Journal Search Engine
Search Advanced Search Adode Reader(link)
Download PDF Export Citaion korean bibliography PMC previewer
ISSN : 1225-1577(Print)
ISSN : 2384-0900(Online)
The Korean Journal of Oral and Maxillofacial Pathology Vol.43 No.1 pp.11-16
DOI : https://doi.org/10.17779/KAOMP.2018.43.1.002

An Expression Profile of Epithelial-Mesenchymal Transition-Related Genes in Porphyromonas gingivalis-Infected Ca9-22 Oral Squamous Cell Carcinoma Cells

Dae Gun Park, Da Jeong Kim, Eun Young Lee, Bok Hee Woo, Ji Hye Lee, Hae Ryoun Park*
Department of Oral Pathology, School of Dentistry, Pusan National University
Correspondence: Hae Ryoun Park, Department of Oral Pathology, School of Dentistry, Pusan National University, Beomeo-ri, Mulgeumeup, Pusan Daehak-ro 49, Yangsan 626-870, South Korea Tel: +82-51-510-8250, Fax: +82-51-510-8249 E-mail: parkhr@pusan.ac.kr
January 31, 2019 February 15, 2019 February 22, 2019

Abstract


Recently chronic inflammation is focused on the association with cancer progression and acquisition of aggressive biologic behaviors, such as invasion, metastasis, and resistance to chemotherapeutic reagents. Due to the close vicinity within oral cavity, oral cancer may be intimately associated with chronic periodontitis. The present study was done to observe the effect of chronic periodontitis on oral cancer cells by utilizing P. gingivalis infection, a major pathogen in chronic periodontitis. We analyzed and compared the mRNA expression levels of epithelial-mesenchymal transition (EMT) markers in non-infected and P. gingivalis-infected oral cancer cells. Eighty-six genes, which are well known as EMT markers, were analyzed using commercially available EMT microarray plates, performed in triplicate. Among the 86 genes, the expression of 26 was increased (≥ 2 fold) by P. gingivalis, whereas that of 7 genes was decreased (≥ 2 fold). Our study suggests that P. gingivalis infection evokes significant changes in EMT-related genes. Further observations on molecular mechanisms underlying these changes may help to clarify the role of chronic periodontitis on cancer progression and to develop more efficient preventive and therapeutic modalities for oral cancer. (182 words)



Porphyromonas gingivalis-감염 Ca9-22 구강편평세포암종 세포에서의 상피-간엽전환 관련 유전자 발현 패턴 분석

박 대근, 김 다정, 이 은영, 우 복희, 이 지혜, 박 혜련*
부산대학교 치의학전문대학원 구강병리학교실

초록


    Pusan National University

    Ⅰ. INTRODUCTION

    Concepts on the correlation between chronic inflammation and cancer are not new and have been widely accepted. For example, inflammatory bowel diseases are known to be related to the development of colon cancer, and cholangitis is linked to the pathogenesis of cholangiocarcinoma1-3). Chronic periodontitis is the most common chronic inflammatory disease of oral cavity, and among oral cancers, more than 90% are oral squamous cell carcinoma4). Thus, it is highly probable that these two types of diseases mutually affect each other. In spite that numerous studies provide and support emerging views on the role of chronic inflammation in development of cancers, correlation between chronic periodontitis and oral cancers has not been actively studied. It has been continuously revealed that infectious agents are main causative factors leading to inflammation-associated cancer1,2). Hepatitis B and C viruses are well known predisposing factors for hepatocellular carcinoma, and infection with a specific subtype of human papilloma virus may initiate cervical cancer5,6). Though numerous examples of infectious agents are found in association with cancers, most microbial agents which are involved in carcinogenesis are not bacterial but viral 8). Not many bacterial agents were recognized as a carcinogen, and the role of bacterial agentinduced inflammation in the process of tumor development has not been defined. Only H. pylori, a causative factor in chronic gastritis, was established as a carcinogen in gastric cancer7). In this study, we used oral squamous cell carcinoma cells and gram-negative anaerobic bacteria, Porphyromonas gingivalis, a major pathogen causing chronic periodontitis, for the study on the effect of chronic periodontitis on oral cancer progression.

