Sirtuin 1 and oral cancer (Review)

  • Authors:
    • Shajedul Islam
    • Yoshihiro Abiko
    • Osamu Uehara
    • Itsuo Chiba
  • View Affiliations

  • Published online on: November 16, 2018     https://doi.org/10.3892/ol.2018.9722
  • Pages: 729-738
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The sirtuins (SIRTs) are a family of highly conserved histone deacetylases (HDACs) consisting of seven members (SIRT1‑SIRT7). Over the past few decades, SIRT1 has been the most extensively studied and garnered tremendous attention in the scientific community due to its emerging role in cancer biology. However, its biological role in the regulation of oral cancer is not yet fully understood. Owing to contradictory findings regarding the role of SIRT1 in oral cancer, debate about it continues. The present study discusses the biological roles and potential therapeutic implications of SIRT1 in precancerous oral lesions and oral cancer.

Introduction

The sirtuin family proteins (SIRT) are class III histone deacetylases (HDACs) comprised of seven members (SIRT1-7). Sirtuin proteins are widely expressed in normal tissues and reported to be involved in several biological processes (14) (Fig. 1). SIRT1 was the first family member to be discovered and is still the most studied. Its biological role in cancer has been studied extensively, yet there are conflicting results regarding the association between the two as SIRT1 is known to suppress or promote cancer depending on its cellular content or type (24). The expression level of SIRT1 has been shown to play an important role in the pathogenesis of oral cancer (58), the sixth most frequent cancer worldwide, with oral squamous cell carcinoma (OSCC) being, by far, the commonest single entity, accounting for about 90% of all malignancies in the oral cavity and posing a major public health problem in many Asian countries (9). The etiologies of oral cancer include betel quid chewing, smoking, alcohol consumption, genetic predisposition, and viruses, including human papillomavirus (HPV) (9,10). The overall 5-year survival rates for patients with OSCC (ranging from 34 to 62.9%) have not significantly improved for decades in spite of advances in the field of oncology. These findings underscore the importance of encouraging new areas of research on factors that modify oral cancer and therapeutic targets to treat it. The purpose of this review is to summarize the findings of recent publications on SIRT1 with regard to oral cancer and to discuss its importance as a possible therapeutic agent. To the best of our knowledge, this is the first review evaluating the biological role of SIRT1 in the modulation of oral cancer.

Overview of SIRT1 functions

SIRT1 is a nicotinamide adenine dinucleotide (NAD+)-dependent class III HDAC protein. High levels of NAD+ induce SIRT1 activity, whereas high NADH levels inhibit its function. Due to its localization, SIRT1 is capable of deacetylating lysine residues on nuclear and cytoplasmic proteins, which is thought to affect their stability, transcriptional activity, and translocation (1). Over the years, SIRT1 has been a major point of focus in biomedical research due to its diversified roles in various pathophysiological states (24). It plays contradictory roles in cancer and has an elaborate network of interactions that are directly involved in tumour biology (1123) (Table I).

Table I.

Overview of SIRT1 functions.

Table I.

Overview of SIRT1 functions.

Author, yearFunctionSubstratesEffects of SIRT1(Refs.)
Peters et al, 2001Genomic stabilityH3K9Ac, H4K16Ac,Modification of chromatin through the(11)
Vaquero et al, 2004 H1K26Ac, Suv39h1,formation of heterochromatin structure(12)
Vaquero et al, 2007 TERTand positive regulation of telomere length(13)
Palacios et al, 2010 (14)
Wang et al, 2008DNA repairγ-H2AX, BRCA1,Induction of HR and NHEJ-mediated DNA(4)
Sawada et al, 2003 Rad51, MRN complex,repair(17)
Yuan et al, 2007 Ku70, BaxInduction of formation of NBSI (nibrin) foci(15)
Jeong et al, 2007 and direct recruitment to DNA damage sites(16)
Induction of Ku70-dependent DNA repair signalling
Brunet et al, 2004Stress responsep53, Foxo3a, p300,Inhibition of apoptosis and promote DNA(18)
Motta et al, 2004 set7/9, MnSODrepair by deacetylase p53(20)
Kobayashi et al, 2005 Promotion of cell survival during oxidative(19)
Chua et al, 2005 stress by inducing DNA repair in cooperation(21)
Yi et al, 2010 with Foxo1(22)
Peng et al, 2011DNA methylationDNMT1, DNMT3bAlteration of DNMTs enzymatic activity by deacetylating different lysines(23)

[i] H3K9ac, histone H3 lysine 9 acetylation; H4K16ac, histone H4 lysine 16 acetylation; H1K26ac, histone H1 lysine 26 acetylation; Suv39h1, suppressor of variegation 3–9 homolog 1; TERT, telomerase reverse transcriptase; γ-H2AX, phosphorylated form of variant histone H2AX; BRCA1, breast cancer type 1 susceptibility protein; Rad51, DNA repair protein Rad51 homolog 1; MRN complex, Mre11-Rad50-Nbs1 complex; Ku70, X-ray repair cross-complementing 6; Bax, BCL2 associated × protein; HR, homologous recombination; NHEJ, nonhomologous end joining; Foxo3a, forkhead box o3 α; p300, E1A binding protein p300; Set7/9, Set domain containing lysine methyltransferase 7; MnSOD, manganese-containing superoxide dismutase; Foxo1, forkhead box protein o1; DNMT1, DNA methyltransferase 1; DNMT3b, DNA methyltransferase 3 β.

