1
|
Ryan N, Lamenza F, Shrestha S, Upadhaya P, Springer A, Jordanides P, Pracha H, Roth P, Kumar R, Wang Y, Vilgelm AE, Satoskar A, Oghumu S. Host derived macrophage migration inhibitory factor expression attenuates anti-tumoral immune cell accumulation and promotes immunosuppression in the tumor microenvironment of head and neck squamous cell carcinoma. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167345. [PMID: 38992847 DOI: 10.1016/j.bbadis.2024.167345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/24/2024] [Accepted: 07/03/2024] [Indexed: 07/13/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a significant public health concern worldwide. Immunomodulatory targets in the HNSCC tumor microenvironment are crucial to enhance the efficacy of HNSCC immunotherapy. Macrophage migration inhibitory factor (MIF) is a pro-inflammatory cytokine that has been linked to poor prognosis in many cancers, but the mechanistic role of MIF in HNSCC remains unclear. Using a murine orthotopic oral cancer model in Mif+/+ or Mif-/- mice, we determined the function of host derived MIF in HNSCC tumor development, metastasis as well as localized and systemic tumor immune responses. We observed that Mif-/- mice have decreased tumor growth and tumor burden compared to their wild-type counterparts. Flow cytometric analysis of immune populations within the primary tumor site revealed increased Th1 and cytotoxic T cell recruitment to the HNSCC tumor microenvironment. Within the tumors of Mif-/- mice, MIF deletion also enhanced the effector function of anti-tumoral effector CD8+ T cells as well as Th1 cells and decreased the accumulation of granulocytic myeloid derived suppressor cells (g-MDSCs) in the tumor microenvironment. Furthermore, MDSCs isolated from tumor bearing mice chemotactically respond to MIF in a dose dependent manner. Taken together, our results demonstrate a chemotactic and immunomodulatory role for host derived MIF in promoting HNSCC and suggest that MIF targeted immunomodulation is a promising approach for HNSCC treatment.
Collapse
Affiliation(s)
- Nathan Ryan
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; College of Medicine, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Felipe Lamenza
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | - Suvekshya Shrestha
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | - Puja Upadhaya
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Anna Springer
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Pete Jordanides
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Hasan Pracha
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Peyton Roth
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Rathan Kumar
- Department of Hematology, The Ohio State University Wexner Medial Center, Columbus, OH 43210, USA
| | - Yinchong Wang
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Anna E Vilgelm
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Abhay Satoskar
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Steve Oghumu
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| |
Collapse
|
2
|
Zhang C, Cai Q, Ke J. Poor Prognosis of Oral Squamous Cell Carcinoma Correlates With ITGA6. Int Dent J 2023; 73:178-185. [PMID: 35820930 PMCID: PMC10023534 DOI: 10.1016/j.identj.2022.05.010] [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: 03/19/2022] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVES Oral cancer is the ninth most common cancer worldwide and a leading cause of cancer-related death. Oral squamous cell carcinoma (OSCC) accounts for 90% of all oral cancers. Autophagy is a conserved essential catabolic process related to OSCC. The aim of this study was to elucidate diagnostic and prognostic autophagy-related biomarkers in OSCC. METHODS The OSCC gene expression data set was obtained from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) between the OSCC samples and adjacent healthy tissues were identified by R software. The Human Autophagy Database was screened, which revealed 222 autophagy-related genes. The autophagy-related DEGs were identified. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were applied. Protein-protein interaction network analysis was performed in the STRING database. cytoHubba in the Cytoscape software was applied to determine the top 10 hub genes. The data set of patients with OSCC from The Cancer Genome Atlas (TCGA) was used to evaluate the prognostic value of the 10 hub genes. The association between prognosis-related hub genes and immune infiltrates was explored. RESULTS Twenty-seven autophagy-related DEGs were identified. The top 10 hub genes were CCL2, CDKN2A, CTSB, CTSD, CXCR4, ITGA6, MAP1LC3A, MAPK3, PARP1, and RAB11A. ITGA6 was identified as the most efficient biomarker. Receiver operating characteristic curve analysis indicated that ITGA6 had the highest diagnostic accuracy for OSCC (area under the curve = 0.925). ITGA6 expression was significantly related to immune infiltrates. CONCLUSIONS The autophagy-related gene ITGA6 might be an efficient diagnostic and prognostic biomarker in OSCC.
