• Nenhum resultado encontrado

Methylation-sensitive high resolution melting (MS-HRM) analysis of SOCS1, P16 and TIMP2 genes in LPS-stimulated human gingival fibroblastos = Análise específica de metilação por melting de alta resolução (MS-HRM) dos genes SOCS1, P16 e TIMP2 em fibroblast

N/A
N/A
Protected

Academic year: 2021

Share "Methylation-sensitive high resolution melting (MS-HRM) analysis of SOCS1, P16 and TIMP2 genes in LPS-stimulated human gingival fibroblastos = Análise específica de metilação por melting de alta resolução (MS-HRM) dos genes SOCS1, P16 e TIMP2 em fibroblast"

Copied!
33
0
0

Texto

(1)

UNIVERSIDADE ESTADUAL DE CAMPINAS

FACULDADE DE ODONTOLOGIA DE PIRACICABA

MAYRA DE FARIA FRANÇA LEITE

METHYLATION-SENSITIVE HIGH RESOLUTION

MELTING (MS-HRM) ANALYSIS OF SOCS1, P16 AND TIMP2

GENES IN LPS-STIMULATED HUMAN GINGIVAL

FIBROBLASTS

ANÁLISE ESPECÍFICA DE METILAÇÃO POR MELTING

DE ALTA RESOLUÇÃO (MS-HRM) DOS GENES SOCS1, P16 E

TIMP2 EM FIBROBLASTOS GENGIVAIS HUMANOS

Piracicaba

2017

(2)

MAYRA DE FARIA FRANÇA LEITE

METHYLATION-SENSITIVE HIGH RESOLUTION

MELTING (MS-HRM) ANALYSIS OF SOCS1, P16 AND TIMP2

GENES IN LPS-STIMULATED HUMAN GINGIVAL

FIBROBLASTS

ANÁLISE ESPECÍFICA DE METILAÇÃO POR MELTING

DE ALTA RESOLUÇÃO (MS-HRM) DOS GENES SOCS1, P16 E

TIMP2 EM FIBROBLASTOS GENGIVAIS HUMANOS

ESTIMULADOS POR LPS

Dissertação apresentada à Faculdade de Odontologia de Piracicaba da Universidade Estadual de Campinas como parte dos requisitos exigidos para a obtenção do título de Mestra em Biologia Buco-Dental, na Área de Histologia e Embriologia

Dissertation presented to the Piracicaba Dental School of the University of Campinas in partial fulfillment of the requirements for the degree of Master in Dental Biology, in Histology and Embryology

Orientador: Profa. Dra. Ana Paula de Souza

Coorientador: Profa. Dra. Aline Cristiane Planello

ESTE EXEMPLAR CORRESPONDE À

VERSÃO FINAL DA DISSERTAÇÃO DEFENDIDA PELA ALUNA MAYRA DE FARIA FRANÇA LEITE E ORIENTADA PELA PROFESSORA ANA PAULA DE SOUZA.

Piracicaba

2017

(3)
(4)
(5)

DEDICATÓRIA

Dedico este trabalho à minha avó Odila, ao meu avô Sérgio (in memorian)

e à minha mãe Márcia por serem os meus grandes exemplos de amor e profissionais

da área da Saúde.

(6)

AGRADECIMENTOS

A Deus e aos meus amados pais pela vida e por todo o amor recebido.

À Faculdade de Odontologia de Piracicaba da Universidade Estadual de

Campinas (FOP/UNICAMP).

À Profa. Dra. Ana Paula de Souza pela oportunidade de ingressar na

Pós-Graduação, pela sua orientação, pelos seus conhecimentos compartilhados, pelo seu

fundamental apoio e principalmente pela sua importância no meu crescimento pessoal

e profissional.

À minha coorientadora Aline Cristina Planello pela sua orientação, pelos

conhecimentos compartilhados e pela amizade.

Ao Prof. Dr. Sérgio Roberto Peres Line, o qual permitiu que eu pudesse

ingressar no universo da pesquisa durante a minha graduação, pelo apoio e amizade.

À CAPES pela Bolsa de Mestrado.

À FAPESP pelo financiamento do projeto de pesquisa.

A todos os Professores da Área de Histologia e Embriologia da

FOP/UNICAMP pelos conhecimentos compartilhados, pelo apoio e amizade.

Aos meus amigos de Departamento e Laboratório, em especial ao Gabriell

Borgato, pela enorme participação na realização do meu projeto de mestrado.

Aos meus irmãos Jaqueline e Rodrigo pelo amor, incentivo e apoio

incondicional.

Ao meu namorado Alan pelo amor, incentivo e apoio incondicional.

Aos meus amigos por todo apoio e carinho, em especial à minha amiga

Joyce.

(7)

RESUMO

Objetivo: O papel do lipopolissacarídeo bacteriano (LPS) para desencadear a

inflamação periodontal é bem conhecido. No entanto, o efeito do LPS na modulação

do estado de metilação do DNA não está completamente claro. Aqui, investigamos o

efeito do LPS de P. gingivalis no perfil de metilação da região reguladora dos genes

SOCS1, P16 e TIMP2 em fibroblastos gengivais humanos cultivados, a fim de avaliar

se o LPS pode alterar o estado de metilação desses genes. Material e Métodos: As

células de fibroblastos gengivais humanos imortalizadas foram cultivadas em meio

suplementado com LPS de P. gingivalis ou com o veículo do LPS de P. gingivalis

durante 12, 24 e 48 horas. A seguir, a viabilidade celular foi avaliada por teste de MTT.

O DNA genômico foi isolado e convertido por bissulfito de sódio. O estado de

metilação da região reguladora dos genes P16, SOCS1 e TIMP2 foi analisado por

PCR específica de metilação seguida por análise de melting de alta resolução

(MS-HRM). Resultados: Tanto fibroblastos estimulados por LPS quanto os fibroblastos

tratados apenas com o veículo do LPS não mostraram diferenças significativas na

região analisada dos genes P16, SOCS1 e TIMP2 durante os períodos de 12, 24 e 48

horas. Conclusão: Pequenos períodos de estimulação com LPS não foram suficientes

para promover alterações de metilação nas regiões analisadas dos genes SOCS1,

P16 e TIMP2 em fibroblastos. Relevância clínica: Compreender o papel das moléculas

derivadas de periodontopatógenos na promoção de alterações de metilação dentro da

região reguladora de genes-chaves associados à periodontite é importante para

aumentar o reconhecimento da área e pode abrir portas para o desenvolvimento de

novas terapias adjuvantes.

