role in tumor growth and angiogenesis
.
Cancer Cell
2004
;
6
:
447
58
.
40.
Aceto
N
,
Duss
S
,
Macdonald
G
,
Meyer
DS
,
Roloff
TC
,
Hynes
NE
, et al
Co-expression of HER2 and HER3 receptor tyrosine kinases enhances invasion of breast cells via stimulation of interleukin-8 autocrine secretion
.
Breast Cancer Res
2012
;
14
:
R131
.
41.
Fernando
RI
,
Castillo
MD
,
Litzinger
M
,
Hamilton
DH
,
Palena
C
. 
IL-8 signaling plays a critical role in the epithelial-mesenchymal transition of human carcinoma cells
.
Cancer Res
2011
;
71
:
5296
306
.
42.
Freund
A
,
Chauveau
C
,
Brouillet
JP
,
Lucas
A
,
Lacroix
M
,
Licznar
A
, et al
IL-8 expression and its possible relationship with estrogen-receptor-negative status of breast cancer cells
.
Oncogene
2003
;
22
:
256
65
.
43.
Freund
A
,
Jolivel
V
,
Durand
S
,
Kersual
N
,
Chalbos
D
,
Chavey
C
, et al
Mechanisms underlying differential expression of interleukin-8 in breast cancer cells
.
Oncogene
2004
;
23
:
6105
14
.
44.
Hartman
ZC
,
Poage
GM
,
den Hollander
P
,
Tsimelzon
A
,
Hill
J
,
Panupinthu
N
, et al
Growth of triple-negative breast cancer cells relies upon coordinate autocrine expression of the proinflammatory cytokines IL-6 and IL-8
.
Cancer Res
2013
;
73
:
3470
80
.
45.
Hartman
ZC
,
Yang
XY
,
Glass
O
,
Lei
G
,
Osada
T
,
Dave
SS
, et al
HER2 overexpression elicits a proinflammatory IL-6 autocrine signaling loop that is critical for tumorigenesis
.
Cancer Res
2011
;
71
:
4380
91
.
46.
Korkaya
H
,
Kim
GI
,
Davis
A
,
Malik
F
,
Henry
NL
,
Ithimakin
S
, et al
Activation of an IL6 inflammatory loop mediates trastuzumab resistance in HER2+ breast cancer by expanding the cancer stem cell population
.
Mol Cell
2012
;
47
:
570
84
.
47.
Iliopoulos
D
,
Hirsch
HA
,
Struhl
K
. 
An epigenetic switch involving NF-kappaB, Lin28, Let-7 MicroRNA, and IL6 links inflammation to cell transformation
.
Cell
2009
;
139
:
693
706
.
48.
Brighenti
E
,
Calabrese
C
,
Liguori
G
,
Giannone
FA
,
Trere
D
,
Montanaro
L
, et al
Interleukin 6 downregulates p53 expression and activity by stimulating ribosome biogenesis: a new pathway connecting inflammation to cancer
.
Oncogene
2014
;
33
:
4396
406
.
49.
Kalluri
R
. 
EMT: when epithelial cells decide to become mesenchymal-like cells
.
J Clin Invest
2009
;
119
:
1417
9
.
50.
Stadler
SC
,
Vincent
CT
,
Fedorov
VD
,
Patsialou
A
,
Cherrington
BD
,
Wakshlag
JJ
, et al
Dysregulation of PAD4-mediated citrullination of nuclear GSK3beta activates TGF-beta signaling and induces epithelial-to-mesenchymal transition in breast cancer cells
.
Proc Natl Acad Sci U S A
2013
;
110
:
11851
6
.