    Experimental studies have reported that P. gingivalis can invade a variety type of host eukaryotic cells, including gingival epithelial cells and endothelial cells8-10). Thus, P. gingivalis, which are present in or near the area of oral mucosa, can invade and may provoke genetic changes in adjacent oral cancer cells. Considering the involvement of H. pylori in gastric cancer progression, investigations on changes in gene expression of oral cancer cells to invading P. gingivalis may be a meaningful strategy to define a role of P. gingivalis in oral cancer progression as well as a link between chronic periodontitis and oral cancer.

    Epithelial-mesenchymal transition (EMT), a phenotypic change which epithelial cells acquire mesenchymal characteristics, is a commonly observed in the cancer progression. Through EMT cancer cells gain not only higher migratory and invasive properties, but also resistance to chemotherapeutic reagents11-15). Information on P. gingivalis-induced changes in the expression level of genes which lead to EMT would contribute to the development of therapeutic modalities against intractable oral cancer. Accordingly, we observed changes of transcriptional profiles in oral cancer cells using microarrays to monitor EMT-related genes influenced by P. gingivalis infection.

    Ⅱ. MATERIALS AND METHODS

    Bacteria and culture conditions

    The P. gingivalis strain 381 was cultured anaerobically in trypticase soy broth, supplemented with yeast extract (1 mg/ml), hemin (5 μg/ml), and menadione (1 μg/ml) at 37°C overnight.

    Oral cancer cell culture

    Ca9-22 oral squamous cell carcinoma (OSCC) cells from Japanese Collection of Research (BioSource Cell Bank, Japan) were cultured as a monolayer in minimum essential medium (MEM; Hyclone, Logan, UT) containing 10% fetal bovine serum (FBS; Atlas Biologicals, Fort Collins, CO) and 1% penicillin-streptomycin (GIBCO-BRL, Rockville, MD, USA) at 37°C in a humidified 5% CO2-95% air incubator.

    Bacterial infection

    Before infection, P. gingivalis were washed twice and resuspended in phosphate-buffered saline (PBS). The number of bacteria in suspension was determined in a spectrophotometer. Oral cancer cells were then incubated with P. gingivalis at a multiplicity of infection (MOI) of 100 for 2 hours in a CO2 incubator and then washed with PBS three times to remove non-invaded adherent and suspended bacteria. Those P. gingivalis–infected oral cancer cells were incubated under a normal cell culture condition before harvest.

    RNA isolation and analysis of gene expression by the EMT PCR array

    To analyze the expression profiles of EMT-related genes, total RNAs from non-infected control and P. gingivalisinfected oral cancer cells were extracted using Trizol (Invitrogen Life Technologies, Carlsbad, CA, USA) and purified by the RNeasy MiniElute Cleanup kit (Qiagen, Valencia, CA, USA). For quality control, RNA purity was evaluated by monitoring OD 260/280 ratio before amplification using Agilent 2100 Bioanalyzer (Agilent Technologies, Palo Alto, USA).

    cDNAs were synthesized using the RT2 first strand kit and were then added to RT2 qPCR Master Mix, which contains SYBR Green and reference dye. Real-time PCR was performed using the ABI 7500 thermal cycler. A RT2 Profiler PCR array panel was used to investigate the changes 84 EMT-related key genes. Fold changes in mRNA expression were defined with the ΔΔCt method. Differences between non-infected and P. gingivalis-infected oral cancer cells were analyzed using Array Data Analysis Web Portal from the following address: http://www.sabiosciences.com/pcrarraydataanalysis.

    Statistical analysis

    Data were obtained from at least 3 separate experiments, and the results were expressed as the mean values ± SD. Genes of a fold regulation greater than 2 or smaller than -2 were considered to display meaningful change. Statistical significance of differential gene expression between two groups was determined using Student t-tests. P values less than 0.05 were considered statistically significant.

    Ⅲ. RESULTS

    As discussed above, it can be suspected that chronic periodontitis may contribute to aggressiveness of oral cancer cells by modulating biologic behaviors of the cells through changes in EMT-related genes. The present study was done to define the role of chronic periodontitis on the oral cancer progression. To mimic chronic periodontitis, oral cancer cells were infected with P. gingivalis, a major pathogen in chronic periodontitis, at an MOI of 1, 10, and 100. The presence of P. gingivalis within oral cancer cells was first confirmed by observing P. gingivalis 16S ribosomal RNA from the cancer cells in a dose-dependent manner (Fig. 1A). The expression of 16S rRNA, an indicator of P. gingivalis, in oral cancer cells which was infected at an MOI of 100, were decreased in a time-dependent manner (Fig. 1B).