Palavras-chave: Epigenética, metilação do DNA, lipopolissacarídeo, LPS,

fibroblasto, MS-HRM

(8)

ABSTRACT

Objective: The role of bacterial lipopolysaccharide (LPS) to trigger periodontal

inflammation is well known. However, the effect of LPS in modulating the methylation

status of DNA is not completely clear. Here, we investigated the effect of P. gingivalis

LPS on the methylation profile of regulatory region of SOCS1, P16 and TIMP2 genes

in cultured human gingival fibroblast in order to assess whether LPS can alter the

methylation status of these genes. Material and Methods: Immortalized human gingival

fibroblast cells were cultured in medium supplemented with P. gingivalis LPS or P.

gingivalis LPS vehicle during 12, 24 and 48 hours. Following, cell viability was

assessed by MTT test. Genomic DNA was isolated and converted by sodium bisulfite.

The methylation status of the regulatory region of P16, SOCS1 and TIMP2 genes was

analyzed by methylation specific PCR followed by high resolution melting analysis

(MS-HRM). Results: Either fibroblast LPS-stimulated or fibroblast treated only with

vehicle did not show significant differences in the analyzed region of P16, SOCS1 and

TIMP2 genes during the periods of 12, 24 and 48 hours. Conclusion: Short periods of

LPS stimulation were not enough to promote methylation changes in the analyzed

regions of SOCS1, P16 and TIMP2 genes in fibroblasts. Clinical Relevance:

Understanding the role of molecules derived from periodontopathogens in promoting

methylation changes within regulatory region of key genes associated with

periodontitis is important to increase the area acknowledgment and can open doors to

the development of new adjuvant therapies.

Key words: Epigenetics, DNA methylation, lipopolysaccharide, LPS, fibroblast,

MS-HRM

(9)

SUMÁRIO

1 INTRODUÇÃO

10

2 ARTIGO: Methylation-Sensitive High Resolution Melting (MS-HRM)

Analysis of SOCS1, P16 And TIMP2 Genes In LPS-Stimulated

Human Gingival Fibroblasts

13

3 CONCLUSÃO

29

REFERÊNCIAS

30

(10)

10

1 INTRODUÇÃO

A periodontite (ou doença periodontal) é uma desordem inflamatória que

afeta os tecidos de suporte dos dentes (periodonto) e que da fase aguda torna-se

crônica. Está claro que o acúmulo de bactérias sobre a superfície da região cervical

dos dentes desencadeia a doença, porém seu agravamento e severidade são

dependentes de fatores como o hábito de fumar, a genética do hospedeiro e a

condição de saúde sistêmica.

A composição genética e epigenética do hospedeiro também podem

modular o prognóstico da doença periodontal. A epigenética pode ser definida como

mudanças no padrão de expressão do gene não envolvendo a sequência do DNA

(Larsson et al., 2015). A metilação do DNA e modificações em histonas são as duas

principais alterações epigenéticas em humanos (Takai et al., 2015). Essas alterações

podem contribuir para o desenvolvimento e manutenção de doenças inflamatórias,

doenças autoimunes e do câncer (Larsson et al., 2015). A metilação do DNA é um

mecanismo epigenético primário caracterizado pela adição de um grupo metil em

citosinas precedidas de guanina em regiões nomeadas ilhas CpG. As ilhas CpG

(CGIs) são pequenas sequências intercaladas de DNA que desviam significantemente

da média do padrão genômico por serem ricas em GC e são predominantemente

não-metiladas. As ilhas CpG evidentemente contém sítios de ínicio de transcrição (TSS),

os quais coincidem com a região promotora dos genes em ratos e humanos. As

características das sequências de DNA partilhadas adaptaram as CGIs para a função

de regiões promotoras, desestabilizando nucleossomos e atraindo proteínas que

criam um estado de cromatina transcricionalmente permissivo (Deaton e Bird, 2011).

A metilação do DNA está associada à várias mudanças na estrutura da

cromatina e ao recrutamento de proteínas para os sítios metilados. A metilação

geralmente leva à obstrução da região promotora, dificultando a transcrição gênica e

subsequentemente causando o silenciamento do gene (Rettori et al., 2013). A

metilação é mediada pelas enzimas da família das DNA metiltransferases (DNMTs),

sendo a função principal da DNMT1 a manutenção da metilação, metilando sítios

hemimetilados gerados durante a replicação do DNA, e as enzimas de novo DNMT3a

e DNMT3b são responsáveis pela metilação do DNA durante o desenvolvimento

embrionário (Abiko et al., 2014). Infecções bacterianas também induzem

(11)

11

modificações epigenéticas em células humanas, e a hipermetilação do DNA tem sido

implicada na doença periodontal em humanos (Larsson et al., 2015).

Patógenos

periodontais

são

fatores

ambientais

cruciais

no

desenvolvimento da doença periodontal. Uma bactéria que é frequentemente

detectada em bolsas periodontais é a Porphyromonas gingivalis. P. gingivalis é uma

bactéria anaeróbica, gram-negativa, que possui muitos fatores de virulência como

enzimas proteolíticas, fímbrias e lipopolissacarídeos (LPS) (Palm et. al., 2015). O LPS

é utilizado como uma toxina bacteriana para provocar respostas agudas em modelos

experimentais de infecção (Ding e Jin, 2014) (Johansson et al., 1996).

Fibroblastos gengivais são o tipo mais comum de células encontradas no

tecido gengival. Além de fornecerem uma estrutura tecidual, os fibroblastos também

demonstram responderem aos patógenos associados a padrões moleculares

(PAMPs) via, por exemplo, receptores ativados por proteases (PARs) e receptores do

tipo toll (TLRs), sendo o TLR2 e o TLR4 (Palm et al., 2015). Além de manter a

integridade do tecido gengival por meio da regulação do metabolismo do colágeno e

dos proteoglicanos, os fibroblastos gengivais produzem várias citocinas

pró-inflamatórias, como a interleucina-1 (IL-1), IL-6 e IL-8 em resposta à estimulação direta

e indireta com LPS (Tabeta et al., 2000). Fibroblastos do ligamento periodontal de

humanos estimulados por um longo período por LPS apresentaram hipermetilação de

genes da matriz extracelular, e as metilações ocorridas nas regiões promotoras dos

genes FANK1, COL4A1-A2, 12A1 e 15A1, LAMA5 e B1, MMP25, POMT1 e EMILIN3

estavam relacionadas com o baixo nível de expressão transcricional desses genes

(Takai et al., 2015).

