1
|
Zhang C, Ren L, Zhang H, Yang S, Deng M, He L, Cao R, Zhao C, Xia J. SESN1, negatively regulated by miR-377-3p, suppresses invasive growth of head and neck squamous cell carcinoma by interaction with SMAD3. Hum Cell 2022; 35:1100-1113. [PMID: 35622213 DOI: 10.1007/s13577-022-00719-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 05/06/2022] [Indexed: 11/30/2022]
Abstract
Sestrin 1 (SESN1) is a stress-inducible protein that suppresses tumors in numerous cancers. However, the function of SESN1 in head and neck squamous cell carcinoma (HNSCC) is not clear and needs to be elucidated. Here, SESN1 expression was downregulated in HNSCC tissues and cell lines, and low SESN1 expression was positively correlated with poor prognosis in patients with HNSCC. Moreover, SESN1 overexpression inhibited the proliferation, migration, and invasion of HSC-6 and CAL-33 cells. In addition, the binding relationship between miR-377-3p and SESN1 was confirmed using luciferase reporter and RNA immunoprecipitation assays. Downregulation of SESN1 expression was consistent with high levels of miR-377-3p in HNSCC tissues. Linear regression analysis of clinical HNSCC tissues revealed a negative correlation between miR-377-3p and SESN1 expression. Moreover, co-immunoprecipitation mass spectrometry analysis revealed that SESN1 interacted with SMAD3, and SMAD3 reversed the increased proliferation, migration, and invasion of HSC-6 and CAL-33 cells caused by SESN1 knockdown. In conclusion, these findings provide evidence that SESN1 functions as a tumor suppressor and reveal the miR-377-3p-SESN1-SMAD3 regulatory axis that contributes to proliferation, migration, and invasion in HNSCC development, which may represent an interventional target for HNSCC therapy.
Collapse
Affiliation(s)
- Chi Zhang
- Hostpital of Stomatology, Sun Yat-Sen University, No.55 Linyuan Xi Road, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, 510055, Guangdong, People's Republic of China
| | - Lin Ren
- Hostpital of Stomatology, Sun Yat-Sen University, No.55 Linyuan Xi Road, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, 510055, Guangdong, People's Republic of China
| | - Hongjian Zhang
- Hostpital of Stomatology, Sun Yat-Sen University, No.55 Linyuan Xi Road, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, 510055, Guangdong, People's Republic of China
| | - Shiwen Yang
- Hostpital of Stomatology, Sun Yat-Sen University, No.55 Linyuan Xi Road, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, 510055, Guangdong, People's Republic of China
| | - Miao Deng
- Hostpital of Stomatology, Sun Yat-Sen University, No.55 Linyuan Xi Road, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, 510055, Guangdong, People's Republic of China
| | - Lihong He
- Hostpital of Stomatology, Sun Yat-Sen University, No.55 Linyuan Xi Road, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, 510055, Guangdong, People's Republic of China
| | - Ruoyan Cao
- Hostpital of Stomatology, Sun Yat-Sen University, No.55 Linyuan Xi Road, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, 510055, Guangdong, People's Republic of China
| | - Chuanjiang Zhao
- Hostpital of Stomatology, Sun Yat-Sen University, No.55 Linyuan Xi Road, Guangzhou, People's Republic of China. .,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, 510055, Guangdong, People's Republic of China.
| | - Juan Xia
- Hostpital of Stomatology, Sun Yat-Sen University, No.55 Linyuan Xi Road, Guangzhou, People's Republic of China. .,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, 510055, Guangdong, People's Republic of China.
| |
Collapse
|
2
|
Xia H, Liu Y, Xu L, Mao H, Zhou Q, Xie Y. APOBEC1 complementation factor facilitates cell migration by promoting nucleus translocation of SMAD3 in renal cell carcinoma cells. In Vitro Cell Dev Biol Anim 2021; 57:501-509. [PMID: 34002283 DOI: 10.1007/s11626-021-00589-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/02/2021] [Indexed: 11/25/2022]
Abstract
Metastasis is inevitable in about 30% of patients with primary renal cell carcinoma after nephrectomy treatment. APOBEC1 complementation factor (A1CF), an RNA binding protein, participates in tumor progressions such as growth, apoptosis, differentiation, and invasion. Here, we explored biological functions of A1CF and provided a new insight into renal cell carcinoma metastasis. Wound healing assay was conducted to detect migration in A1CF overexpression and knockdown stable cell lines. Quantitative PCR and western blot assays were utilized to test transcriptional and translation levels of A1CF and SMAD3 in A1CF overexpression and knockdown renal carcinoma cells. Nuclear and cytoplasmic protein separation assays were conducted to evaluate the subcellular distribution of A1CF and SMAD3. Immunoprecipitation assay was conducted to detect the interaction between A1CF and SMAD3. Our study demonstrated A1CF overexpression facilitated cell migration in renal carcinoma cells. A1CF deficiency downregulated expression of SMAD3, Snail1, and N-cadherin. In addition, A1CF promoted nucleus translocation of SMAD3 and interacted with SMAD3. SMAD3 knockdown attenuated cell migration induced by A1CF overexpression. Our study suggested A1CF facilitated cell migration by promoting nucleus translocation of SMAD3 in renal cell carcinoma cells.