Collapse
Affiliation(s)
- Churen Zhang
- Department of Stomatology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, China.
| | - Qiaoling Cai
- Department of Stomatology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, China.
| | - Jianguo Ke
- Department of Stomatology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, China.
| |
Collapse
|
3
|
Liu Y, Zhang M, Liao Y, Chen H, Su D, Tao Y, Li J, Luo K, Wu L, Zhang X, Yang R. Human umbilical cord mesenchymal stem cell-derived exosomes promote murine skin wound healing by neutrophil and macrophage modulations revealed by single-cell RNA sequencing. Front Immunol 2023; 14:1142088. [PMID: 36999022 PMCID: PMC10044346 DOI: 10.3389/fimmu.2023.1142088] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/22/2023] [Indexed: 03/08/2023] Open
Abstract
IntroductionFull-thickness skin wound healing remains a serious undertaking for patients. While stem cell-derived exosomes have been proposed as a potential therapeutic approach, the underlying mechanism of action has yet to be fully elucidated. The current study aimed to investigate the impact of exosomes derived from human umbilical cord mesenchymal stem cells (hucMSC-Exosomes) on the single-cell transcriptome of neutrophils and macrophages in the context of wound healing.MethodsUtilizing single-cell RNA sequencing, the transcriptomic diversity of neutrophils and macrophages was analyzed in order to predict the cellular fate of these immune cells under the influence of hucMSC-Exosomes and to identify alterations of ligand-receptor interactions that may influence the wound microenvironment. The validity of the findings obtained from this analysis was subsequently corroborated by immunofluorescence, ELISA, and qRT-PCR. Neutrophil origins were characterized based on RNA velocity profiles.ResultsThe expression of RETNLG and SLC2A3 was associated with migrating neutrophils, while BCL2A1B was linked to proliferating neutrophils. The hucMSC-Exosomes group exhibited significantly higher levels of M1 macrophages (215 vs 76, p < 0.00001), M2 macrophages (1231 vs 670, p < 0.00001), and neutrophils (930 vs 157, p < 0.00001) when compared to control group. Additionally, it was observed that hucMSC-Exosomes elicit alterations in the differentiation trajectories of macrophages towards more anti-inflammatory phenotypes, concomitant with changes in ligand-receptor interactions, thereby facilitating healing.DiscussionThis study has revealed the transcriptomic heterogeneity of neutrophils and macrophages in the context of skin wound repair following hucMSC-Exosomes interventions, providing a deeper understanding of cellular responses to hucMSC-Exosomes, a rising target of wound healing intervention.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Medical School of Chinese People’s Liberation Army, Beijing, China
- Department of Dermatology, the Seventh Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Mingwang Zhang
- Department of Dermatology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Yong Liao
- Department of Dermatology, the Seventh Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Hongbo Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Shenzhen, China
| | - Dandan Su
- School of Pharmaceutical Sciences, Sun Yat-sen University, Shenzhen, China
| | - Yuandong Tao
- Department of Pediatric Urology, the Seventh Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Jiangbo Li
- Bioinformatics Center of Academy of Military Medical Sciences, Beijing, China
| | - Kai Luo
- Biomedical Treatment Center, the Seventh Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Lihua Wu
- Biomedical Treatment Center, the Seventh Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Xingyue Zhang
- Department of Dermatology, the Seventh Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Rongya Yang
- Department of Dermatology, the Seventh Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
- *Correspondence: Rongya Yang,
| |
Collapse
|
4
|
Na S, Cui H, Guo Z, Liang X, Sakran KA, Guo X, Li X, Xie L, Zhu Y, Qi H, Tu J. Overexpression of Mena is associated with tumor progression and poor prognosis in oral squamous cell carcinoma via EMT. Front Oncol 2022; 12:1052375. [PMID: 36620546 PMCID: PMC9822539 DOI: 10.3389/fonc.2022.1052375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Background Mena, a cytoskeletal regulatory protein, is involved in actin-based regulation of cell motility and adhesion, and contributes to tumor invasion and metastasis. However, the role of Mena in oral squamous cell carcinoma remains unclear. This is the first research focusing on the prognostic value of Mena in OSCC. In this study, we aimed to investigate the correlation between Mena expression and clinicopathological significance, as well as prognostic value in OSCC. Methods Mena gene expression profiles of OSCC and normal tissues were