SIRT1 and tumour development

SIRT1 plays a key role in epigenetic regulation of gene expression by changing the structure of chromatin. It has been reported to deacetylate both histone and non-histone proteins. Deacetylation of histones by SIRT1 has been shown to induce chromatin condensation, whereas acetylation by histone acetyltransferases (HATs) causes chromatin decondensation. This balance is crucial for normal cellular functions, and any disturbance of it will be related to cancer (24). SIRT1-mediated deacetylation of non-histone proteins has been suggested to be more important in cancer than histones (24,25). In tumour biology, SIRT1 seems to play contradictory roles and deregulation of SIRT1 expression has frequently been reported in many human malignancies (4,2638) (Table II).

Table II.

SIRT1 and tumour development.

Table II.

SIRT1 and tumour development.

A, Tumor promoter

Author, yearEffects of SIRT1Types of tumour(Refs.)
Huffman et al, 2007Inhibition of HIC-1 expression via epigenetic modificationProstate cancer(26)
Chen et al, 2012Induction of expression of c-Myc, EMT markers; therebyHCC(27)
Hao et al, 2014increasing resistance to the chemotherapeutic agent (28)
Chen et al, 2014Induction of genomic instability by maintaining the characteristics of CSCsCRC(29)
Zhao et al, 2013Induction of chemoresistance via dysregulation of HIC-1Pancreatic carcinoma(30)
Stunkel et al, 2007Survival and proliferation of cancer cellsColon carcinoma(31)
Ford et al, 2005Induction of growth and survivalCervical cancer(32)
Induction of c-Myc activityThyroid carcinoma
He et al, 2016Induction of metastatic phenotype of cancerous cellsESCC(33)
Hida et al, 2007Dysregulation of the RB1 pathwayNMSC(34)
Bradbury et al, 2005Induction of drug resistanceAML(35)

B, Tumor suppressor

Author, yearEffects of SIRT1Types of tumour(Refs.)

Wang et al, 2008Maintenance of genomic integrity & activation ofGlioblastoma(4)
DNA repair mechanismsOvarian cancer
Hepatic cancer
Breast cancer
Bladder cancer
Prostate cancer
Jung et al, 2012Maintenance of genetic integrity and inhibition of β catenin,CRC(36)
Jang et al, 2013survivin expression (37)
Firestein et al, 2008Deacetylation and suppression of β-cateninColon cancer(38)

[i] HIC1, hypermethylated in cancer 1; AFP, α-fetoprotein; TERT, telomerase reverse transcriptase; EMT, epithelial mesenchymal transition; CSCs, cancer stem cells; RB1, retinoblastoma 1; HCC, hepatocellular carcinoma; CRC, colorectal carcinoma; ESCC, esophageal squamous cell carcinoma; NMSC, nonmelanoma skin cancer; AML, acute myeloid leukemia; β catenin, catenin β 1.

On the one hand, SIRT1 has been reported to deacetylate and inactivate p53, thereby allowing cells to bypass p53-induced apoptosis (22,39). Similarly, during cellular oxidative stress, SIRT1-mediated deacetylation of forkhead box O3 alpha (Foxo3a) has been shown to induce cell survival rather than apoptosis (4,22,39). This is good for normal cells to prolong their lifespan but in tumour cells, this effect is not at all desirable since it aggravates tumour growth. Meanwhile, overexpression of SIRT1 has been demonstrated to induce angiogenesis in cancer cells via increases in the expression of angiogenic growth factors (40). Taken together, these studies suggest that SIRT1 may bring more nutrition to cancer cells and lead to their enhanced growth, proliferation, and survival.

As opposed to what occurs during cellular oxidative stress, SIRT1 has been shown to induce mitochondrial translocation of p53, leading to enhanced p53-independent mitochondrial apoptosis (22). The DNA repair mechanisms and genomic stability functions of SIRT1 imply a protective effect against cancer (reviewed in 2–4). Therefore, the question arises as to whether SIRT1 acts primarily as an oncogene or a tumour suppressor. It is, however, strongly evident that SIRT1 is a critical regulator in the pathogenesis of tumours. To clarify the contradictory roles of SIRT1 in tumorigenesis, further studies are necessary.

Modulation of SIRT1 in oral cancer

The regulatory role of SIRT1 in oral cancer is vigorously debated owing to the belief that it can have both tumorigenic and non-tumorigenic roles (58) (Table III). Altered levels of SIRT1 expression have a significant impact on the pathophysiology of oral cancer. Downregulation of SIRT1 expression is correlated with the metastatic phenotype, whereas upregulation of this protein results in opposite effects (58). It has been reported that stable expression of SIRT1 aids in maintaining epithelial integrity by inducing the expression of epithelial-cadherin (E-cadherin), and this contributes to the prevention of both invasion and metastasis in oral cancer (57). Conflicting data have also been reported for prostate carcinoma. It has been reported that SIRT1 mediates deacetylation of histone H3, causing transcriptional repression of E-cadherin and leading to invasion and metastasis (41). However, SIRT1 has been demonstrated to inhibit transforming growth factor-beta (TGF-β)-mediated malignant transformation, invasion and metastasis in oral cancer (5). TGF-β is a growth factor and its overexpression has been frequently reported to be involved in precancerous oral lesions, leading to oral cancer (42,43). Increased expression of TGF-β has been shown to enhance malignant transformation, invasion and metastasis in oral epithelial cells by inducing its downstream targets (56,43). Similarly, overexpressed TGF-β acts on fibroblasts and has been reported to increase myofibroblastic transdifferentiation (42,43). Myofibroblasts are the major source for collagen synthesis in the extracellular matrix (ECM) of connective tissues, and continuous increases in the deposition of collagen lead to the pathogenesis of oral submucous fibrosis (OSF), a precancerous condition (42,43) (Fig. 2). The malignant transformation rate in patients with OSF ranges from 7–13% (10). SIRT1 has been shown to induce transcriptional suppression of TGF-β-mediated downstream targets in fibroblasts and to prevent malignant transformation (44). Based on these observations, SIRT1 may have the ability to prevent malignant transformation, invasion, and metastasis.