Collapse
Affiliation(s)
- Hua Xia
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Yuzhong District, Chongqing, 400016, China
| | - Yamin Liu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Lei Xu
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Yuzhong District, Chongqing, 400016, China
| | - Huajie Mao
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Yuzhong District, Chongqing, 400016, China
| | - Qin Zhou
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Yuzhong District, Chongqing, 400016, China
| | - Yajun Xie
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
3
|
Karatas OF, Capik O, Barlak N, Aydin Karatas E. Comprehensive in silico analysis for identification of novel candidate target genes, including DHX36, OPA1, and SENP2, located on chromosome 3q in head and neck cancers. Head Neck 2020; 43:288-302. [PMID: 33006201 DOI: 10.1002/hed.26493] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/27/2020] [Accepted: 09/21/2020] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Major milestones of head and neck carcinogenesis have been associated with various genetic abnormalities; however, a clear picture of the molecular networks deregulated during the carcinogenesis of head and neck squamous cell carcinoma (HNSC) has not yet completely revealed. METHODS In this study, we used in silico tools and online data sets to evaluate the underlying reasons for the expressional changes of genes residing within the chromosome 3q and to help understanding their contributions to HNSC carcinogenesis. RESULTS We found that 13 of 20 most upregulated genes in HNSC are localized to 3q. Further analysis revealed a gene signature consisting of DHX36, OPA1, and SENP2, which showed significant correlation in HNSC samples and potentially be deregulated through similar mechanisms including DNA amplification, transcriptional, and posttranscriptional regulation. CONCLUSIONS Considering our findings, we suggest DHX36, OPA1, and SENP2 genes as overexpressed in HNSC tumors and that might be concurrently involved in HNSC carcinogenesis, tumor progression, and induction of angiogenic pathways.
Collapse
Affiliation(s)
- Omer Faruk Karatas
- Molecular Biology and Genetics Department, Erzurum Technical University, Erzurum, Turkey.,Molecular Cancer Biology Laboratory, High Technology Application and Research Center, Erzurum Technical University, Erzurum, Turkey
| | - Ozel Capik
- Molecular Biology and Genetics Department, Erzurum Technical University, Erzurum, Turkey.,Molecular Cancer Biology Laboratory, High Technology Application and Research Center, Erzurum Technical University, Erzurum, Turkey
| | - Neslisah Barlak
- Molecular Biology and Genetics Department, Erzurum Technical University, Erzurum, Turkey.,Molecular Cancer Biology Laboratory, High Technology Application and Research Center, Erzurum Technical University, Erzurum, Turkey
| | - Elanur Aydin Karatas
- Molecular Biology and Genetics Department, Erzurum Technical University, Erzurum, Turkey.,Molecular Cancer Biology Laboratory, High Technology Application and Research Center, Erzurum Technical University, Erzurum, Turkey
| |
Collapse
|
4
|
Li Z, Liu FY, Kirkwood KL. The p38/MKP-1 signaling axis in oral cancer: Impact of tumor-associated macrophages. Oral Oncol 2020; 103:104591. [PMID: 32058294 PMCID: PMC7136140 DOI: 10.1016/j.oraloncology.2020.104591] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 02/04/2020] [Indexed: 02/07/2023]
Abstract
Oral squamous cell carcinomas (OSCC) constitute over 95% of all head and neck malignancies. As a key component of the tumor microenvironment (TME), chronic inflammation contributes towards the development, progression, and regional metastasis of OSCC. Tumor associated macrophages (TAMs) associated with OSSC promote tumorigenesis through the production of cytokines and pro-inflammatory factors that are critical role in the various steps of malignant transformation, including tumor growth, survival, invasion, angiogenesis, and metastasis. The mitogen-activated protein kinases (MAPKs) can regulate inflammation along with a wide range of cellular processes including cell metabolism, proliferation, motility, apoptosis, survival, differentiation and play a crucial role in cell growth and survival in physiological and pathological processes including innate and adaptive immune responses. Dual specificity MAPK phosphatases (MKPs) deactivates MAPKs. MKPs are considered as an important feedback control mechanism that limits MAPK signaling and subsequent target gene expression. This review outlines the role of MKP-1, the founding member of the MKP family, in OSCC and the TME. Herein, we summarize recent progress in understanding the regulation of p38 MAPK/MKP-1 signaling pathways via TAM-related immune responses in OSCC development, progression and treatment outcomes.