    Gene expression differences between P. gingivalis-infected and non-infected OSCC cells were compared using RT2 PCR Array which profiles levels of 84 EMT-related genes. Only genes with a fold change greater than twofold and statistical significance were listed. Genes of multiple signaling pathways which drive EMT were upregulated in P. gingivalis-infected oral cancer cells in compared to non-infected cells. For example, P. gingivalis-infected oral cancer cells were found to 5 fold overexpress NOTCH1, one of the Notch family of trans-membrane receptors, relative to non-infected cells. In addition, factors of Wnt/β-catenin signaling pathway as well as BMPs were significantly upregulated. TIMP1, an inhibitor of matrix metalloproteinase, were downregulated (-2.5 fold), suggesting that invasive ability of P. gingivalis-infected oral cancer cells might be promoted by that MMPs were unrestrained (Table 1 & 2).

    The array data showed significant changes in many genes involved in tumor invasion and metastasis, supporting that both chronic inflammation and EMT are known to be related to the aggressiveness of cancer cells, such as invasive and metastatic abilities. The upregulated expression of several genes in P. gingivalis-infected OSCC cells was further validated by real-time PCR. The result on such as expression of MMP2 gene strongly suggests that P. gingivalis may increase invasiveness of oral cancer cells by degrading extracellular matrix components within tumor microenvironment. In addition, expressions of important transcription factors and its upstream and/or downstream effectors in the process of EMT, such as wnt and TCF4, were found to be significantly upregulated in P. gingivalis-infected oral cancer cells, implying that P. gingivalis could strongly induce EMT (Fig. 2).

    Ⅳ. DISCUSSION

    Inflammatory stimuli, which are induced by immune cells as well as the presence of microbial agent in tumor microenvironment, are known to modulate numerous factors that are involved in promoting EMT of cancer cells. The cells which encounter those stimuli exhibit higher durability and resistance to therapeutic agents by acquiring EMT characteristics11-15). In the present study, we aimed to investigate whether inflammatory insults by P. gingivalis, which is a representative pathogen in chronic periodontitis, could evoke transcriptional changes in oral cancer cells using a microarray analysis. Before considering EMT-related changes, induction of inflammatory signal by P. gingivalis infection was first identified by observing increased expression of SERPINE1. Then we found significant differences in the EMT-related gene expression profiles of P. gingivalis-infected oral cancer cells, including the genes SNAI1 and SNAI3, which are major transcription factors involved in EMT. It is well that these upregulated transcriptional factors suppress the expression of E-cadherin and increase the expression of vimentin or N-cadherin, mesenchymal markers. Increased expression of CDH2(N-cadherin gene), which is a subsequent phenomenon of SNAI1 increase, was also observed, suggesting that P. gingivalis infection leads to formation of aggressive oral cancer cells that can be highly invasive and metastatic.

    Wnt/β-catenin signaling is one of the most important pathways that are involved in EMT as well as the aggravation of cancer malignancy. Binding of wnt ligands to cell membrane activates the signaling pathway, and β-catenin is stabilized and translocated into nucleus. Wnt/β-catenin signaling pathway activate expression of numerous genes by forming a complex with nuclear TCF416). The increased expression of β-catenin/ TCF4 complex-responsive genes play a role in tumor progression and acquisition of aggressive behavior of cancer cells17,18). Upregulated WNT ligands in P. gingivalis-infected OSCC cells in the present study suggest that periodontal pathogen may contribute to oral cancer progression by activating Wnt/β-catenin signaling. P. gingivalis infection additionally activate transcriptional activity of β-catenin in oral cancer cells by upregulating TCF4 gene expression, emphasizing the role of chronic periodontitis in oral cancer progression.

    Though a direct correlation between P. gingivalis infection and aggressiveness of oral cancer cells cannot be concluded from this study and more reasonable evidences for the association are needed, this study showed alterations of EMT-related genes in P. gingivalis-infected oral cancer cells, suggesting a putative contribution of chronic periodontitis to the aggressiveness of OSCC cells. Further study based on the current findings would reveal the role of chronic periodontitis in oral cancer progression and provide a useful insight into the development of therapeutic modalities against inflammation-related oral cancer

    ACKNOWLEDGEMENTS

    This work was supported by a 2-year Research Grant of Pusan National University

    Figure

    KAOMP-43-1-11_F1.gif

    Quantitative real-time PCR assay (qPCR) determining relative quantity of P. gingigvalis rRNA. (A) Ca9-22 oral squamous cell carcinoma (OSCC) cells were infected with P. gingivalis for 2 h at an MOI of 1, 10, 100. After 24 h, total RNA was extracted from the cells. Relative level of infection was detected by the presence of 16S rRNA detected with specific primers. (B) Ca9-22 OSCC cells were infected with P. gingivalis at an MOI of 100 for 2h. Levels of 16S rRNA in P. gingivalis-infected OSCC cells were observed at 3, 6, 24, and 48 h after P. gingivalis infection.