Figure 2.

Possible regulatory mechanism of SIRT1 in oral cancer induced by betel quid chewing: (A, a) TGF-β is a growth factor and its overexpression has been frequently reported in precancerous oral lesions leading to oral cancer, The TGF-β ligand binds to its receptor and induces the phosphorylation of smad2/3; (A, b) phosphorylated smad2/3 binds with acetylated smad4 and forms a complex known as the SMAD complex; (A, c, d) this SMAD complex translocates to the nucleus and binds with its co-activator CBP/p300, a histone acetyltransferase, and induces TGF-β-mediated invasion and metastasis. (A, e) SIRT1 can inhibit phosphorylation of smad2/3 and remove acetyl groups from smad4 protein. These effects help to prevent the formation of the SMAD complex; (A, f) at the nucleus, SIRT1 binds in the promoter region of TGF-β, inhibits CBP/p300-mediated acetylation via the deacetylation mechanism and results in transcriptional suppression of TGF-β-mediated malignant transformation, invasion, and metastasis in the oral mucosa of betel quid chewers. (B, a) Arecoline is the major alkaloid in betel quid, and is known to downregulate SIRT1 expression in oral epithelial cells, leading to enhanced TGF-β-mediated invasion and metastasis; (B, b) arecoline-mediated downregulation of SIRT1, followed by upregulation of TGF-β, acts on fibroblasts and enhances the pathogenesis of OSF, a precancerous condition. SIRT1, sirtuin 1; TGF-β, transforming growth factor beta; OSF, oral submucous fibrosis.

Table III.

Regulatory role of SIRT1 in oral cancer.

Table III.

Regulatory role of SIRT1 in oral cancer.

Author, yearType of functionRole of SIRT1(Refs.)
Chen et al, 2014Tumour suppressorMaintenance of epithelial polarity by increases the expression of(5)
Kang et al, 2017 E-cadherin(6)
Murofushi et al, 2017 Suppression of expression of N-cadherin and vimentin(7)
Downregulation of genes involved in migration and invasion, such as
CSK2A2, FRA1, ACTB, and SLUG
Downregulation of TGF-β downstream targets
Induction of p21 and G1/S phase cell cycle arrest
Xiong et al, 2011Tumour promoterInduction of cisplatin resistance by increasing the expression of annexin A4, stathmin, SOD2 and thioredoxin(8)
Induction of growth and survival of cancerous cells

[i] SOD2, superoxide dismutase 2; CSK2A2, casein kinase II subunit α; FRA1, fos related antigen 1; ACTB, beta-actin; SLUG, snail family transcriptional repressor 2, TGF-β, transforming growth factor β.

Recently, our group evaluated a significant association between arecoline and the expression of SIRT1 in oral epithelial cells. Arecoline, the major alkaloid in betel quid, has been reported to be involved in the pathogenesis of oral cancer by facilitating the cellular transformation and transcriptional repression of tumour suppressor genes (TSGs) (10,45,46). We discovered that arecoline significantly induced DNA hypermethylation, followed by downregulation of SIRT1 expression in oral epithelial cells. The frequency of DNA hypermethylation was found to be associated with precancerous oral lesions (data not shown). Our data suggest that arecoline-mediated downregulation of SIRT1 expression may be involved in the initial stage of transformation of normal cells to oral cancer and the development of precancerous oral lesions induced by betel quid chewing. Our results fit well with the observation of an association of SIRT1 and TGF-β, wherein arecoline-mediated downregulation of SIRT1 expression in oral epithelial cells fails to prevent TGF-β-induced malignant transformation in the oral mucosa of betel quid chewers. Taken together, these results suggest that SIRT1 could serve as a tumour suppressor in oral cancer. However, it remains unclear how it directly affects this process.

Conversely, despite evidence of the tumour-suppressing effects of SIRT1, some studies have demonstrated the promoting effects of this protein (8). Upregulation of annexin A4 has been shown to promote the progression and chemoresistance of numerous tumours (47). Overexpression of SIRT1 has been reported to induce cisplatin resistance in oral cancer by elevating the level of annexin A4 (8), and chemical inhibitors of SIRT1 significantly abolish this action (8). Hypoxia within the tumour microenvironment has a well-documented role to promote tumorigenesis. Recent reports investigating the role of SIRT1 under hypoxia have demonstrated that it promotes tumorigenesis via incorporation with hypoxia-inducible factor-1 alpha (HIF-1α) (48). Based on these findings, SIRT1 might have a significant tumour-inducing effect. Thus, further studies are needed to clarify this issue and evaluate new therapeutic approaches.

Therapeutic potential of HDACs in malignancy

Activators and inhibitors of HDACs have been developed in recent years and, to date, three histone deacetylase inhibitors (HDACis) have received United States Food and Drug Administration approval for therapeutic use (4963) (Table IV).

Table IV.

HDAC inhibitors/activators in the treatment of various tumours.

Table IV.

HDAC inhibitors/activators in the treatment of various tumours.