Collapse
Affiliation(s)
- Zhenning Li
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, China
- Department of Medical Genetics, China Medical University, Shenyang, China
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY, USA
| | - Fa-yu Liu
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, China
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY, USA
| | - Keith L. Kirkwood
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY, USA
- Department of Head and Neck/Plastic and Reconstructive Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
5
|
Lin LH, Chang KW, Cheng HW, Liu CJ. SMAD4 Somatic Mutations in Head and Neck Carcinoma Are Associated With Tumor Progression. Front Oncol 2019; 9:1379. [PMID: 31867281 PMCID: PMC6909744 DOI: 10.3389/fonc.2019.01379] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/22/2019] [Indexed: 12/19/2022] Open
Abstract
As the incidence and the mortality rate of head and neck squamous cell carcinoma (HNSCC) is increasing worldwide, gaining knowledge about the genomic changes which happen in the carcinogenesis of HNSCC is essential for the diagnosis and therapy of the disease. SMAD4 (DPC4) is a tumor suppressor gene. It is located at chromosome 18q21.1 and a member of the SMAD family. Which mediates the TGF-β signaling pathway, thereby controlling the growth of epithelial cells. In the study presented here, we analyzed tumor samples by multiplex PCR-based next-generation sequencing (NGS) and found deleterious mutations of SMAD4 in 4.1% of the tumors. Knock-down experiments of endogenous and exogenous SMAD4 expression demonstrated that SMAD4 is involved in the migration and invasion of HNSCC cells. Functional analysis of a missense mutation in the MH1 domain of SMAD4 may be responsible for the loss of function in suppressing tumor progression. Missense SMAD4 mutations, therefore, could be useful prognostic determinants for patients affected by HNSCCs. This report is the first study where NGS analysis based on multiplex-PCR is used to demonstrate the imminent occurrence of missense SMAD4 mutations in HNSCC cells. The gene analysis that we performed may support the identification of SMAD4 mutations as a diagnostic marker or even as a potential therapeutic target in head and neck cancer. Moreover, the analytic strategy proposed for the detection of mutations in the SMAD4 gene may be validated as a platform to assist mutation screening.
Collapse
Affiliation(s)
- Li-Han Lin
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Kuo-Wei Chang
- Department of Stomatology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Dentistry, Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan
| | - Hui-Wen Cheng
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chung-Ji Liu
- School of Dentistry, Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan.,Department of Oral and Maxillofacial Surgery, Taipei MacKay Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
6
|
Hernandez AL, Wang Y, Somerset HL, Keysar SB, Aisner DL, Marshall C, Bowles DW, Karam SD, Raben D, Jimeno A, Varella-Garcia M, Wang XJ. Inter- and intra-tumor heterogeneity of SMAD4 loss in head and neck squamous cell carcinomas. Mol Carcinog 2019; 58:666-673. [PMID: 30575147 DOI: 10.1002/mc.22958] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 12/04/2018] [Accepted: 12/15/2018] [Indexed: 12/16/2022]
Abstract
Reports regarding the frequency of SMAD4 loss in human head and neck squamous cell carcinoma (HNSCC) vary significantly. We have shown that SMAD4 deletion contributes to HNSCC initiation and progression. Therefore, accurately detecting genetic SMAD4 loss is critical to determine prognosis and therapeutic interventions in personalized medicine. We developed a SMAD4 fluorescence in situ hybridization (FISH) assay to identify chromosomal SMAD4 loss at the single cell level of primary HNSCC specimens and patient derived xenograft (PDX) tumors derived from HNSCCs. SMAD4 heterozygous loss was detected in 35% of primary HNSCCs and 41.3% of PDX tumors. Additionally, 4.3% of PDX tumors had SMAD4 homozygous loss. These frequencies of SMAD4 loss were similar to those in The Cancer Genome Atlas (TCGA). However, we identified significant heterogeneities of SMAD4 loss (partial or complete) among cells within each tumor. We also found that aneuploidy (monosomy and polysomy) contributed greatly to how to define chromosomal SMAD4 deletion. Furthermore, in cultured PDX tumors, SMAD4 mutant cells outcompeted SMAD4 wildtype cells, resulting in establishing homogenous SMAD4 mutant HNSCC cell lines with partial or complete genomic SMAD4 loss, suggesting a survival advantage of SMAD4 mutant cells. Taken together, our study reveals inter- and intra-tumor heterogeneities of SMAD4 chromosomal loss in HNSCCs. Further, SMAD4 FISH assay provides a platform for future clinical diagnosis of SMAD4 chromosomal loss that potentially serves as a molecular marker for prognosis and therapeutic intervention in cancer patients.