collected from Oncomine, TCGA, and GEO databases. Biological function was analyzed through GO, KEGG and GSEA enrichment. Further, the expression level of Mena and tumor-related markers in 151 OSCC specimens was examined by IHC staining based on tissue microarray. Kaplan-Meier analysis was used to assess the prognostic performance of Mena in OSCC. Result Mena was generally upregulation in various malignancies, especially OSCC. The functional analyses indicated that Mena was involved in the assembly and regulation of actin, cell movement, and EMT. IHC staining revealed that high expression of Mena in OSCC was correlated with Lymphatic metastasis, TNM stage, E-cadherin, Vimentin, and MMP-2, but insignificantly Ki67. Kaplan-Meier analysis demonstrated that elevated expression of Mena was significantly associated with poor overall survival and disease-free survival of OSCC patients. Conclusion Mena could be a novel biomarker for predicting the prognosis of OSCC patients, which supports a theoretical basis for developing molecular target therapy.
Collapse
Affiliation(s)
- Sijia Na
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China,Department of Oral and Maxillofacial Surgery, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Hao Cui
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China,Department of Oral and Maxillofacial Surgery, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Zhichen Guo
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China,Department of Oral and Maxillofacial Surgery, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xiang Liang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China,Department of Oral and Maxillofacial Surgery, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Karim Ahmed Sakran
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xiaomei Guo
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China,Department of Pathology, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xingqiang Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China,Department of Oral and Maxillofacial Surgery, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Linyang Xie
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China,Department of Oral and Maxillofacial Surgery, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yifei Zhu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China,Department of Oral and Maxillofacial Surgery, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Hong Qi
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China,Department of Pathology, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China,*Correspondence: Hong Qi, ; Junbo Tu,
| | - Junbo Tu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China,Department of Oral and Maxillofacial Surgery, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi, China,*Correspondence: Hong Qi, ; Junbo Tu,
| |
Collapse
|
5
|
Chiu WC, Ou DL, Tan CT. Mouse Models for Immune Checkpoint Blockade Therapeutic Research in Oral Cancer. Int J Mol Sci 2022; 23:ijms23169195. [PMID: 36012461 PMCID: PMC9409124 DOI: 10.3390/ijms23169195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/05/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
The most prevalent oral cancer globally is oral squamous cell carcinoma (OSCC). The invasion of adjacent bones and the metastasis to regional lymph nodes often lead to poor prognoses and shortened survival times in patients with OSCC. Encouraging immunotherapeutic responses have been seen with immune checkpoint inhibitors (ICIs); however, these positive responses to monotherapy have been limited to a small subset of patients. Therefore, it is urgent that further investigations into optimizing immunotherapies are conducted. Areas of research include identifying novel immune checkpoints and targets and tailoring treatment programs to meet the needs of individual patients. Furthermore, the advancement of combination therapies against OSCC is also critical. Thus, additional studies are needed to ensure clinical trials are successful. Mice models are advantageous in immunotherapy research with several advantages, such as relatively low costs and high tumor growth success rate. This review paper divided methods for establishing OSCC mouse models into four categories: syngeneic tumor models, chemical carcinogen induction, genetically engineered mouse, and humanized mouse. Each method has advantages and disadvantages that influence its application in OSCC research. This review comprehensively surveys the literature and summarizes the current mouse models used in immunotherapy, their advantages and disadvantages, and details relating to the cell lines for oral cancer growth. This review aims to present evidence and considerations for choosing a suitable model establishment method to investigate the early diagnosis, clinical treatment, and related pathogenesis of OSCC.