Author, yearHDAC inhibitors (class I, II, IV)Clinical status(Refs.)
Ceccacci et al, 2016VorinostatApproved for cutaneous T-cell lymphoma(49)
PanobinostatApproved for multiple myeloma
RomidepsinApproved for peripheral T-cell lymphoma
HDAC inhibitors (Class III)
Ceccacci et al, 2016NAM (SIRT 1–6)Phase III clinical trial-laryngeal cancer(49)
Hu et al, 2014 Leukemia, prostate carcinoma(50)
Jin et al, 2015Tenovin-6 (SIRT 1–2)Chronic myeloblastic leukemia(51)
Dai et al, 2016 Acute myeloblastic leukemia(52)
Eckschlager et al, 2017BDF4 (1/2a/-b/2d) (SIRT 1–2)Colon carcinoma, glioblastoma(53)
Eckschlager et al, 2017Sirtinol (SIRT 1–2)Breast, lung and prostate carcinomas(53)
Ota et al, 2006 (54)
Eckschlager et al, 2017EX-527 (SIRT 1)Leukaemia(53)
Süssmuth et al, 2015 Phase I/II clinical trials-Huntington disease(56)
Bhalla et al, 2017 (55)
Heltweg et al, 2006CMB (SIRT 1–2)Burkitt lymphoma(57)
Kalle et al, 2010JGB-1741 (SIRT 1)Breast cancer(58)
Eckschlager et al, 2017AC-93253 (SIRT 1–3)Prostate, pancreas and lung carcinoma(53)
Lai et al, 2017 (59)
Lara et al, 2009Salermide (SIRT 1–2)Colorectal carcinoma(61)
Rotili et al, 2012 (60)
HDAC activators (Class III)
Jiang et al, 2017Resveratrol (SIRT1)Leukemia, prostate carcinoma and skin cancer(62)
Chauhan et al, 2011SRT1720 (SIRT1)Multiple myeloma(63)

[i] Vorinostat, suberanilohydroxamic acid; NAM, nicotinamide; BDF4, benzodeazoxaflavins; Sirtinol, sir two inhibitor naphthol; CMB, cambinol.

The use of HDACis in combination with conventional chemotherapeutic agents has already been reported to be a promising strategy against oral cancer (6471) (Table V). The class III HDAC SIRT1 is considered to be both a promoter and suppressor. Inhibitors of SIRT1 have attracted interest and been found to induce apoptosis in various cancer cell lines. Similarly, activators of SIRT1 have been shown to possess the ability to prevent numerous cancers, including leukaemia, skin cancer, prostate cancer and multiple myeloma (25,34,62,63,72). Therefore, the conflicting data reported in the literature support the use of both activators and inhibitors of SIRT1 as strategies for cancer therapy; hence, care needs to be taken regarding the cytotoxicity and the dose of this protein when it is administered as a therapeutic agent.

Table V.

HDACis in oral squamous cell carcinoma treatment.

Table V.

HDACis in oral squamous cell carcinoma treatment.

Author, yearHDACisEffectsMechanism(Refs.)
Suzuki et al, 2009VorinostatInduction of chemosensitizationMaintainence of epithelial polarity(66)
Bruzzese et al, 2011 to cisplatin, thereby increasingInduction of p53, CytC, and(65)
Tasoulas et al, 2015 apoptosis and inhibiting survival, proliferation, migration, and invasioncaspase-3 dependent apoptosis(64)
Tasoulas et al, 2015Trichostatin AInduction of chemosensitizationIncreased cathepsin activity and(64)
Eriksson et al, 2013 to cisplatinreduced LAMP-2 level(67)
Tasoulas et al, 2015BenzamideIncrease in radiation-induced cellInduction of G1/S phase cell(64)
Sato et al, 2006 cycle arrest and apoptosiscycle arrest(68)
Tasoulas et al, 2015DepsipeptideInduction of apoptosis and cellInduction of p21 and G2/M arrest(64)
Sato et al, 2006 cycle arrest (68)
Tasoulas et al, 2015Valproic acid +5Inhibition of proliferation andInduction of H3 and H4 acetylation(64)
Shoji et al, 2012azacytidineinduces radiosensitizationand decrease of Rad51 expression(69)
Gan et al, 2012 (70)
Gong et al, 2010Sodium butyrateSuppression of growth and proliferationInduction of p27 expression and caspase-dependent apoptosis(71)
Downregulation of Akt pathway

[i] Vorinostat, suberanilohydroxamic acid; CytC, Cytochrome c; LAMP-2, lysosomal-associated membrane protein 2; Rad51, DNA repair protein Rad51 homolog 1; Akt, protein kinase B; HDACis, histone deacetylase inhibitors.

Future research perspectives and possible therapeutic applications of SIRT1 in oral cancer

In spite of being involved in various physiological and pathological processes, the effects of altered SIRT1 expression in oral cancer are inconsistent. Reactive oxygen species (ROSs) are the strongest risk factor associated with the pathogenesis of oral cancer (10). Habits such as betel quid chewing have been reported to induce generation of ROSs in the oral epithelium and lead to genetic instability by damaging DNA and other macromolecules (10,45). SIRT1 has been shown to increase the synthesis of antioxidant enzymes such as glutathione and superoxide dismutase 2 and prevent ROS-mediated genomic alterations (73). Consistent with this premise, it is hypothesized that SIRT1 may play a significant role in inhibiting the synthesis of ROSs and prevent DNA and macromolecule damage in the oral mucosa of betel quid chewers. Further studies are thus warranted to evaluate the regulatory mechanism of SIRT1 in ROS generation in oral epithelial cells. Moreover, as demonstrated in some of the studies cited above, SIRT1 may prevent TGF-β-mediated invasion and metastasis in oral cancer (5,42,43). TGF-β is a growth factor and remains hidden in its inactive state in the ECM (74). αvβ6 integrin has been shown to facilitate the activation of the TGF-β downstream pathway by allowing it to bind with its receptors (74) (Fig. 3).