Collapse
Affiliation(s)
- Ariel L Hernandez
- Department of Pathology, School of Medicine, Molecular Biology Program, Medical Scientist Training Program, University of Colorado, Aurora, Colorado
| | - Ying Wang
- University of Colorado Cancer Center, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado, Aurora, Colorado
| | - Hilary L Somerset
- Department of Pathology, School of Medicine, Molecular Biology Program, Medical Scientist Training Program, University of Colorado, Aurora, Colorado
| | - Stephen B Keysar
- Department of Medical Oncology, University of Colorado, Aurora, Colorado
| | - Dara L Aisner
- Department of Pathology, School of Medicine, Molecular Biology Program, Medical Scientist Training Program, University of Colorado, Aurora, Colorado
| | - Carrie Marshall
- Department of Pathology, School of Medicine, Molecular Biology Program, Medical Scientist Training Program, University of Colorado, Aurora, Colorado
| | - Daniel W Bowles
- Veterans Affairs Medical Center, VA Eastern Colorado Health Care System, Aurora, Colorado
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado, Aurora, Colorado
| | - David Raben
- Department of Radiation Oncology, University of Colorado, Aurora, Colorado
| | - Antonio Jimeno
- Department of Medical Oncology, University of Colorado, Aurora, Colorado
| | - Marileila Varella-Garcia
- University of Colorado Cancer Center, Molecular Pathology/Cytogenetics Shared Resource, University of Colorado, Aurora, Colorado
- Department of Medical Oncology, University of Colorado, Aurora, Colorado
| | - Xiao-Jing Wang
- Department of Pathology, School of Medicine, Molecular Biology Program, Medical Scientist Training Program, University of Colorado, Aurora, Colorado
- Veterans Affairs Medical Center, VA Eastern Colorado Health Care System, Aurora, Colorado
| |
Collapse
|
7
|
Wei M, Mao S, Lu G, Li L, Lan X, Huang Z, Chen Y, Zhao M, Zhao Y, Xia Q. Valproic acid sensitizes metformin-resistant human renal cell carcinoma cells by upregulating H3 acetylation and EMT reversal. BMC Cancer 2018; 18:434. [PMID: 29665787 PMCID: PMC5902941 DOI: 10.1186/s12885-018-4344-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 04/08/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Metformin (Met) is a widely available diabetic drug and shows suppressed effects on renal cell carcinoma (RCC) metabolism and proliferation. Laboratory studies in RCC suggested that metformin has remarkable antitumor activities and seems to be a potential antitumor drug. But the facts that metformin may be not effective in reducing the risk of RCC in cancer clinical trials made it difficult to determine the benefits of metformin in RCC prevention and treatment. The mechanisms underlying the different conclusions between laboratory experiments and clinical analysis remains unclear. The goal of the present study was to determine whether long-term metformin use can induce resistance in RCC, whether metformin resistance could be used to explain the disaccord in laboratory and clinical studies, and whether the drug valproic acid (VPA), which inhibits histone deacetylase, exhibits synergistic cytotoxicity with metformin and can counteract the resistance of metformin in RCC. METHODS We performed CCK8, transwell, wound healing assay, flow cytometry and western blotting to detect the regulations of proliferation, migration, cell cycle and apoptosis in 786-O, ACHN and metformin resistance 786-O (786-M-R) cells treated with VPA, metformin or a combination of two drugs. We used TGF-β, SC79, LY294002, Rapamycin, protein kinase B (AKT) inhibitor to treat the 786-O or 786-M-R cells and detected the regulations in TGF-β /pSMAD3 and AMPK/AKT pathways. RESULTS 786-M-R was refractory to metformin-induced antitumor effects on proliferation, migration, cell cycle and cell apoptosis. AMPK/AKT pathways and TGF-β/SMAD3 pathways showed low sensibilities in 786-M-R. The histone H3 acetylation diminished in the 786-M-R cells. However, the addition of VPA dramatically upregulated histone H3 acetylation, increased the sensibility of AKT and inhibited pSMAD3/SMAD4, letting the combination of VPA and metformin remarkably reappear the anti-tumour effects of metformin in 786-M-R cells. CONCLUSIONS VPA not only exhibits synergistic cytotoxicity with metformin but also counteracts resistance to metformin in renal cell carcinoma cell. The re-sensitization to metformin induced by VPA in metformin-resistant cells may help treat renal cell carcinoma patients.
Collapse
Affiliation(s)
- Muyun Wei
- Department of Center Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, 544 Jingsi Road, Jinan, 250001, Shandong Province, China
| | - Shaowei Mao
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, 9677 Jingshidong Road, Jinan, 250001, Shandong Province, China
| | - Guoliang Lu
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, 9677 Jingshidong Road, Jinan, 250001, Shandong Province, China
| | - Liang Li
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, 9677 Jingshidong Road, Jinan, 250001, Shandong Province, China
| | - Xiaopeng Lan
- Department of Urology, Qingdao center Hospital, Qingdao, 266042, Shandong Province, China
| | - Zhongxian Huang
- Department of Urology, Jinan center Hospital, Jinan, 250001, Shandong Province, China
| | - Yougen Chen
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, 9677 Jingshidong Road, Jinan, 250001, Shandong Province, China
| | - Miaoqing Zhao
- Department of pathology, Shandong Provincial Hospital Affiliated to Shandong University, 324, Jingwu weiqi Road, Jinan, 250001, Shandong Province, China
| | - Yueran Zhao
- Department of Center Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, 544 Jingsi Road, Jinan, 250001, Shandong Province, China
| | - Qinghua Xia
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, 9677 Jingshidong Road, Jinan, 250001, Shandong Province, China.