Collapse
Affiliation(s)
- Wei-Chiao Chiu
- Department of Medical Research, Fu-Jen Catholic University Hospital, Fu-Jen Catholic University, New Taipei City 24352, Taiwan
- Department of Otolaryngology, National Taiwan University Hospital, Taipei City 100225, Taiwan
| | - Da-Liang Ou
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei City 10051, Taiwan
- YongLin Institute of Health, National Taiwan University, Taipei City 10672, Taiwan
| | - Ching-Ting Tan
- Department of Otolaryngology, National Taiwan University Hospital, Taipei City 100225, Taiwan
- Stem Cell Core Laboratory, Center of Genomic Medicine, National Taiwan University, Taipei City 10051, Taiwan
- Department of Otolaryngology, College of Medicine, National Taiwan University, Taipei City 100233, Taiwan
- Department of Otolaryngology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu 302058, Taiwan
- Correspondence: ; Tel.: +886-2-23123456 (ext. 88649)
| |
Collapse
|
6
|
Ryan NM, Lamenza FF, Upadhaya P, Pracha H, Springer A, Swingler M, Siddiqui A, Oghumu S. Black raspberry extract inhibits regulatory T-cell activity in a murine model of head and neck squamous cell carcinoma chemoprevention. Front Immunol 2022; 13:932742. [PMID: 36016924 PMCID: PMC9395668 DOI: 10.3389/fimmu.2022.932742] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/18/2022] [Indexed: 11/29/2022] Open
Abstract
Head and neck squamous cell carcinomas (HNSCC) are one of the most diagnosed malignancies globally, with a 5-year survival rate of approximately 40% to 50%. Current therapies are limited to highly invasive surgery, aggressive radiation, and chemotherapies. Recent reports have demonstrated the potential phytochemical properties of black raspberries in inhibiting the progression of various cancers including HNSCCs. However, the effects of black raspberry extracts on immune cells of the tumor microenvironment, specifically regulatory T cells during HNSCC, have not been investigated. We used a mouse model of 4-nitroquinoline-1-oxide (4NQO) chemically induced HNSCC carcinogenesis to determine these effects. C57BL/6 mice were exposed to 4NQO for 16 weeks and regular water for 8 weeks. 4NQO-exposed mice were fed the AIN-76A control mouse diet or the AIN76 diet supplemented with black raspberry extract. At terminal sacrifice, tumor burdens and immune cell recruitment and activity were analyzed in the tumor microenvironment, draining lymph nodes, and spleens. Mice fed the BRB extract-supplemented diet displayed decreased tumor burden compared to mice provided the AIN-76A control diet. Black raspberry extract administration did not affect overall T-cell populations as well as Th1, Th2, or Th17 differentiation in spleens and tumor draining lymph nodes. However, dietary black raspberry extract administration inhibited regulatory T-cell recruitment to HNSCC tumor sites. This was associated with an increased cytotoxic immune response in the tumor microenvironment characterized by increased CD8+ T cells and enhanced Granzyme B production during BRB extract-mediated HNSCC chemoprevention. Interestingly, this enhanced CD8+ T-cell antitumoral response was localized at the tumor sites but not at spleens and draining lymph nodes. Furthermore, we found decreased levels of PD-L1 expression by myeloid populations in draining lymph nodes of black raspberry-administered carcinogen-induced mice. Taken together, our findings demonstrate that black raspberry extract inhibits regulatory T-cell recruitment and promotes cytotoxic CD8 T-cell activity at tumor sites during HNSCC chemoprevention. These results demonstrate the immunomodulatory potential of black raspberry extracts and support the use of black raspberry-derived phytochemicals as a complementary approach to HNSCC chemoprevention and treatment.