αv is the only integrin that can form a dimer with β6, and β6 integrin is normally not expressed in healthy adult epithelial cells, whereas its overexpression is associated with preneoplastic epithelial phenotypes. As a result, formation of a stable αvβ6 integrin dimer is related to TGF-β-mediated invasion and metastasis in the oral epithelium (75). CREB-binding protein (CBP) is a HAT and it has been reported that CBP mediates acetylation in the promoter region of β6 integrin and induces its expression in preneoplastic epithelium. This induced expression of β6 integrin enhances the formation of αvβ6 dimers and results in invasion and metastasis in oral cancer (76). Since SIRT1 is a deacetylase, it is hypothesized that it may induce transcriptional suppression of β6 integrin via deacetylation in the promoter region and prevent invasion and metastasis in oral cancer. Thus, further studies are warranted to evaluate the use of SIRT1-based therapeutic approaches in oral cancer.

Concluding remarks

Based on results of the studies referred to above and our current data, it is hypothesized that SIRT1 may play a significant tumour-suppressive role in oral cancer. Future studies will undoubtedly pinpoint the molecular mechanisms via which SIRT1 influences oral carcinogenesis and identify efficacious SIRT1 activators for the prevention or treatment of precancerous oral lesions that can lead to oral cancer.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets generated and/or analyzed during the present study are not publicly available due to the data containing information that may compromise the consent of the participants but are available from the corresponding author on reasonable request.

Authors' contributions

SI and YA conducted the literature review and wrote the manuscript. OU and IC contributed to the study design and the writing of the manuscript, and made corrections. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Carafa V, Rotili D, Forgione M, Cuomo F, Serretiello E, Hailu GS, Jarho E, Lahtela-Kakkonen M, Mai A and Altucci L: Sirtuin functions and modulation: From chemistry to the clinic. Clin Epigenetics. 8:612016. View Article : Google Scholar : PubMed/NCBI

2 

Deng CX: SIRT1, is it a tumor promoter or tumor suppressor? Int J Biol Sci. 5:147–152. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Bosch-Presegué L and Vaquero A: The dual role of sirtuins in cancer. Genes Cancer. 2:648–662. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Wang RH, Sengupta K, Li C, Kim HS, Cao L, Xiao C, Kim S, Xu X, Zheng Y, Chilton B, et al: Impaired DNA damage response, genome instability, and tumorigenesis in SIRT1 mutant mice. Cancer Cell. 14:312–323. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Chen IC, Chiang WF, Huang HH, Chen PF, Shen YY and Chiang HC: Role of SIRT1 in regulation of epithelial-to-mesenchymal transition in oral squamous cell carcinoma metastasis. Mol Cancer. 13:2542014. View Article : Google Scholar : PubMed/NCBI

6 

Kang YY, Sun FL, Zhang Y and Wang Z: SIRT1 acts as a potential tumor suppressor in oral squamous cell carcinoma. J Chin Med Assoc. 81:416–422. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Murofushi T, Tsuda H, Mikami Y, Yamaguchi Y and Suzuki N: CAY10591, a SIRT1 activator, suppresses cell growth, invasion, and migration in gingival epithelial carcinoma cells. J Oral Sci. 59:415–423. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Xiong P, Li YX, Tang YT and Chen HG: Proteomic analyses of Sirt1-mediated cisplatin resistance in OSCC cell line. Protein J. 30:499–508. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Warnakulasuriya S: Global epidemiology of oral and oropharyngeal cancer. Oral Oncol. 45:309–316. 2009. View Article : Google Scholar : PubMed/NCBI

10 

IARC Working Group on the Evaluation of Carcinogenic Risks to Humans: Betel-quid and areca-nut chewing and some areca-nut-derived nitrosamines. IARC Monogr Eval Carcinog Risks Hum. 85:1–334. 2004.PubMed/NCBI

11 

Peters AH, O'Carroll D, Scherthan H, Mechtler K, Sauer S, Schöfer C, Weipoltshammer K, Pagani M, Lachner M, Kohlmaier A, et al: Loss of the Suv39h histone methyltransferases impairs mammalian heterochromatin and genome stability. Cell. 107:323–337. 2001. View Article : Google Scholar : PubMed/NCBI

12 

Vaquero A, Scher M, Lee D, Erdjument-Bromage H, Tempst P and Reinberg D: Human SirT1 interacts with histone H1 and promotes formation of facultative heterochromatin. Mol Cell. 16:93–105. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Vaquero A, Scher M, Erdjument-Bromage H, Tempst P, Serrano L and Reinberg D: SIRT1 regulates the histone methyl-transferase SUV39H1 during heterochromatin formation. Nature. 450:440–444. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Palacios JA, Herranz D, De Bonis ML, Velasco S, Serrano M and Blasco MA: SIRT1 contributes to telomere maintenance and augments global homologous recombination. J Cell Biol. 191:1299–1313. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Yuan Z, Zhang X, Sengupta N, Lane WS and Seto E: SIRT1 regulates the function of the Nijmegen breakage syndrome protein. Mol Cell. 27:149–162. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Jeong J, Juhn K, Lee H, Kim SH, Min BH, Lee KM, Cho MH, Park GH and Lee KH: SIRT1 promotes DNA repair activity and deacetylation of Ku70. Exp Mol Med. 39:8–13. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Sawada M, Sun W, Hayes P, Leskov K, Boothman DA and Matsuyama S: Ku70 suppresses the apoptotic translocation of Bax to mitochondria. Nat Cell Biol. 5:320–329. 2003. View Article : Google Scholar : PubMed/NCBI

18 

Brunet A, Sweeney LB, Sturgill JF, Chua KF, Greer PL, Lin Y, Tran H, Ross SE, Mostoslavsky R, Cohen HY, et al: Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science. 303:2011–2015. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Kobayashi Y, Furukawa-Hibi Y, Chen C, Horio Y, Isobe K, Ikeda K and Motoyama N: SIRT1 is a critical regulator of FOXO-mediated transcription in response to oxidative stress. Int J Mol Med. 16:237–243. 2005.PubMed/NCBI