O estudo da epigenética, especialmente a metilação do DNA, oferece um

enorme potencial para a identificação de marcadores biológicos que podem ser

utilizados no diagnóstico dos primeiros estágios de desenvolvimento do câncer, e para

uma avaliação mais precisa do risco individual de desenvolver doenças (Calmon et al,

2007). A proliferação celular é regulada em parte pelo produto proteico do gene

supressor de tumor P16 (Cody et al, 1999). Tipicamente, p16 induz a parada do ciclo

celular e impede a divisão celular através da inibição das quinases dependentes de

ciclina CDK4 e CDK6, bem como a fosforilação mediada por CDK do gene do

retinoblastoma (Conceição et al, 2015). O gene P16 foi um dos primeiros genes

(12)

12

investigados em câncer de cabeça e pescoço associado com a metilação do promotor

do gene, e é amplamente investigado em câncer oral (Ha e Califano, 2006).

As metaloproteinases da matriz (MMPs) são consideradas enzimas de

grande importância na destruição do tecido conjuntivo. Essas enzimas eventualmente

degradam o ligamento periodontal e as proteínas da matriz óssea. Inibidores teciduais

das metaloproteinases da matriz (TIMPs) são inibidores específicos das MMPs, e

desempenham um papel relevante no metabolismo da matriz extracelular periodontal

através do controle da atividade das MMPs (Kubota et al., 2008). O inibidor tecidual

de metaloproteinases-2 (TIMP2) é conhecido por antagonizar a atividade das

metaloproteinases da matriz e suprimir o crescimento tumoral, angiogênese, invasão

e metástases (Parashar e Capalash, 2012). Um desequilíbrio na interação MMP-TIMP

está implicado na etiologia da doença periodontal, onde a degradação da matriz

extracelular é uma característica principal (de Souza et al., 2005).

Muitas citocinas pró-inflamatórias desempenham papéis fundamentais na

iniciação de vias de sinalização intracelular específicas. Umas das mais importantes

cascatas de sinalização induzidas por citocinas é a via JAK/STAT (JAK (Janus kinase)

/ STAT (Signal Transducer e Activator of Transcription) (Andia et al., 2015). Os

supressores de sinalização de citocinas (SOCS) são uma família de proteínas

intracelulares que participam da regulação da resposta de células imunes ao estímulo

de várias citocinas pró-inflamatórias. SOCS1 é um potente inibidor de sinalização de

citocinas (Andia et al., 2015). A metilação do DNA é um importante mecanismo que

controla os níveis de transcrição do gene SOCS1. A Ilha CpG do gene SOCS1 invade

a região do primeiro e segundo éxon (De Souza et al., 2014). A hipermetilação do

gene SOCS1 é capaz de controlar a expressão de outros genes através da interação

enhancer/promotor, e este evento tem grande importância na patogênese da

inflamação crônica (Planello, 2014).

O presente estudo teve como objetivo analisar por MS-HRM (Methylation

Sensitive High Resolution Melting) a porcentagem da metilação dos genes P16,

SOCS1 e TIMP2 em cultura de fibroblastos gengivais humanos expostos ao LPS no

(13)

13

2 ARTIGO: METHYLATION-SENSITIVE HIGH RESOLUTION MELTING (MS-HRM)

ANALYSIS SOCS1, P16 AND TIMP2 GENES IN LPS-STIMULATED HUMAN

GINGIVAL FIBROBLASTS

Submetido ao periódico Clinical Oral Investigation (Anexo 1)

Mayra de Faria França Leite, Gabriell Borgatto, Aline Cristiane Planello, Ana Paula de

Souza.

Department of Morphology Division of Histology, Piracicaba Dental School - UNICAMP, Piracicaba-SP, Brazil

Corresponding author:

Ana Paula de Souza, PhD, Department of Morphology Piracicaba Dental School – UNICAMP

Limeira Avenue, 901, 13414-018, Piracicaba, SP, Brazil +55 (19) 21065381

apsfop@unicamp.br

Financial Support: This study was supported by Fundação de Amparo a Pesquisa do Estado de São Paulo (FAPESP 2015/24749-6). Mayra de Faria França Leite and Gabriell Borgatto was supported by Conselho Nacional de Pesquisa (CNPq). Aline Cristiane Planello was supported by Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP 2014/05867-5).

(14)

14

ABSTRACT

Objective: The role of bacterial lipopolysaccharide (LPS) in triggering periodontal inflammation is well known. However, the effect of LPS in modulating the methylation status of DNA is not completely clear. Here, we investigated the effect of P. gingivalis LPS on the methylation profile of regulatory region of SOCS1, P16 and TIMP2 genes in cultured human gingival fibroblast in order to assess whether LPS can alter the methylation status of these genes.

Material and Methods: Immortalized human gingival fibroblast cells were cultured in medium supplemented with P. gingivalis LPS or P. gingivalis LPS vehicle during 12, 24 and 48 hours. Following, cell viability was assessed by MTT test. Genomic DNA was isolated and converted by sodium bisulfite. The methylation status of the regulatory region of P16, SOCS1 and TIMP2 genes was analyzed by Methylation-Sensitive High Resolution Melting (MS-HRM).

Results: Neither fibroblast LPS-stimulated nor fibroblast treated only with vehicle showed significant differences in the analyzed region of P16, SOCS1 and TIMP2 genes during the periods of 12, 24 and 48 hours of treatment.

Conclusion: Short periods of LPS stimulation were not enough to promote methylation changes in the analyzed regions of SOCS1, P16 and TIMP2 genes in fibroblasts.

Clinical Relevance: Understanding the role of molecules derived from periodontopathogens in promoting methylation changes within regulatory region of key genes associated with periodontitis is important to increase the area acknowledgment - opening doors to the development of new adjuvant therapies.

(15)

15

INTRODUCTION

Periodontitis represents the most important oral disease of industrialized countries, responsible for teeth-lost in more than 20% of adult population [1]. It is an infection-associated inflammation disease induced by bacteria-derived molecules, such as lipopolysaccharide (LPS), that promotes the collagen type I breakdown and its solubilization from soft and hard periodontal tissues [2, 3].

In the last decades, explosion of studies have focused on the molecular basis of periodontitis in order to find the pathways that regulate the host tissues destruction. The understanding of how signal factors, cytokines, growth factors and transcriptional factors act during periodontitis have raised the knowledge of area. However, the investigation of factors directly associated with gene function is essential to clarify the real mechanisms.