Collapse
Affiliation(s)
- Nathan M. Ryan
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Felipe F. Lamenza
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Microbiology, The Ohio State University, Columbus, OH, United States
| | - Puja Upadhaya
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Hasan Pracha
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Anna Springer
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Michael Swingler
- Department of Microbiology, Immunology, and Inflammation, Center of Neurovirology and Gene Editing, School of Medicine, Temple University, Philadelphia, PA, United States
| | - Arham Siddiqui
- Kentucky College of Osteopathic Medicine, University of Pikeville, Pikeville, KY, United States
| | - Steve Oghumu
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- *Correspondence: Steve Oghumu,
| |
Collapse
|
7
|
Yu Y, Tian J, Hou Y, Zhang X, Li L, Cong P, Ji L, Wang X. A signature of immune-related gene pairs (IRGPs) for risk stratification and prognosis of oral cancer patients. World J Surg Oncol 2022; 20:227. [PMID: 35804390 PMCID: PMC9264557 DOI: 10.1186/s12957-022-02630-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 05/06/2022] [Indexed: 11/23/2022] Open
Abstract
Background With low response to present immunotherapy, it is imperative to identify new immune-related biomarkers for more effective immunotherapies for oral cancer. Methods RNA profiles for 390 oral cancer patients and 32 normal samples were downloaded from The Cancer Genome Atlas (TCGA) database and differentially expressed genes (DEGs) were analyzed. Immune genesets from ImmPort repository were overlapped with DEGs. After implementing univariate Cox analysis and the least absolute shrinkage and selection operator (LASSO) Cox regression analysis, key immune-related gene pairs (IRGPs) among the overlapped DEGs for predicting the survival risk were obtained. Then, the cutoff of risk score was calculated by the receiver operating characteristic (ROC) curve to stratify oral cancer patients into high and low-risk groups. Multivariate Cox analysis was used to analyze independent prognostic indicators for oral cancer. Besides, infiltration of immune cells, functional annotation, and mutation analysis of IRGPs were conducted. Biological functions correlated with IRGPs were enriched by Gene Set Enrichment Analysis (GSEA) method. Results We identified 698 differentially expressed genes (DEGs) in response to oral cancer. 17 IRGPs among the DEGs were identified and integrated into a risk score model. Patients in the high-risk group have a significantly worse prognosis than those in the low-risk group in both training (P<0.001) and test (P=0.019) cohorts. Meanwhile, the IRGP model was identified as an independent prognostic factor for oral cancer. Different infiltration patterns of immune cells were found between the high- and low-risk groups that more types of T and B cells were enriched in the low-risk group. More immune-related signaling pathways were highly enriched in the low-risk group and Tenascin C (TNC) was the most frequently mutated gene. We have developed a novel 17-IRGPs signature for risk stratification and prognostic prediction of oral cancer. Conclusion Our study provides a foundation for improved immunotherapy and prognosis and is beneficial to the individualized management of oral cancer patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12957-022-02630-1.
Collapse
Affiliation(s)
- Yanling Yu
- Department of Stomatology, Weihai Central Hospital, Weihai, China
| | - Jing Tian
- Department of Stomatology, Feicheng Hospital Affiliated to Shandong First Medical University, Taian, China
| | - Yanni Hou
- Department of Special Dental Care Clinic, Wendeng Stomatology Hospital, Weihai, Shandong, China
| | - Xinxin Zhang
- Department of Stomatology, Feicheng Hospital Affiliated to Shandong First Medical University, Taian, China
| | - Linhua Li
- Repair Department of Stomatology, Shouguang Stomatology Hospital, Weifang, China
| | - Peifu Cong
- Department of Stomatology, Weihai Central Hospital, Weihai, China
| | - Lei Ji
- Operating room, Weihai Central Hospital, Weihai, China
| | - Xuri Wang
- Department of Stomatology, Weihai Central Hospital, Weihai, China.