    KAOMP-43-1-11_F2.gif

    Differential mRNA expression of non-infected and P. gingivalis-infected Ca9-22 OSCC cells. Gene expression was assessed using quantitative real-time PCR assay.

    Table

    Upregulated genes

    Downregulated genes

    Reference

    1. Balkwill F and Mantovani A: Inflammation and cancer: back to Virchow? Lancet 2001;357:539-545.
    2. Mantovani A, Allavena P, Sica A and Balkwill F: Cancer-related inflammation. Nature 2008;454: 436-444.
    3. Folseraas T, Boberg KM: Cancer Risk and Surveillance in Primary Sclerosing Cholangitis. Clin Liver Dis 2016; 20:79-98.
    4. Natarajan E, Eisenberg E: Contemporary concepts in the diagnosis of oral cancer and precancer. Dent Clin North Am 2011;55:63-88.
    5. Arzumanyan A, Reis HM, and Feitelson MA: Pathogenic mechanisms in HBV-HCV-associated hepatocellular carcinoma. Nat Rev Cancer 2013;13:123-135.
    6. Moody CA and Laimins LA: Human papillomavirus oncoproteins: pathways to transformation. Nat Rev Cancer 2010;10:550-560.
    7. Polk DB, Peek RM: Helicobacter pylori gastric cancer and beyond. Nat Rev Cancer 2010;10:403-414.
    8. Belanger M, Rodriugues PH, Dunn WA, Progulske-Fox A: Autophagy: a highway for porphyromonas gingivalis in endothelial cells. Autophagy 2006;2:165-170.
    9. Belton CM, Izutsu KT, Goodin PC, Park Y, and Lamont RJ: Fluorescence image analysis of the association between Porphyromonas gingivalis and gingival epithelial cells. Cell Microbiol 1999;1:215-224.
    10. Dorn BR, Dunn WA Jr, Progulske-Fox A: Porphyromonas gingivalis traffics to autophagosomes in human coronary artery endothelial cells. Infect Immun 2001;69:5698-708.
    11. Masui T, Ota I, Yook JI, Mikami S, Yane K, Yamanaka T et al: Snail-induced epithelial-mesenchymal transition promotes cancer stem cell-like phenotype in head and neck cancer cells. Int J Oncol 2014; 44:693-699.
    12. Wang X, Ling MT, Guan XY, Tsao SW, Cheung HW, Lee DT et al: Identification of a novel function of TWIST, a bHLH protein, in the development of acquired taxol resistance in human cancer cells. Oncogene 2004;23:474-482.
    13. Vega S, Morales AV, Ocaña OH, Valdés F, Fabregat I, Nieto MA: Snail blocks the cell cycle and confers resistance to cell death. Genes & development. 2004; 18:1131-1143.
    14. Polyak K, Weinberg RA: Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits. Nat Rev Cancer. 2009; 9:265-273.
    15. Kong D, Li Y, Wang Z, Sarkar FH: Cancer Stem Cells and Epithelial-to-Mesenchymal Transition (EMT)-Phenotypic Cells: Are They Cousins or Twins? Cancers (Basel). 2011; 3:716-729.
    16. Zhan T, Rindtorff N, Boutros M: Wnt signaling in cancer. Oncogene 2017; 36:1461-1473.
    17. Emons G, Spitzner M, Reineke S, Möller J, Auslander N et al.: Chemoradiotherapy Resistance in Colorectal Cancer Cells is Mediated by Wnt/β-catenin Signaling. Mol Cancer Res 2017;15:1481-1490.
    18. Hrckulak D, Janeckova L, Lanikova L, Kriz V, Horazna M et al.: Wnt Effector TCF4 Is Dispensable for Wnt Signaling in Human Cancer Cells. Genes (Basel) 2018;9. pii: E439.
    오늘하루 팝업창 안보기 닫기