Addressed to this topic, epigenetic emerges as the field committed to investigate the genome regulation. Epigenetic is the study of DNA/histone chemical changes inherited after mitotically and meiotically cell divisions that control chromatin structure and, consequently, gene functions [4,5]. Several proteins and enzymes take part in the molecular basis of epigenetics that involves histones modifications, noncoding RNAs and DNA methylation. Surely, DNA methylation does represent the most studied epigenetic change in the human genome that is carried out by DNA methyltransferases (DNMTs): the maintenance methyltransferase DNMT1 and de novo methyltransferases DNMT3A and DNMT3B. CpG enriched regions (named as CpG islands) are not randomly distributed around the whole genome and tends to be concentrated within regulatory regions such as gene promoters [6]. Ordinarily, CpG islands are found unmethylated but can become methylated during diseases, especially during tumor development and chronic inflammation.

We have reported that LPS can modify the transcription level of some chromatin modifying enzymes in human keratinocytes cells [7]. Adding, a few studies have shown that LPS-stimulated cells can answer differentially to epigenetic drugs [8] as well as extracellular matrix-related genes can become hypermethylated in human periodontal fibroblasts stimulated for a prolonged period with LPS derived from P. gingivalis [9].

Therefore, in the present paper we hypothesized whether LPS derived from P. gingivalis could be able to promote DNA methylation changes in the regulatory region of three genes that contain CpG islands and are implicated in different cell functions: SOCS1 a member of Suppressor Of Cytokine Signaling family that acts as classic negative feedback inhibitors of JAK/STAT pathway and is key physiological regulator of both innate and adaptive immunity [10]; P16 (also known as P16INK4a or CDKN2A) a tumor suppressor gene implicated in the prevention of several cancers, including oropharyngeal squamous cell carcinoma, cervical cancer, esophageal cancer [11], considered a key factor in controlling the G1 phase of cell

(16)

16

cycle; and TIMP2 (Tissue Inhibitor of Metalloproteinases-2) a member of TIMP family responsible for the physiological inhibition of matrix metalloproteinases (MMPs), whose imbalance can lead to connective tissue destruction [12]. Our analysis were carried on human gingival fibroblasts and MS-HRM assay was performed in order to quantify the percentage of DNA methylation in LPS-stimulated and non-stimulated cells.

MATERIAL AND METHODS

Materials

DMEM medium (Dulbecco’s Modified Eagle’s medium, Sigma Chemical Co.) was obtained from Cultilab (São Paulo, Brazil). Heat-inactivated fetal bovine serum (FBS) and trypsin/ ethylenediamine tetraacetic acid (EDTA) solution used in all experiments were purchased from LGC Biotecnologia (São Paulo, Brazil). The antibiotics (penicillin/streptomycin/) were purchased from GIBCO (Grand Island, NY, USA). Tissue culture multiwell plates were obtained from TPP® (Switzerland). P. gingivalis lipopolysaccharide (P. gingivalis LPS), extracted from P. gingivalis strain ATCC 3327, was obtained from InvivoGen (cat. code: tlrl-pglps, San Diego, CA, USA). For the viability analysis, a MTT assay was purchased from Invitrogen, Thermo Fisher Scientific. Quick-DNA Universal kit was obtained from Zymo Research. MethyISEQr Bisulfite Conversion kit was purchased from Life (Carlsbad, CA, USA). LightCycler®480 HRM Master Mix was purchased from Roche (Mannheim, Germany). EpiTect PCR Control DNA Set was obtained from Qiagen.

Cell Culture and LPS Treatment

Immortalized human gingival fibroblast cell line (HGF-1, ATCC® CRL-201) were cultured in DMEM medium supplemented with 10% FBS (fetal bovine serum) and 1% antibiotics in the humidified atmosphere (5% CO2) at 37oC with subculture every 3 days. At harvest, the cells were plated at a concentration of 1x105/mL and were exposed to P. gingivalis LPS at concentration of 1g/mL in serum re-supplemented with 10% FCS. The experiments were carried on at 12, 24 and 48 hours and after each 24 hour new amount of DMEM medium containing 10% FBS and LPS (1g/mL) was replaced. Culture treated with LPS vehicle (deionized water used to dilute LPS) was used as control. The experiment was performed in triplicate.

(17)

17

Viability Cell Assay

Cell viability was assessed by MTT reduction to formazan crystals by mitochondrial enzyme reductases using a MTT biochemical assay in 96-well plates. 1x105 cells/mL were seeded in wells and the assay was carried on in triplicate for each experimental time. MTT assay was performed following the manufacturer ́s protocol (Invitrogen). Finally, the multiwell plate was mixed until complete salt crystal dissolution and absorbance was measured in an ELISA reader (Molecular Devices, CA, USA) using the software VersaMax (test wavelength 570 nm, reference wavelength 630 nm).

DNA Extraction and Bisulfite Treatment

DNA was purified using Quick-DNA Universal kit, according to the manufacturer ́s protocol. After, 0.5 g of gDNA was bisulfite converted by MethyISEQr Bisulfite Conversion kit, following the manufacturer ́s protocol. The samples were short time storage at 04C and used for MS-HRM.

Methylation Sensitive High Resolution Melting (MS-HRM)

Real-time PCR followed by HRM was carried out in Light Cycler 480 II (Roche, Mannheim, Germany). The sequence of the primer set used is shown in Table 1. The reaction mixture consisted of 18 ng of bisulfite-converted DNA, 1x LightCycler®480 HRM Master Mix, 150 nM of each primer, 3 mM of MgCl+2 in a final volume of 20 μl. The reaction conditions were performed in a equal start for all genes at 95°C for 10 minutes, followed by specific cycles for each gene: P16 – 45 cycles at 95°C for 10 seconds, 64°C for 10 seconds, 72 °C for 10 seconds; TIMP2 – 45 cycles at 95°C for 10 seconds, 59°C for 05 seconds, 72 °C for 10 seconds; SOCS1 – 45 cycles at 95°C for 10 seconds, 55°C for 04 seconds, 72 °C for 10 seconds. MS-HRM analyses were performed at the temperature ramping and fluorescence acquisition settings recommended by the manufacturer, 1 minute at 95 °C, a hold at 70 °C for 1 minute to allow re-anneling of all PCR product, then the acquisition step started ramping from 70 °C to 95 °C, rising by 0,2 °C/s with 25 acquisitions per °C.