20 

Motta MC, Divecha N, Lemieux M, Kamel C, Chen D, Gu W, Bultsma Y, McBurney M and Guarente L: Mammalian SIRT1 represses forkhead transcription factors. Cell. 116:551–563. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Chua KF, Mostoslavsky R, Lombard DB, Pang WW, Saito S, Franco S, Kaushal D, Cheng HL, Fischer MR, Stokes N, et al: Mammalian SIRT1 limits replicative life span in response to chronic genotoxic stress. Cell Metab. 2:67–76. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Yi J and Luo J: SIRT1 and p53, effect on cancer, senescence and beyond. Biochim Biophys Acta. 1804:1684–1689. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Peng L, Yuan Z, Ling H, Fukasawa K, Robertson K, Olashaw N, Koomen J, Chen J, Lane WS and Seto E: SIRT1 deacetylates the DNA methyltransferase 1 (DNMT1) protein and alters its activities. Mol Cell Biol. 31:4720–4734. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Glozak MA, Sengupta N, Zhang X and Seto E: Acetylation and deacetylation of non-histone proteins. Gene. 363:15–23. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Glozak MA and Seto E: Histone deacetylases and cancer. Oncogene. 26:5420–5432. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Huffman DM, Grizzle WE, Bamman MM, Kim JS, Eltoum IA, Elgavish A and Nagy TR: SIRT1 is significantly elevated in mouse and human prostate cancer. Cancer Res. 67:6612–6618. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Chen HC, Jeng YM, Yuan RH, Hsu HC and Chen YL: SIRT1 promotes tumorigenesis and resistance to chemotherapy in hepatocellular carcinoma and its expression predicts poor prognosis. Ann Surg Oncol. 19:2011–2019. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Hao C, Zhu PX, Yang X, Han ZP, Jiang JH, Zong C, Zhang XG, Liu WT, Zhao QD, Fan TT, et al: Overexpression of SIRT1 promotes metastasis through an epithelial-mesenchymal transition in hepatocellular carcinoma. BMC Cancer. 14:9782014. View Article : Google Scholar : PubMed/NCBI

29 

Chen X, Sun K, Jiao S, Cai N, Zhao X, Zou H, Xie Y, Wang Z, Zhong M and Wei L: High levels of SIRT1 expression enhance tumorigenesis and associate with a poor prognosis of colorectal carcinoma patients. Sci Rep. 4:74812014. View Article : Google Scholar : PubMed/NCBI

30 

Zhao G, Qin Q, Zhang J, Liu Y, Deng S, Liu L, Wang B, Tian K and Wang C: Hypermethylation of HIC1 promoter and aberrant expression of HIC1/SIRT1 might contribute to the carcinogenesis of pancreatic cancer. Ann Surg Oncol. 20 Suppl 3:S301–S311. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Stunkel W, Peh BK, Tan YC, Nayagam VM, Wang X, Salto-Tellez M, Ni B, Entzeroth M and Wood J: Function of the SIRT1 protein deacetylase in cancer. Biotechnol J. 2:1360–1368. 2007. View Article : Google Scholar : PubMed/NCBI

32 

Ford J, Jiang M and Milner J: Cancer-specific functions of SIRT1 enable human epithelial cancer cell growth and survival. Cancer Res. 65:10457–10463. 2005. View Article : Google Scholar : PubMed/NCBI

33 

He Z, Yi J, Jin L, Pan B, Chen L and Song H: Overexpression of Sirtuin-1 is associated with poor clinical outcome in esophageal squamous cell carcinoma. Tumour Biol. 37:7139–7148. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Hida Y, Kubo Y, Murao K and Arase S: Strong expression of a longevity-related protein, SIRT1, in Bowen's disease. Arch Dermatol Res. 299:103–106. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Bradbury CA, Khanim FL, Hayden R, Bunce CM, White DA, Drayson MT, Craddock C and Turner BM: Histone deacetylases in acute myeloid leukaemia show a distinctive pattern of expression that changes selectively in response to deacetylase inhibitors. Leukemia. 19:1751–1759. 2005. View Article : Google Scholar : PubMed/NCBI

36 

Jung W, Hong KD, Jung WY, Lee E, Shin BK, Kim HK, Kim A and Kim BH: SIRT1 expression is associated with good prognosis in colorectal cancer. Korean J Pathol. 47:332–339. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Jang SH, Min KW, Paik SS and Jang KS: Loss of SIRT1 histone deacetylase expression associates with tumour progression in colorectal adenocarcinoma. J Clin Pathol. 65:735–739. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Firestein R, Blander G, Michan S, Oberdoerffer P, Ogino S, Campbell J, Bhimavarapu A, Luikenhuis S, de Cabo R, Fuchs C, et al: The SIRT1 deacetylase suppresses intestinal tumorigenesis and colon cancer growth. PLoS One. 3:e20202008. View Article : Google Scholar : PubMed/NCBI

39 

Voelter-Mahlknecht S and Mahlknecht U: The sirtuins in the pathogenesis of cancer. Clin Epigenetics. 1:71–83. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Potente M, Ghaeni L, Baldessari D, Mostoslavsky R, Rossig L, Dequiedt F, Haendeler J, Mione M, Dejana E, Alt FW, et al: SIRT1 controls endothelial angiogenic functions during vascular growth. Genes Dev. 21:2644–2658. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Byles V, Zhu L, Lovaas JD, Chmilewski LK, Wang J, Faller DV and Dai Y: SIRT1 induces EMT by cooperating with EMT transcription factors and enhances prostate cancer cell migration and metastasis. Oncogene. 31:4619–4629. 2012. View Article : Google Scholar : PubMed/NCBI