| |
Collapse
|
8
|
Wu F, Weigel KJ, Zhou H, Wang XJ. Paradoxical roles of TGF-β signaling in suppressing and promoting squamous cell carcinoma. Acta Biochim Biophys Sin (Shanghai) 2018; 50:98-105. [PMID: 29206939 PMCID: PMC5846704 DOI: 10.1093/abbs/gmx127] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 11/07/2017] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor β (TGF-β) signaling either promotes or inhibits tumor formation and/or progression of many cancer types including squamous cell carcinoma (SCC). Canonical TGF-β signaling is mediated by a number of downstream proteins including Smad family proteins. Alterations in either TGF-β or Smad signaling can impact cancer. For instance, defects in TGF-β type I and type II receptors (TGF-βRI and TGF-βRII) and in Smad2/3/4 could promote tumor development. Conversely, increased TGF-β1 and activated TGF-βRI and Smad3 have all been shown to have tumor-promoting effects in experimental systems of human and mouse SCCs. Among TGF-β/Smad signaling, only TGF-βRII or Smad4 deletion in mouse epithelium causes spontaneous SCC in the mouse model, highlighting the critical roles of TGF-βRII and Smad4 in tumor suppression. Herein, we review the dual roles of the TGF-β/Smad signaling pathway and related mechanisms in SCC, highlighting the potential benefits and challenges of TGF-β/Smad-targeted therapies.
Collapse
Affiliation(s)
- Fanglong Wu
- State Key Laboratory of Oral Diseases, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kelsey J Weigel
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Hongmei Zhou
- State Key Laboratory of Oral Diseases, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xiao-Jing Wang
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
9
|
Ozawa H, Ranaweera RS, Izumchenko E, Makarev E, Zhavoronkov A, Fertig EJ, Howard JD, Markovic A, Bedi A, Ravi R, Perez J, Le QT, Kong CS, Jordan RC, Wang H, Kang H, Quon H, Sidransky D, Chung CH. SMAD4 Loss Is Associated with Cetuximab Resistance and Induction of MAPK/JNK Activation in Head and Neck Cancer Cells. Clin Cancer Res 2017; 23:5162-5175. [PMID: 28522603 DOI: 10.1158/1078-0432.ccr-16-1686] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 12/01/2016] [Accepted: 05/15/2017] [Indexed: 01/01/2023]
Abstract
Purpose: We previously demonstrated an association between decreased SMAD4 expression and cetuximab resistance in head and neck squamous cell carcinoma (HNSCC). The purpose of this study was to further elucidate the clinical relevance of SMAD4 loss in HNSCC.Experimental Design: SMAD4 expression was assessed by IHC in 130 newly diagnosed and 43 patients with recurrent HNSCC. Correlative statistical analysis with clinicopathologic data was also performed. OncoFinder, a bioinformatics tool, was used to analyze molecular signaling in TCGA tumors with low or high SMAD4 mRNA levels. The role of SMAD4 was investigated by shRNA knockdown and gene reconstitution of HPV-negative HNSCC cell lines in vitro and in vivoResults: Our analysis revealed that SMAD4 loss was associated with an aggressive, HPV-negative, cetuximab-resistant phenotype. We found a signature of prosurvival and antiapoptotic pathways that were commonly dysregulated in SMAD4-low cases derived from TCGA-HNSCC dataset and an independent oral cavity squamous cell carcinoma (OSCC) cohort obtained from GEO. We show that SMAD4 depletion in an HNSCC cell line induces cetuximab resistance and results in worse survival in an orthotopic mouse model in vivo We implicate JNK and MAPK activation as mediators of cetuximab resistance and provide the foundation for the concomitant EGFR and JNK/MAPK inhibition as a potential strategy for overcoming cetuximab resistance in HNSCCs with SMAD4 loss.Conclusions: Our study demonstrates that loss of SMAD4 expression is a signature characterizing the cetuximab-resistant phenotype and suggests that SMAD4 expression may be a determinant of sensitivity/resistance to EGFR/MAPK or EGFR/JNK inhibition in HPV-negative HNSCC tumors. Clin Cancer Res; 23(17); 5162-75. ©2017 AACR.