To estimate the methylation level of the samples, converted fully methylated and fully unmethylated DNA (EpiTect PCR Control DNA Set) were used to prepare dilution series as controls. The dilution series of relevant methylated DNA in a background of unmethylated DNA

(18)

18

were prepare in the concentration of 0%, 5%, 10%, 25%, 50% and 100% of methylation to build standards curves. PCR bias toward unmethylated DNA was reversed following the guideline published by Wojdacz (22). Standard curves and no template controls were included in each experimental run. As a negative control, genomic unconverted DNA was tested once with each primer. Initial assays were first run using 50% and 0% controls until the proportionality of amplification was achieved. MS-HRM data were normalized with the Light Cycler 480 II analysis software to compensate for varying starting fluorescence levels. The amplicon melting profile of each sample was compared to standard curves, and based on that, samples of each individual was classified into different methylation categories; 0-5, 6-10, 11- 25, 26-50 and 51-100%. The experiment was performed in triplicate.

RESULTS

MTT assay showed no effect of LPS on HGF cells viability at concentration of 1g/mL as well this LPS concentration did not alter the morphology of cells when we observed them by microscopy (Figure 1).

Following, human gingival fibroblasts were treated with P. gingivalis LPS in order to analyze the potential of this molecule derived from bacteria in promoting methylation changes in the regulatory region of P16, TIMP2 and SOCS1 genes. The assays were performed using MS-HRM approach in order to quantify methylation changes in those genes. Either exposure of HGF cells to LPS for 12h, 24h or for 48h did not promote significant differences in the methylation profile of the analyzed regions of these genes. The MS-HRM results of P16, TIMP2 and SOCS1 genes are shown in Figure 2, 3 and 4, respectively.

DISCUSSION

Cell functions such as activation, proliferation, and survival are differently controlled during chronic inflammation. It occurs not only due the number of new cascades of signal transduction pathways installed during inflammation, that activate different transcriptions factors that take part in the immune-inflammatory response, but also due the chromatin-remodeling that allows or prohibits the access of transcription factors to different regions of genome [13,14].

(19)

19

LPS is a component of the cell wall of gram-negative bacteria responsible for inducing expression of a range of pro-inflammatory cytokines such as TNF-α, IL-1β and IL-6 [15]. Injection of sub-lethal doses of LPS into animals demonstrated that a state of “LPS tolerance” was induced in the animals [16]. Interesting, while some genes became unable to answer to LPS-stimulation another group of genes entitled “non-tolerizable” escape from the suppressive effect of LPS tolerance [15]. Addressed to it, studies have demonstrated that gene promoters can be regulated in order to confer their tolerized status and apparently, the mechanism associated to it is chromatin-remodeling. Histone-modifications are found associated with LPS stimulation. Different histone marks are found as consequence of LPS stimulation and depending on if acute or whether prolonged (chronic) stimulation, these marks are different and induce an open or compacted chromatin status. For example, upon an acute LPS stimulation, H3K9 unmethylated is found in naïve human monocytes, while H3K9 dimethylation is observed in the same but tolerized cells [17, 18, 19].

In addition to histone modifications, DNA methylation is other epigenetic event able to regulate gene expression and confers gene tolerance to LPS. Unfortunately, DNA methylation was not widely investigated in acquire LPS-tolerance until moment, but is hypothesized that DNA methylation could be the final step in the tolerization process [15]. Recently, we published a study reporting that short period of LPS stimulation modulates gene expression of some enzymes implicate with epigenetic changes, such as DNA methyltransferases (DNMT1 and DNMT3A) [7].

P. gingivalis is a distinguished gram-negative periodontal pathogen able to lead consequences on host cells and LPS is a key bacteria-derived molecule that recognizes and binds to toll-like receptors TLR2 and TLR4 to trigger its effects [8]. In this study we challenged human gingival fibroblast cells with LPS derived from P. gingivalis in order to assess how short period of LPS-stimulation could modify the methylation profile of SOCS1, P16 and TIMP2 genes that are associated with different cell functions. Performing MS-HRM analysis, it is possible to verify percentages changes of methylation within DNA in a scale less than 1%.

SOCS family is composed by eight members that share similar structure. However, only SOCS1 and SOCS3 contain in the N-terminus sequence of 12-amino acids capable to inhibit the kinase activity of JAK2, due the presence of a pseudo-substrate for JAK2 in this additional region. Transcription of SOCS1 can be induced by cytokines that trigger JAK-STAT signaling pathway. Although gene promoter of SOCS1 shows motif to STAT-binding, the expression of SOCS1 leads the inhibition of JAK-STAT signaling via down-regulation of STAT activation due the blockage of kinase activity by the pseudo-substrate present in SOCS1. Reports have suggested that SOCS1 down-regulates the TLR signaling via LPS sensitivity

(20)

20

modulation. SOCS1-/- and INF-γ-/- mice showed an increased response to LPS when compared to only INF-γ-/- mice. Added, SOCS1-/- macrophages were hyper-responsive to LPS, indicating that SOCS1 can play a key role in the regulation of TLR via LPS signaling [20].

The analysis of SOCS1 genes did not shown any methylation changes in DNA when a short period of LPS-stimulation was assayed. Epigenetic changes in the intragenic region of SOCS1 have been frequently associated with chronic inflammation [21]. SOCS1 is a small gene that contains a CpG island (CGI) inside the promoter region that overlaps the first and second exons. This CGI is frequently observed hypermethylated in chronic disease as hepatitis and some types of cancer, taking part in the CGI methylator phenotype (CIMP) panel in colon cancer. Positive correlation between presence of methylation in regulatory region of SOCS1 and down-regulation of gene transcription is not always found. Recently, we published a paper showing that this same region of SOCS1 may act as an enhancer element over other genes of chromosome 16 in gingival tissue [21].

The tumor suppressor gene P16 (P16INK4a) plays a key role in cell cycle regulation by

decelerating cells progression from G1 phase to S phase. The protein is encoded by the CDKN2A locus that is located within human chromosome 9p21. This protein is a member of INK4 family of cyclin-dependent kinase inhibitors, mainly known to form a complex with CDK4 and CDK6 [23]. Although the mainly function of P16 is associated with tumor prevention, other different functions have been described for P16 in chronic inflammation. Proliferation of synovial fibroblasts is an ordinary event in rheumatoid arthritis trigged by molecules of inflammation. The intra-articular gene transfer of P16 was capable to inhibit synovial hyperplasia, immune-inflammatory cells infiltration, bone and cartilage joint destruction [24,25]. These findings are credit to the ability of P16 in inhibiting CDK4 and CDK6 that are responsible to release E2F transcription factors that allow cell-cycle progression [26]. P16 can also modulate the pro-inflammatory transcription factors NF-kB and AP-1 via its ankyrin motifs that bind c-jun [27] and NF-kB [28] and inhibit their activity. However, this P16 function is not available apparently in all cell types.