42 

Verrecchia F and Mauviel A: Transforming growth factor-beta and fibrosis. World J Gastroenterol. 13:3056–3062. 2007. View Article : Google Scholar : PubMed/NCBI

43 

Ekanayaka RP and Tilakaratne WM: Oral submucous fibrosis: Review on mechanisms of malignant transformation. Oral Surg Oral Med Oral Pathol Oral Radiol. 122:192–199. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Chang YC, Lin CW, Yu CC, Wang BY, Huang YH, Hsieh YC, Kuo YL and Chang WW: Resveratrol suppresses myofibroblast activity of human buccal mucosal fibroblasts through the epigenetic inhibition of ZEB1 expression. Oncotarget. 7:12137–12149. 2016.PubMed/NCBI

45 

Uehara O, Takimoto K, Morikawa T, Harada F, Takai R, Adhikari BR, Itatsu R, Nakamura T, Yoshida K, Matsuoka H, et al: Upregulated expression of MMP-9 in gingival epithelial cells induced by prolonged stimulation with arecoline. Oncol Lett. 14:1186–1192. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Chiba I, Muthumala M, Yamazaki Y, Uz Zaman A, Iizuka T, Amemiya A, Shibata T, Kashiwazaki H, Sugiura C and Fukuda H: Characteristics of mutations in the p53 gene of oral squamous-cell carcinomas associated with betel-quid chewing in Sri Lanka. Int J Cancer. 77:839–842. 1998. View Article : Google Scholar : PubMed/NCBI

47 

Wei B, Guo C, Liu S and Sun MZ: Annexin A4 and cancer. Clin Chim Acta. 447:72–78. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Laemmle A, Lechleiter A, Roh V, Schwarz C, Portmann S, Furer C, Keogh A, Tschan MP, Candinas D, Vorburger SA and Stroka D: Inhibition of SIRT1 impairs the accumulation and transcriptional activity of HIF-1α protein under hypoxic conditions. PLoS One. 7:e334332012. View Article : Google Scholar : PubMed/NCBI

49 

Ceccacci E and Minucci S: Inhibition of histone deacetylases in cancer therapy: Lessons from leukaemia. Br J Cancer. 114:605–611. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Hu J, Jing H and Lin H: Sirtuin inhibitors as anticancer agents. Future Med Chem. 6:945–966. 2014. View Article : Google Scholar : PubMed/NCBI

51 

Jin Y, Cao Q, Chen C, Du X, Jin B and Pan J: Tenovin-6-mediated inhibition of SIRT1/2 induces apoptosis in acute lymphoblastic leukemia (ALL) cells and eliminates ALL stem/progenitor cells. BMC Cancer. 15:2262015. View Article : Google Scholar : PubMed/NCBI

52 

Dai W, Zhou J, Jin B and Pan J: Class III-specific HDAC inhibitor Tenovin-6 induces apoptosis, suppresses migration and eliminates cancer stem cells in uveal melanoma. Sci Rep. 6:226222016. View Article : Google Scholar : PubMed/NCBI

53 

Eckschlager T, Plch J, Stiborova M and Hrabeta J: Histone deacetylase inhibitors as anticancer drugs. Int J Mol Sci. 18(pii): E14142017. View Article : Google Scholar : PubMed/NCBI

54 

Ota H, Tokunaga E, Chang K, Hikasa M, Iijima K, Eto M, Kozaki K, Akishita M, Ouchi Y and Kaneki M: Sirt1 inhibitor, Sirtinol, induces senescence-like growth arrest with attenuated Ras-MAPK signaling in human cancer cells. Oncogene. 25:176–185. 2006. View Article : Google Scholar : PubMed/NCBI

55 

Bhalla S and Gordon LI: Functional characterization of NAD dependent de-acetylases SIRT1 and SIRT2 in B-cell chronic lymphocytic leukemia (CLL). Cancer Biol Ther. 17:300–309. 2016. View Article : Google Scholar : PubMed/NCBI

56 

Süssmuth SD, Haider S, Landwehrmeyer GB, Farmer R, Frost C, Tripepi G, Andersen CA, Di Bacco M, Lamanna C, Diodato E, et al: An exploratory double-blind, randomized clinical trial with selisistat, a SirT1 inhibitor, in patients with Huntington's disease. Br J Clin Pharmacol. 79:465–476. 2015. View Article : Google Scholar : PubMed/NCBI

57 

Heltweg B, Gatbonton T, Schuler AD, Posakony J, Li H, Goehle S, Kollipara R, Depinho RA, Gu Y, Simon JA and Bedalov A: Antitumor activity of a small-molecule inhibitor of human silent information regulator 2 enzymes. Cancer Res. 66:4368–4377. 2006. View Article : Google Scholar : PubMed/NCBI

58 

Kalle AM, Mallika A, Badiger J, Alinakhi, Talukdar P and Sachchidanand: Inhibition of SIRT1 by a small molecule induces apoptosis in breast cancer cells. Biochem Biophys Res Commun. 401:13–19. 2010. View Article : Google Scholar : PubMed/NCBI

59 

Lai YH, Lin SY, Wu YS, Chen HW and Chen JJW: AC-93253 iodide, a novel Src inhibitor, suppresses NSCLC progression by modulating multiple Src-related signaling pathways. J Hematol Oncol. 10:1722017. View Article : Google Scholar : PubMed/NCBI

60 

Rotili D, Tarantino D, Nebbioso A, Paolini C, Huidobro C, Lara E, Mellini P, Lenoci A, Pezzi R, Botta G, et al: Discovery of salermide-related sirtuin inhibitors: Binding mode studies and antiproliferative effects in cancer cells including cancer stem cells. J Med Chem. 55:10937–10947. 2012. View Article : Google Scholar : PubMed/NCBI