Collapse
Affiliation(s)
- Hiroyuki Ozawa
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Ruchira S Ranaweera
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Evgeny Izumchenko
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Eugene Makarev
- Insilico Medicine, Inc, ETC, Johns Hopkins University, Baltimore, Maryland
| | - Alex Zhavoronkov
- Insilico Medicine, Inc, ETC, Johns Hopkins University, Baltimore, Maryland
| | - Elana J Fertig
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
- Department of Health Science Informatics, Johns Hopkins University, Baltimore, Maryland
| | - Jason D Howard
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Ana Markovic
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Atul Bedi
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Rajani Ravi
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jimena Perez
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Quynh-Thu Le
- Department of Pathology, Stanford University School of Medicine Stanford, California
| | - Christina S Kong
- Department of Pathology, Stanford University School of Medicine Stanford, California
| | - Richard C Jordan
- Departments of Orofacial Sciences and Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Hao Wang
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Hyunseok Kang
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Harry Quon
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland
| | - David Sidransky
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christine H Chung
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, Florida
| |
Collapse
|
10
|
Patient-Specific Screening Using High-Grade Glioma Explants to Determine Potential Radiosensitization by a TGF-β Small Molecule Inhibitor. Neoplasia 2017; 18:795-805. [PMID: 27978994 PMCID: PMC5156509 DOI: 10.1016/j.neo.2016.08.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/26/2016] [Accepted: 08/29/2016] [Indexed: 12/26/2022] Open
Abstract
High-grade glioma (HGG), a deadly primary brain malignancy, manifests radioresistance mediated by cell-intrinsic and microenvironmental mechanisms. High levels of the cytokine transforming growth factor-β (TGF-β) in HGG promote radioresistance by enforcing an effective DNA damage response and supporting glioma stem cell self-renewal. Our analysis of HGG TCGA data and immunohistochemical staining of phosphorylated Smad2, which is the main transducer of canonical TGF-β signaling, indicated variable levels of TGF-β pathway activation across HGG tumors. These data suggest that evaluating the putative benefit of inhibiting TGF-β during radiotherapy requires personalized screening. Thus, we used explant cultures of seven HGG specimens as a rapid, patient-specific ex vivo platform to test the hypothesis that LY364947, a small molecule inhibitor of the TGF-β type I receptor, acts as a radiosensitizer in HGG. Immunofluorescence detection and image analysis of γ-H2AX foci, a marker of cellular recognition of radiation-induced DNA damage, and Sox2, a stem cell marker that increases post-radiation, indicated that LY364947 blocked these radiation responses in five of seven specimens. Collectively, our findings suggest that TGF-β signaling increases radioresistance in most, but not all, HGGs. We propose that short-term culture of HGG explants provides a flexible and rapid platform for screening context-dependent efficacy of radiosensitizing agents in patient-specific fashion. This time- and cost-effective approach could be used to personalize treatment plans in HGG patients.
Collapse
|
11
|
Foroutan M, Cursons J, Hediyeh-Zadeh S, Thompson EW, Davis MJ. A Transcriptional Program for Detecting TGFβ-Induced EMT in Cancer. Mol Cancer Res 2017; 15:619-631. [PMID: 28119430 DOI: 10.1158/1541-7786.mcr-16-0313] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/19/2016] [Accepted: 01/04/2017] [Indexed: 11/16/2022]
Abstract
Most cancer deaths are due to metastasis, and epithelial-to-mesenchymal transition (EMT) plays a central role in driving cancer cell metastasis. EMT is induced by different stimuli, leading to different signaling patterns and therapeutic responses. TGFβ is one of the best-studied drivers of EMT, and many drugs are available to target this signaling pathway. A comprehensive bioinformatics approach was employed to derive a signature for TGFβ-induced EMT which can be used to score TGFβ-driven EMT in cells and clinical specimens. Considering this signature in pan-cancer cell and tumor datasets, a number of cell lines (including basal B breast cancer and cancers of the central nervous system) show evidence for TGFβ-driven EMT and carry a low mutational burden across the TGFβ signaling pathway. Furthermore, significant variation is observed in the response of high scoring cell lines to some common cancer drugs. Finally, this signature was applied to pan-cancer data from The Cancer Genome Atlas to identify tumor types with evidence of TGFβ-induced EMT. Tumor types with high scores showed significantly lower survival rates than those with low scores and also carry a lower mutational burden in the TGFβ pathway. The current transcriptomic signature demonstrates reproducible results across independent cell line and cancer datasets and identifies samples with strong mesenchymal phenotypes likely to be driven by TGFβ.Implications: The TGFβ-induced EMT signature may be useful to identify patients with mesenchymal-like tumors who could benefit from targeted therapeutics to inhibit promesenchymal TGFβ signaling and disrupt the metastatic cascade. Mol Cancer Res; 15(5); 619-31. ©2017 AACR.