P16 gene is frequently mutated or deleted in a variety of cancer types as well as it can be silencing by hypermethylation. The P16 gene is found methylated in oral, esophageal and lung squamous cell carcinoma, in all of them the hypermethylation suppresses the mRNA transcription and, consequently, the protein expression [29]. In this study we tested the thesis of P16 gene becomes methylated in fibroblast as a consequence of LPS-stimulation. However, the results showed P16 completely unmethylated after LPS-stimulation for a short period.

TIMP-2 is one of a group of four members that compose the TIMP family, the natural tissue inhibitors of matrix metalloproteinases (MMPs). The inhibition is reached by the

(21)

N-21

terminal region of TIMP proteins that binds to the catalytic domain of MMPs. TIMP-2 specifically binds to MT1-MMP and this bi-molecular complex takes part in the process of pro-MMP2 activation [30]. Thus, TIMP-2 represents an inhibitor and activator of MMPs. Although TIMP-2 is secreted in the extracellular matrix, having well know functions outside cells, a number of reports have suggested a other functions to TIMP proteins since these proteins link to membrane receptors of different types of cells [31]. TIMP2 gene transcription is regulated epigenetically. CpG island that overlapping TIMP2 gene promoter is commonly seen hypermethylated in MCF-7 cells, silencing the gene transcription [31]. We did not detected methylation in the TIMP2 gene after LPS-stimulation. However, we emphasize the same could not be repeated whether other assay conditions would be performed such as LPS-stimulation for long period.

Previous study reported hypermethylation of extracellular matrix-related genes when fibroblasts were induced by LPS-stimulation for a prolonged period [9]. The LPS-stimulation period was basically the difference between our study and the other one. These results agree with the clinical findings in patients. Gingivitis is an acute inflammation that shows a set of clinical signs, microscopic and molecular features completely different from the set of findings that are seen in chronic periodontitis. Probably, the long-term exposure to LPS may promote epigenetic changes that are not detectable when we performed a short-term assay.

In conclusion, LPS stimulation did not promote methylation changes in the promoter region of SOCS1, P16 and TIMP2 genes when human gingival fibroblasts were stimulated for a short period. There is no guarantee the same would be found if others genome regions, others genes, others cell types or others assay-periods would be tested. Then, we have an “open sea” of hypothesis that must be still assayed in order to increase our knowledge about the effects of LPS in promoting epigenetic changes.

(22)

22

Table 1: Sequence of MS-HRM primers set, amplicons size and number of CpG sites

analyzed in the fragments.

Gene

Primer sequences (CpG sites

underlined and converted Cs as

capital Ts or As). (5'

→ 3').

Amplicon

CpGs between

the primers set

P16

F: ggagTTttcggTtgaTtggTtggTT

R: aAcaAcgcccgcacctcctcta

69bp

05

TIMP2

F: gTaTcggggagTagTtgTagg

R: cgctcgAcctcctActActAA

66bp

06

SOCS1

F: tcgcggTtgTTatTTaggtgaaag

R: cgaAcccgtAAAcaccttcctA

140bp

12

(23)

23

Figure 1. The figure represents the % of cells viability after HGF cells exposed to P.

gingivalis LPS (1

g/mL) for 12h, 24h and 48h or only vehicle (deionized water/control)

at the same periods of time. No significant differences were observed on the HGF cells

viability after the treatments (n=3/group).

Figure 2. MS-HRM assay of P16 gene. Different percentages of DNA methylation are

shown in different color in the curves. The percentages are obtained from the mix of

methylated and unmethylated control DNAs what allowed to build the assay calibration

curve. Samples are shown as strong blue color near to 0% (brown lines). The result

indicates that the promoter of P16 gene is completely unmethylated (0%) in control

and HGF cells after all periods of LPS treatment.

0 20 40 60 80 100 120 12h 24h 48h

% Cell Viability

Control LPS(1µg/mL) 0% 5% 10% 25% 50 and 100%

(24)

24

Figure 3. MS-HRM assay of TIMP2 gene. Different percentages of DNA methylation

are shown in different color in the curves. The percentages are obtained from the mix

of methylated and unmethylated control DNAs what allowed to build the assay

calibration curve. Samples are shown as strong blue color near to 0% (blue lines). The

result indicates that the promoter of TIMP2 gene is completely unmethylated (0%) in

the control and HGF cells after all periods of LPS treatment.

Figure 4. MS-HRM assay of SOCS1 gene. Different percentages of DNA methylation

are shown in different color in the curves. The percentages are obtained from the mix

of methylated and unmethylated control DNAs what allowed to build the assay

calibration curve. Samples are shown as strong blue color near to 0% (red lines). The

result indicates that the promoter of SOCS1 gene is completely unmethylated in the

control and HGF cells after all periods of LPS treatment.

0% 5% 10% 25% 50% 100% 5% 25% 50% 100% 0% 10%

(25)

25

REFERENCES

1. Pihlstrom BL, Michalowicz BS, Johnson NW (2005). Periodontal diseases. Lancet 366:1809-1820.

2. Armitage GC (1995). Clinical evaluation of periodontal diseases. Periodontol 2000 7:39-53.

3. Scheres N, Laine ML, de Vries TJ, Everts V, van Winkelhoff AJ (2010). Gingival and periodontal ligament fibroblasts differ in their inflammatory response to viable Porphyromonas gingivalis. J Periodontol Res 45:262-270.

4. Bird AP (1986). CpG-rich islands and the function of DNA methylation. Nature 321: 209-213.

5. Bird A(2007). Perceptions of epigenetics. Nature 447, 396-398.

6. Jones PA(2012). Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet 13: 484-492.

7. de Camargo Pereira G, Guimarães GN, Planello AC, Santamaria MP, de Souza AP, Line SR, Marques MR (2013). Porphyromonas gingivalis LPS stimulation downregulates DNMT1, DNMT3a, and JMJD3 gene expression levels in human HaCaT keratinocytes. Clin Oral Investig 17: 1279-1285.

8. Shen J, Wu S, Guo W, Liang S, Li X, Yang X (2016). Epigenetic regulation of pro-inflammatory cytokine genes in lipopolysaccharide-stimulated peripheral blood mononuclear cells from broilers. Immunobiol 2985: 30370-30379.

9.Takai R, Uehara O, Harada F, Utsunomiya M, Chujo T, Yoshida K, Sato J, Nishimura M, Chiba I, Abiko Y (2016). DNA hypermethylation of extracellular matrix-related genes in human periodontal fibroblasts induced by stimulation for a prolonged period with lipopolysaccharide derived from Porphyromonas gingivalis. J Periodontal Res 51:508-517.