61 

Lara E, Mai A, Calvanese V, Altucci L, Lopez Nieva P, Martinez Chantar ML, Varela Rey M, Rotili D, Nebbioso A, Ropero S, et al: Salermide, a Sirtuin inhibitor with a strong cancer-specific proapoptotic effect. Oncogene. 28:781–791. 2009. View Article : Google Scholar : PubMed/NCBI

62 

Jiang Z, Chen K, Cheng L, Yan B, Qian W, Cao J, Li J, Wu E, Ma Q and Yang W: Resveratrol and cancer treatment: Updates. Ann N Y Acad Sci. 1403:59–69. 2017. View Article : Google Scholar : PubMed/NCBI

63 

Chauhan D, Bandi M, Singh AV, Ray A, Raje N, Richardson P and Anderson KC: Preclinical evaluation of a novel SIRT1 modulator SRT1720 in multiple myeloma cells. Br J Haematol. 155:588–598. 2011. View Article : Google Scholar : PubMed/NCBI

64 

Tasoulas J, Giaginis C, Patsouris E, Manolis E and Theocharis S: Histone deacetylase inhibitors in oral squamous cell carcinoma treatment. Expert Opin Investig Drugs. 24:69–78. 2015. View Article : Google Scholar : PubMed/NCBI

65 

Bruzzese F, Leone A, Rocco M, Carbone C, Piro G, Caraglia M, Di Gennaro E and Budillon A: HDAC inhibitor vorinostat enhances the antitumor effect of gefitinib in squamous cell carcinoma of head and neck by modulating ErbB receptor expression and reverting EMT. J Cell Physiol. 226:2378–2390. 2011. View Article : Google Scholar : PubMed/NCBI

66 

Suzuki M, Endo M, Shinohara F, Echigo S and Rikiishi H: Enhancement of cisplatin cytotoxicity by SAHA involves endoplasmic reticulum stress-mediated apoptosis in oral squamous cell carcinoma cells. Cancer Chemother Pharmacol. 64:1115–1122. 2009. View Article : Google Scholar : PubMed/NCBI

67 

Eriksson I, Joosten M, Roberg K and Ollinger K: The histone deacetylase inhibitor trichostatin A reduces lysosomal pH and enhances cisplatin-induced apoptosis. Exp Cell Res. 319:12–20. 2013. View Article : Google Scholar : PubMed/NCBI

68 

Sato T, Suzuki M, Sato Y, Echigo S and Rikiishi H: Sequence-dependent interaction between cisplatin and histone deacetylase inhibitors in human oral squamous cell carcinoma cells. Int J Oncol. 28:1233–1241. 2006.PubMed/NCBI

69 

Shoji M, Ninomiya I, Makino I, Kinoshita J, Nakamura K, Oyama K, Nakagawara H, Fujita H, Tajima H, Takamura H, et al: Valproic acid, a histone deacetylase inhibitor, enhances radiosensitivity in esophageal squamous cell carcinoma. Int J Oncol. 40:2140–2146. 2012.PubMed/NCBI

70 

Gan CP, Hamid S, Hor SY, Zain RB, Ismail SM, Wan Mustafa WM, Teo SH, Saunders N and Cheong SC: Valproic acid: growth inhibition of head and neck cancer by induction of terminal differentiation and senescence. Head Neck. 34:344–353. 2012. View Article : Google Scholar : PubMed/NCBI

71 

Gong L, Wang WM, Ji Y, Wang Y and Li DW: Effects of sodium butyrate on proliferation of human oral squamous carcinoma cell line and expression of p27Kip1. Zhonghua Kou Qiang Yi Xue Za Zhi. 45:619–622. 2010.PubMed/NCBI

72 

Lin Z and Fang D: The roles of SIRT1 in cancer. Genes Cancer. 4:97–104. 2013. View Article : Google Scholar : PubMed/NCBI

73 

Salminen A, Kaarniranta K and Kauppinen A: Crosstalk between oxidative stress and SIRT1: Impact on the ageing process. Int J Mol Sci. 14:3834–3859. 2013. View Article : Google Scholar : PubMed/NCBI

74 

Agarwal SK: Integrins and cadherins as therapeutic targets in fibrosis. Front Pharmacol. 5:1312014. View Article : Google Scholar : PubMed/NCBI

75 

Xue H, Atakilit A, Zhu W, Li X, Ramos DM and Pytela R: Role of the avb6 integrin in human oral squamous cell carcinoma growth in vivo and in vitro. Biochem Biophys Res Commun. 288:610–618. 2008. View Article : Google Scholar

76 

Xu M, Yin L, Cai Y, Hu Q, Huang J, Ji Q, Hu Y, Huang W, Liu F, Shi S and Deng X: Epigenetic regulation of integrin β6 transcription induced by TGF-β1 in human oral squamous cell carcinoma cells. J Cell Biochem. 119:4193–4204. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2019
Volume 17 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Islam S, Abiko Y, Uehara O and Chiba I: Sirtuin 1 and oral cancer (Review). Oncol Lett 17: 729-738, 2019
APA
Islam, S., Abiko, Y., Uehara, O., & Chiba, I. (2019). Sirtuin 1 and oral cancer (Review). Oncology Letters, 17, 729-738. https://doi.org/10.3892/ol.2018.9722
MLA
Islam, S., Abiko, Y., Uehara, O., Chiba, I."Sirtuin 1 and oral cancer (Review)". Oncology Letters 17.1 (2019): 729-738.
Chicago
Islam, S., Abiko, Y., Uehara, O., Chiba, I."Sirtuin 1 and oral cancer (Review)". Oncology Letters 17, no. 1 (2019): 729-738. https://doi.org/10.3892/ol.2018.9722