| |
Collapse
|
8
|
Ni Y, Low JT, Silke J, O’Reilly LA. Digesting the Role of JAK-STAT and Cytokine Signaling in Oral and Gastric Cancers. Front Immunol 2022; 13:835997. [PMID: 35844493 PMCID: PMC9277720 DOI: 10.3389/fimmu.2022.835997] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 05/16/2022] [Indexed: 12/12/2022] Open
Abstract
When small proteins such as cytokines bind to their associated receptors on the plasma membrane, they can activate multiple internal signaling cascades allowing information from one cell to affect another. Frequently the signaling cascade leads to a change in gene expression that can affect cell functions such as proliferation, differentiation and homeostasis. The Janus kinase-signal transducer and activator of transcription (JAK-STAT) and the tumor necrosis factor receptor (TNFR) are the pivotal mechanisms employed for such communication. When deregulated, the JAK-STAT and the TNF receptor signaling pathways can induce chronic inflammatory phenotypes by promoting more cytokine production. Furthermore, these signaling pathways can promote replication, survival and metastasis of cancer cells. This review will summarize the essentials of the JAK/STAT and TNF signaling pathways and their regulation and the molecular mechanisms that lead to the dysregulation of the JAK-STAT pathway. The consequences of dysregulation, as ascertained from founding work in haematopoietic malignancies to more recent research in solid oral-gastrointestinal cancers, will also be discussed. Finally, this review will highlight the development and future of therapeutic applications which modulate the JAK-STAT or the TNF signaling pathways in cancers.
Collapse
Affiliation(s)
- Yanhong Ni
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jun T. Low
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - John Silke
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Lorraine A. O’Reilly
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
9
|
STAT1 is regulated by TRIM24 and promotes immunosuppression in head and neck squamous carcinoma cells, but enhances T cell antitumour immunity in the tumour microenvironment. Br J Cancer 2022; 127:624-636. [PMID: 35595823 PMCID: PMC9381763 DOI: 10.1038/s41416-022-01853-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 05/02/2022] [Accepted: 05/09/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is a significant problem and is frequently resistant to current treatments. STAT1 is important in anti-tumour immune responses against HNSCC. However, the role of STAT1 expression by tumour cells and its regulation during HNSCC is unclear. METHODS We determined the effects of STAT1 inhibition on tumour development and immunity in CAL27 and UMSCC22A HNSCC cell lines in vitro and in a HNSCC carcinogen-induced model in vivo. RESULTS STAT1 siRNA knockdown in human HNSCC cells impaired their proliferation and expression of the immunosuppressive marker PD-L1. Stat1-deficient mice displayed increased oral lesion incidence and multiplicity during tumour carcinogenesis in vivo. Immunosuppressive markers PD-1 in CD8+ T cells and PD-L1 in monocytic MDSCs and macrophages were reduced in oral tumours and draining lymph nodes of tumour-bearing Stat1-deficient mice. However, STAT1 was required for anti-tumour functions of T cells during HNSCC in vivo. Finally, we identified TRIM24 to be a negative regulator of STAT1 that plays a similar tumorigenic function to STAT1 in vitro and thus may be a potential target when treating HNSCC. CONCLUSION Our findings indicate that STAT1 activity plays an important role in tumorigenicity and immunosuppression during HNSCC development.