Collapse
Affiliation(s)
- Momeneh Foroutan
- The University of Melbourne Department of Surgery, St. Vincent's Hospital, Parkville, Victoria, Australia.,Division of Bioinformatics, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Joseph Cursons
- Division of Bioinformatics, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Systems Biology Laboratory, Melbourne School of Engineering, The University of Melbourne, Parkville, Victoria, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Melbourne School of Engineering, The University of Melbourne, Parkville, Victoria, Australia
| | - Soroor Hediyeh-Zadeh
- Division of Bioinformatics, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Erik W Thompson
- The University of Melbourne Department of Surgery, St. Vincent's Hospital, Parkville, Victoria, Australia.,Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology, Queensland, Australia.,Translational Research Institute, Wooloongabba, Queensland, Australia
| | - Melissa J Davis
- Division of Bioinformatics, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia. .,Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dentistry and Health, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
12
|
HUI LIAN, WU HUA, YANG NING, GUO XING, JANG XUEJUN. Identification of prognostic microRNA candidates for head and neck squamous cell carcinoma. Oncol Rep 2016; 35:3321-30. [DOI: 10.3892/or.2016.4698] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 01/11/2016] [Indexed: 11/05/2022] Open
|
13
|
Huang MY, Lin CH, Huang CM, Tsai HL, Huang CW, Yeh YS, Chai CY, Wang JY. Relationships between SMAD3 expression and preoperative fluoropyrimidine-based chemoradiotherapy response in locally advanced rectal cancer patients. World J Surg 2015; 39:1257-1267. [PMID: 25561186 DOI: 10.1007/s00268-014-2917-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND SMAD3, which is accumulated in the nucleus, transcriptionally regulates TGF-β target genes, playing a significant role in mediating the activities of TGF-β. In this study, we assessed the roles of TGF-β1, SMAD3, and phosphorylated SMAD3 expressions in patients with locally advanced rectal cancer following preoperative fluoropyrimidine-based chemoradiotherapy. METHODS Using immunohistochemistry, we examined TGF-β1, SMAD3, and phosphorylated SMAD3 expressions in pre-chemoradiotherapy cancer tissues from 86 locally advanced rectal cancer patients. After chemoradiotherapy, 64 of 86 (74.4 %) locally advanced rectal cancer patients were classified as responders (pathological tumor regression grades of 2-4). RESULTS A multivariate analysis showed that phosphorylated SMAD3 overexpression correlated to poor preoperative chemoradiotherapy responses (P = 0.015; OR 7.218; 95 % CI 1.479-35.229). Furthermore, a poor response (pathological tumor regression grades of 0-1) was an independent predictor of postoperative relapse (P = 0.021; OR 5.452; 95 % CI 1.286-23.113). Additionally, patients with phosphorylated SMAD3 overexpression were found to have a worse disease-free survival (P = 0.023). CONCLUSIONS Our data suggested that analyzing pre-chemoradiotherapy tumors for phosphorylated SMAD3 overexpression would assist physicians in identifying locally advanced rectal cancer patients who may have a poor response risk to preoperative fluoropyrimidine-based chemoradiotherapy.
Collapse
Affiliation(s)
- Ming-Yii Huang
- Department of Radiation Oncology, Cancer Center, Kaohsiung Medical University Hospital, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Clinical and Molecular Implications of MED15 in Head and Neck Squamous Cell Carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1114-22. [DOI: 10.1016/j.ajpath.2014.12.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 12/18/2014] [Accepted: 12/23/2014] [Indexed: 11/21/2022]
|
15
|
Aberrant expression of p-Smad3 in oral carcinogenesis. Clin Oral Investig 2014; 19:613-8. [DOI: 10.1007/s00784-014-1281-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 07/10/2014] [Indexed: 01/12/2023]
|
16
|
Ohnuki H, Jiang K, Wang D, Salvucci O, Kwak H, Sánchez-Martín D, Maric D, Tosato G. Tumor-infiltrating myeloid cells activate Dll4/Notch/TGF-β signaling to drive malignant progression. Cancer Res 2014; 74:2038-49. [PMID: 24520074 DOI: 10.1158/0008-5472.can-13-3118] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Myeloid cells that orchestrate malignant progression in the tumor microenvironment offer targets for a generalized strategy to attack solid tumors. Through an analysis of tumor microenvironments, we explored an experimental model of lung cancer that uncovered a network of Dll4/Notch/TGF-β1 signals that links myeloid cells to cancer progression. Myeloid cells attracted to the tumor microenvironment by the tumor-derived cytokines CCL2 and M-CSF expressed increased levels of the Notch ligand Dll4, thereby activating Notch signaling in the tumor cells and amplifying tumor-intrinsic Notch activation. Heightened Dll4/Notch signaling in tumor cells magnified TGF-β-induced pSMAD2/3 signaling and was required to sustain TGF-β-induced tumor cell growth. Conversely, Notch blockade reduced TGF-β signaling and limited lung carcinoma tumor progression. Corroborating these findings, by interrogating RNAseq results from tumor and adjacent normal tissue in clinical specimens of human head and neck squamous carcinoma, we found evidence that TGF-β/Notch crosstalk contributed to progression. In summary, the myeloid cell-carcinoma signaling network we describe uncovers novel mechanistic links between the tumor microenvironment and tumor growth, highlighting new opportunities to target tumors where this network is active.