10. Starr R, Willson TA, Viney EM, Murray LJ, Rayner JR, Jenkins BJ, Gonda TJ, Alexander WS, Metcalf D, Nicola NA, Hilton DJ (1997). A family of cytokine-inducible inhibitors of signalling. Nature 387: 917–21.

(26)

26

11. Nobori T, Miura K, Wu DJ, Lois A, Takabayashi K, Carson DA (1994). Deletions of the cyclin-dependent kinase-4 inhibitor gene in multiple human cancers. Nature 368: 753–6.

12. Overall CM, Tam E, McQuibban GA, Morrison C, Wallon UM, Bigg HF, King AE, Roberts CR (2000). Domain interactions in the gelatinase A.TIMP-2.MT1-MMP activation complex. The ectodomain of the 44-kDa form of membrane type-1 matrix metalloproteinase does not modulate gelatinase A activation. J Biol Chem 275: 39497–506.

13. Natoli G (2009). Control of NF-kappaB-dependent transcriptional responses by chromatin organization. Cold Spring Harb Perspect Biol. 1:a000224.

14. Grabiec AM, Tak PP, Reedquist KA (2008). Targeting histone deacetylase activity in rheumatoid arthritis and asthma as prototypes of inflammatory disease: should we keep our HATs on? Arthritis Res Ther. 10:1-13.

15. Seeley JJ, Ghosh S (2016). Molecular mechanisms of innate memory and tolerance to LPS. J Leukoc Biol. 101: 1-11.

16. Beeson PB (1946). Development of tolerance to typhoid bacterial pyrogen and its abolition by reticulo-endothelial blockade. Proc Soc Exp Biol Med. 61:248-50.

17. El Gazzar M, Yoza BK, Hu JY, Cousart SL, McCall CE (2007). Epigenetic silencing of tumor necrosis factor alpha during endotoxin tolerance. J Biol Chem. 282:26857-64.

18. Chen X, El Gazzar M, Yoza BK, McCall CE (2009). The NF-kappaB factor RelB and histone H3 lysine methyltransferase G9a directly interact to generate epigenetic silencing in endotoxin tolerance. J Biol Chem. 284:27857-65.

19. Chan C, Li L, McCall CE, Yoza BK (2005). Endotoxin tolerance disrupts chromatin remodeling and NF-kappaB transactivation at the IL-1beta promoter. J Immunol. 175:461-8.

20. Zhang J, Li H, Yu JP, Wang SE, Ren XB (2012). Role of SOCS1 in tumor progression and therapeutic application. Int J Cancer. 130:1971-80.

21. Planello AC, Singhania R, Kron KJ, Bailey SD, Roulois D, Lupien M, Line SR, de Souza AP, De Carvalho DD (2016). Pre-neoplastic epigenetic disruption of transcriptional enhancers in chronic inflammation. Oncotarget. 7:15772-86.

(27)

27

22. Wojdacz TK, Dobrovic A, Hansen LL (2008). Methylation-sensitive high-resolution melting. Nat Prot. 3:1903-1908.

23. Cudejko C, Wouters K, Fuentes L, Hannou SA, Paquet C, Bantubungi K, Bouchaert E, Vanhoutte J, Fleury S, Remy P, Tailleux A, Chinetti-Gbaguidi G, Dombrowicz D, Staels B, Paumelle R (2011). p16INK4a deficiency promotes IL-4-induced polarization and inhibits proinflammatory signaling in macrophages. Blood. 118:2556-66.

24. Taniguchi K, Kohsaka H, Inoue N, Terada Y, Ito H, Hirokawa K, Miyasaka N (1999). Induction of the p16INK4a senescence gene as a new therapeutic strategy for the treatment of rheumatoid arthritis. Nat Med. 5:760-7.

25. Nasu K, Kohsaka H, Nonomura Y, Terada Y, Ito H, Hirokawa K, Miyasaka N (2000). Adenoviral transfer of cyclin-dependent kinase inhibitor genes suppresses collagen-induced arthritis in mice. J Immunol. 165:7246-52.

26. Murakami Y, Mizoguchi F, Saito T, Miyasaka N, Kohsaka H (2012). p16(INK4a) exerts an anti-inflammatory effect through accelerated IRAK1 degradation in macrophages. J Immunol. 189:5066-72.

27. Choi BY, Choi HS, Ko K, Cho YY, Zhu F, Kang BS, Ermakova SP, Ma WY, Bode AM, Dong Z (2005). The tumor suppressor p16(INK4a) prevents cell transformation through inhibition of c-Jun phosphorylation and AP-1 activity. Nat Struct Mol Biol. 12:699-707.

28. Wolff B, Naumann M (1999). INK4 cell cycle inhibitors direct transcriptional inactivation of NF-kappaB. Oncogene. 18:2663-6.

29. Terayama Y, Matsuura T, Ozaki K (2016). Lack of Correlation between Aberrant p16, RAR-β2, TIMP3, ERCC1, and BRCA1 Protein Expression and Promoter Methylation in Squamous Cell Carcinoma Accompanying Candida albicans-Induced Inflammation. PLoS One. 11:e0159090.

30. Chernov AV, Sounni NE, Remacle AG, Strongin AY (2009). Epigenetic control of the invasion-promoting MT1-MMP/MMP-2/TIMP-2 axis in cancer cells. J Biol Chem. 284:12727-34.

(28)

28

31. Bourboulia D, Stetler-Stevenson WG (2010). Matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs): Positive and negative regulators in tumor cell adhesion. Semin Cancer Biol. 20:161-8.

(29)

29

3 CONCLUSÃO

No presente estudo, nós concluímos que fibroblastos gengivais humanos

estimulados por LPS em um curto período não apresentaram alterações de metilação

nos promotores dos genes P16, TIMP2 e SOCS1. Outros períodos de ensaio devem

ser realizados a fim de verificar se o LPS promoverá alterações na metilação dos

promotores desses genes, ou em outras regiões desses genes. Portanto, temos

inúmeras hipóteses que ainda devem ser ensaiadas, a fim de aumentar o nosso

conhecimento sobre os efeitos do LPS na promoção de alterações epigenéticas.

(30)

30

REFERÊNCIAS

*

Abiko Y, Uehara O, Fukumoto S, Ohta T. Epigenetics of oral infection and inflammatory

diseases – DNA methylation changes in infections and inflammation diseases. J Oral

Biosci. 2014 Nov.56(4)105-9.

Andia DC, Planello AC, Portinho D, da Silva RA, Salmon CR, Sallum EA, et al. DNA

methylation analysis of SOCS1, SOCS3, and LINE-1 in microdissected gingival tissue.

Clin Oral Investig. 2015 Dec;19(9):2337-44. doi: 10.1007/s00784-015-1460-1.