Collapse
|
10
|
Yin J, Zheng Z, Zeng X, Zhao Y, Ai Z, Yu M, Wu Y, Jiang J, Li J, Li S. lncRNA MALAT1 mediates osteogenic differentiation of bone mesenchymal stem cells by sponging miR-129-5p. PeerJ 2022; 10:e13355. [PMID: 35480561 PMCID: PMC9037136 DOI: 10.7717/peerj.13355] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/08/2022] [Indexed: 01/15/2023] Open
Abstract
Background Bone mesenchymal stem cells (BMSCs) have good osteogenic differentiation potential and have become ideal seed cells in bone tissue engineering. However, the osteogenic differentiation ability of BMSCs gradually weakens with age, and the regulatory mechanism is unclear. Method We conducted a bioinformatics analysis, dual-luciferase reporter (DLR) experiment, and RNA binding protein immunoprecipitation (RIP) to explore the hub genes that may affect BMSC functions. Results The expression level of long non-coding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (Malat1) was significantly higher in the BMSCs from elderly than younger mice, while miR-129-5p showed the opposite trend. The results of alkaline phosphatase staining, quantitative reverse transcription PCR and western blot experiments indicated that inhibiting the expression of Malat1 inhibits the osteogenic differentiation of BMSCs. This effect can be reversed by reducing the expression of miR-129-5p. Additionally, DLR and RIP experiments confirmed that Malat1 acts as a sponge for miR-129-5p. Conclusion Overall, our study findings indicated that lncRNA Malat1 may play a critical role in maintaining the osteoblast differentiation potential of BMSCs by sponging miR-129-5p.
Collapse
Affiliation(s)
- Junhao Yin
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Zhanglong Zheng
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Xiaoli Zeng
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Yijie Zhao
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
- Department of Prosthodontics, School and Hospital of Stomatology, Tongji University, Shanghai, China
| | - Zexin Ai
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Miao Yu
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Yang'ou Wu
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
- Department of Oral and Maxillofacial Surgery, Shanghai Xuhui District Dental Center, Jiaotong University, Shanghai, China
| | - Jirui Jiang
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Jia Li
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
- Department of Prosthodontics, School and Hospital of Stomatology, Tongji University, Shanghai, China
| | - Shengjiao Li
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| |
Collapse
|
11
|
Lin L, Song C, Wei Z, Zou H, Han S, Cao Z, Zhang X, Zhang G, Ran J, Cai Y, Han W. Multifunctional photodynamic/photothermal nano-agents for the treatment of oral leukoplakia. J Nanobiotechnology 2022; 20:106. [PMID: 35246146 PMCID: PMC8895861 DOI: 10.1186/s12951-022-01310-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/14/2022] [Indexed: 02/06/2024] Open
Abstract
Oral leukoplakia (OLK) has gained extensive attention because of the potential risk for malignant transformation. Photosensitizers (PSs) played an indispensable role in the photodynamic therapy (PDT) of OLK, but the poor light sensitivity greatly hampered its clinical application. Herein, a novel organic photosensitive ITIC-Th nanoparticles (ITIC-Th NPs) were developed for OLK photodynamic/photothermal therapy (PTT). ITIC-Th NPs present both high photothermal conversion efficiency (~ 38%) and suitable reactive oxygen species (ROS) generation ability under 660 nm laser irradiation, making them possess excellent PDT and PTT capability. In 4-nitroquinoline 1-oxide (4NQO)-induced oral precancerous animal models, ITIC-Th NPs effectively suppress the OLK's cancerization without apparent topical or systemic toxicity in vivo. This study offers a promising therapeutic strategy for PDT and PTT in OLK treatment, and this study is the first interdisciplinary research in the field of multimodal therapy for OLK.