Collapse
Affiliation(s)
- Hidetaka Ohnuki
- Authors' Affiliations: Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, NIH; Department of Intramural Research National Institutes of Neurological Disorders and Stroke, NIH, Bethesda, Maryland; and W2Motif, LLC, San Diego, California
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Karathanasi V, Tosios KI, Nikitakis NG, Piperi E, Koutlas I, Trimis G, Sklavounou A. TGF-β1, Smad-2/-3, Smad-1/-5/-8, and Smad-4 signaling factors are expressed in ameloblastomas, adenomatoid odontogenic tumors, and calcifying cystic odontogenic tumors: an immunohistochemical study. J Oral Pathol Med 2012; 42:415-23. [PMID: 23157422 DOI: 10.1111/jop.12016] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2012] [Indexed: 12/20/2022]
Abstract
OBJECTIVES The TGF-β/Smad signaling pathway regulates diverse cellular functions, including tooth development, and is involved in numerous pathological processes such as tumorigenesis. The aim of this study was to investigate the immunoexpression of the TGF-β/Smad signaling pathway members in ameloblastoma (AM), calcifying cystic odontogenic tumor (CCOT), and adenomatoid odontogenic tumor (AOT). MATERIALS AND METHODS This retrospective cross-sectional study included 65 tissue specimens: 34 AMs, 13 CCOTs, and 18 AOTs. Serial sections were immunohistochemically stained with TGF-β1, Smad-4, Smad-1/-5/-8, and Smad-2/-3 antibodies, and a semiquantitative measurement of the positive cells was carried out by two oral pathologists using a 0-3 scale (0: no immunoreactivity, 1: <20% positive cells, 2: 20-50% positive cells, 3: >50% positive cells). RESULTS All biomarkers studied were found significantly decreased in AM compared to CCOT and AOT. AOT and CCOT expressed Smad-1/-5/-8 more strongly compared to AM (OR = 11.66, P < 0.001 and OR = 5.34, P = 0.013, respectively), and Smad-2/-3 immunostaining was found significantly increased in CCOT (OR = 10.42, P = 0.001) and AOT (OR = 5.16, P < 0.004) compared to AM. Similarly, Smad-4 was expressed more strongly in AOT and CCOT compared to AM (P = 0.001), while AOT demonstrated a fivefold higher chance to express TGF-β1 compared to AM (P = 0.011). CONCLUSION TGF-β/Smad signaling pathway is activated in AM, AOT, and CCOT. The statistically significant reduced TGF-β1/Smad immunoexpression in AM compared to AOT/CCOT could be associated with the more aggressive biological behavior of AM including increased cell proliferation and reduced apoptosis and differentiation. Thus, the biomarkers TGF-β, Smad-4, Smad-1/-5/-8, and Smad-2/-3 could serve as supplementary diagnostic indices between odontogenic tumors of high and low neoplastic dynamics.
Collapse
Affiliation(s)
- Vasiliki Karathanasi
- Department of Oral Pathology and Medicine, Dental School, University of Athens, Athens, Greece.
| | | | | | | | | | | | | |
Collapse
|
18
|
Chen YK, Huang AHC, Cheng PH, Yang SH, Lin LM. Overexpression of Smad proteins, especially Smad7, in oral epithelial dysplasias. Clin Oral Investig 2012; 17:921-32. [PMID: 22669485 DOI: 10.1007/s00784-012-0756-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 05/14/2012] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Transforming growth factor β, via membrane-bound receptors and downstream Smad2-4, 7, can modulate tumorigenesis. Smad2 and Smad3 heterodimerize with Smad4, and the complex migrates to the nucleus to regulate the expression of target genes. Smad7 is a key negative regulator of this signaling pathway. This study aimed to examine Smad2-4, 7 expression and phosphorylated Smad2-3 (p-Smad2-3) in oral epithelial dysplasia and compared it with normal oral mucosa, hyperkeratosis/epithelial hyperplasia and squamous cell carcinoma (SCC). MATERIALS AND METHODS Immunohistochemical staining of Smad2-4, 7 and p-Smad2-3, was performed for 75 samples of human oral mucosa, including hyperkeratosis/epithelial hyperplasia (n = 20), mild epithelial dysplasia (n = 11), moderate to severe epithelial dysplasia (n = 11), and SCC (n = 43). Normal buccal mucosa samples (n = 9) were also included. RESULTS A significant increase in Smad7 expression was observed in the ascending order of samples of normal oral mucosa, hyperkeratosis/epithelial hyperplasia/mild oral epithelial dysplasia, moderate to severe oral epithelial dysplasia, and well-differentiated oral SCC/moderately to poorly differentiated oral SCC. Additionally, significant increases in Smad7 expression were noted as compared with expression of Smad2-4 and p-Smad2-3 in lesions of hyperkeratosis/epithelial hyperplasia, mild oral epithelial dysplasia, moderate to severe oral epithelial dysplasia, well-differentiated oral SCC, and moderately to poorly differentiated oral SCC. CONCLUSIONS Our results indicate that Smad proteins, particularly Smad7, in oral epithelial dysplasia and SCC could contribute to the attenuation of Smads anti-proliferative signaling in cancer development. CLINICAL RELEVANCE Smad7 could be a marker for risk of malignant transformation of oral epithelial dysplasia.
Collapse
Affiliation(s)
- Yuk-Kwan Chen
- Division of Oral Pathology and Diagnosis, Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung, Taiwan
| | | | | | | | | |
Collapse
|