Calmon MF, Colombo J, Carvalho F, Souza FP, Filho JF, Fukuyama EE, et al.

Methylation profile of genes CDKN2A (p14 and p16), DAPK1, CDH1, and ADAM23 in

head and neck cancer. Cancer Genet Cytogenet. 2007 Feb;173(1):31-7.

Cody DT 2nd, Huang Y, Darby CJ, Johnson GK, Domann FE. Differential DNA

methylation of the p16 INK4A/CDKN2A promoter in human oral cancer cells and

normal human oral keratinocytes. Oral Oncol. 1999 Sep;35(5):516-22.

Conceição AL, Babeto E, Candido NM, Franco FC, de Campos Zuccari DA, Bonilha

JL, et al. Differential Expression of ADAM23, CDKN2A (P16), MMP14 and VIM

Associated with Giant Cell Tumor of Bone. J Cancer. 2015 May 23;6(7):593-603. doi:

10.7150/jca.11238.

Deaton AM, Bird A. CpG islands and the regulation of transcription. Genes Dev. 2011

May 15;25(10):1010-22. doi: 10.1101/gad.2037511.

De Souza AP, Planello AC, Marques MR, De Carvalho DD, Line SR. High-throughput

DNA analysis shows the importance of methylation in the control of imune inflammatory

gene transcription in chronic periodontitis. Clin Epigenetics. 2014 Aug 12;6(1):15. doi:

10.1186/1868-7083-6-15.

De Souza AP, Trevilatto PC, Scarel-Caminaga RM, de Brito RB Jr, Barros SP, Line

SR. Analysis of the MMP-9 (C-1562 T) and TIMP-2 (G-418C) gene promoter

polymorphisms in patients with chronic periodontitis. J Clin Periodontol. 2005

Feb;32(2):207-11.

* De acordo com as normas da UNICAMP/FOP, baseadas na padronização do International Committee of Medical Journal Editors - Vancouver Group. Abreviatura dos periódicos em conformidade com o PubMed.

(31)

31

Ding PH, Jin LJ. The role of lipopolysaccharide-binding protein in innate immunity: a

revisit and its relevance to oral/periodontal health. J Periodontal Res. 2014

Feb;49(1):1-9. doi: 10.1111/jre.12081.

Egger G, Liang G, Aparicio A, Jones PA. Epigenetics in human disease and prospects

for epigenetic therapy. Nature. 2004 May 27;429(6990):457-63. Ha PK, Califano JA.

Promoter methylation and inactivation of tumour-suppressor genes in oral squamous

cell carcinoma. Lancet Oncol. 2006 Jan;7(1):77-82.

Johansson A, Bergenholtz A, Holm SE. Strong cytotoxicity to human gingival

fibroblasts by Porphyromonas gingivalis ATCC 33277. J Periodontal Res. 1996

Oct;31(7):477-82.

Kubota T, Itagaki M, Hoshino C, Nagata M, Morozumi T, Kobayashi T, et al. Altered

gene expression levels of matrix metalloproteinases and their inhibitors in

periodontitis-affected

gingival

tissue.

J

Periodontol.

2008

Jan;79(1):166-73.

doi:

10.1902/jop.2008.070159 .

Larsson L, Castilho RM, Giannobile WV. Epigenetics and its role in periodontal

diseases: a state-of-the-art review. J Periodontol. 2015 Apr;86(4):556-68. doi:

10.1902/jop.2014.140559.

Palm E, Demirel I, Bengtsson T, Khalaf H. The role of toll-like and protease-activated

receptors in the expression of cytokines by gingival fibroblasts stimulated with the

periodontal pathogen Porphyromonas gingivalis. Cytokine. 2015 Dec;76(2):424-32.

doi: 10.1016/j.cyto.2015.08.263.

Parashar G, Capalash N. Expression of the TIMP2 gene is not regulated by promoter

hypermethylation in the Caski cell line. Oncol Lett. 2012 May;3(5):1079-1082.

Planello AC. Epigenetics Analysis of SOCS1 Gene and Its Association with Chronic

Periodontitis [tese]. Piracicaba: Faculdade de Odontologia de Piracicaba,

Universidade Estadual de Campinas; 2014.

Rettori MM, de Carvalho AC, Longo AL, de Oliveira CZ, Kowalski LP, Carvalho AL, et

al. TIMP3 and CCNA1 hypermethylation in HNSCC is associated with na increased

incidence of second primary tumors. J Transl Med. 2013 Dec 20;11:316. doi:

10.1186/1479-5876-11-316.

(32)

32

Tabeta K, Yamazaki K, Akashi S, Miyake K, Kumada H, Umemoto T, et al. Toll-like

receptors confer responsiveness to lipopolysaccharide from Porphyromonas gingivalis

in human gingival fibroblasts. Infect Immun. 2000 Jun;68(6):3731-5.

Takai R, Uehara O, Harada F, Utsunomiya M, Chujo T, Yoshida K, et al. DNA

hypermethylation of extracellular matrix-related genes in human periodontal fibroblasts

induced by stimulation for a prolonged period with lipopolysaccharide derived from

Porphyromonas gingivalis. J Periodontal Res. 2016 Aug;51(4):508-17. doi:

10.1111/jre.12330.

(33)

33

Referências

Documentos relacionados

The RT-qPCR analysis showed increased expression of BCL2L11 in ameloblastomas compared with dental follicles, in agreement with the methylation analysis results, while there was

Apesar do alto consumo enérgético durante o processo de secagem da biomassa, a grande disponibiladade da matéria prima usada como biossorvente, o baixo custo durante

Two tumor suppressor genes, CDKN2B/ p15 and CDKN2A/ p16 (cyclin-dependent kinase inhibitors 2B and 2A, respectively), are lo- cated in the 9p21 region and can be targets

In order to validate the importance of the methylation profile in the regulation of gene expression, the transcription of tumor suppressor genes was performed in two

As etapas de criação e expressão dos modelos e analogias proporcionaram à professora, às pesquisadoras e aos pares acesso às ideias que embasavam o entendimento dos estudantes sobre

Note: Cumulative change in the price and composition effects of the between-groups variance of the logarithm of real hourly wages (2005 prices) with respect to 1995.

As corre- laões observadas entre a variação nas freqüências alélicas das isozimas de indivíduos portadores de diferentes padrões de asas e que foram evidencia- das pelo

We studied p16 INK4a expression in DPCs and inhibited p16 INK4a expression in DPCs by adenovirus-mediated RNA interfer- ence to explore the possible mechanisms of cultured human