Collapse
Affiliation(s)
- Lin Lin
- Department of Oral Medicine, Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China.,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, No 30 Zhongyang Road, Nanjing, 210008, China
| | - Chuanhui Song
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, No 30 Zhongyang Road, Nanjing, 210008, China.,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, No 30 Zhongyang Road, Nanjing, 210008, China.,Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Zheng Wei
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, No 30 Zhongyang Road, Nanjing, 210008, China.,Pediatric Dentistry, Nanjing Stomatology Hospital, Medical School of Nanjing University, No 30 Zhongyang road, Nanjing, 210008, China
| | - Huihui Zou
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, No 30 Zhongyang Road, Nanjing, 210008, China.,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, No 30 Zhongyang Road, Nanjing, 210008, China
| | - Shengwei Han
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, No 30 Zhongyang Road, Nanjing, 210008, China
| | - Zichen Cao
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, No 30 Zhongyang Road, Nanjing, 210008, China.,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, No 30 Zhongyang Road, Nanjing, 210008, China
| | - Xinyu Zhang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, No 30 Zhongyang Road, Nanjing, 210008, China.,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, No 30 Zhongyang Road, Nanjing, 210008, China
| | - Guorong Zhang
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, No 30 Zhongyang Road, Nanjing, 210008, China
| | - Jianchuan Ran
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, No 30 Zhongyang Road, Nanjing, 210008, China.,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, No 30 Zhongyang Road, Nanjing, 210008, China
| | - Yu Cai
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, 310014, Zhejiang, China.
| | - Wei Han
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, No 30 Zhongyang Road, Nanjing, 210008, China.
| |
Collapse
|
12
|
Nedungadi D, Ryan N, Anderson K, Lamenza FF, Jordanides PP, Swingler MJ, Rakotondraibe L, Riedl KM, Iwenofu H, Oghumu S. Modulation of the oral glucocorticoid system during black raspberry mediated oral cancer chemoprevention. Carcinogenesis 2021; 43:28-39. [PMID: 34888650 DOI: 10.1093/carcin/bgab118] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 11/17/2021] [Accepted: 06/06/2021] [Indexed: 12/24/2022] Open
Abstract
Recent reports suggest that glucocorticoids (GCs), which can be synthesized in the oral mucosa, play an important role in cancer development. Therefore, the objectives of this study were to characterize the role of the oral GC system in oral cancer, and determine the effect of black raspberry (BRB) administration on GC modulation during oral cancer chemoprevention. We determined the expression of GC enzymes in various oral cancer cell lines, and investigated the role of the GC inactivating enzyme HSD11B2 on CAL27 oral cancer cells using siRNA mediated knockdown approaches. Using two in vivo models of oral carcinogenesis with 4-nitroquinoline-1-oxide (4NQO) carcinogen on C57Bl/6 mice and F344 rats, we determined the effect of BRB on GC modulation during HNSCC chemoprevention. Our results demonstrate that HSD11B2, which inactivates cortisol to cortisone, is downregulated during oral carcinogenesis in clinical and experimental models. Knockdown of HSD11B2 in oral cancer cells promotes cellular proliferation, invasion and expression of angiogenic biomarkers EGFR and VEGFA. An ethanol extract of BRB increased HSD11B2 expression on oral cancer cells. Dietary administration of 5% BRB increased Hsd11b2 gene and protein expression and reduced the active GC, corticosterone, in cancer-induced mouse tongues. Our results demonstrate that the oral GC system is modulated during oral carcinogenesis, and black raspberry administration upregulates Hsd11b2 during oral cancer chemoprevention. In conclusion, our findings challenge the use of synthetic glucocorticoids in head and neck cancer, and support the use of natural product alternatives that potentially modulate GC metabolism in a manner that supports oral cancer chemoprevention.
Collapse
Affiliation(s)
- Divya Nedungadi
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Nathan Ryan
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA.,Division of Anatomy, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Kelvin Anderson
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Felipe F Lamenza
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA.,Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Pete P Jordanides
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Michael J Swingler
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Liva Rakotondraibe
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Kenneth M Riedl
- Department of Food Science, Parker Food Science Building, The Ohio State University, Columbus, Ohio, USA
| | - Hans Iwenofu
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Steve Oghumu
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
13
|
Tight junction protein CLDN17 serves as a tumor suppressor to reduce the invasion and migration of oral cancer cells by inhibiting epithelial-mesenchymal transition. Arch Oral Biol 2021; 133:105301. [PMID: 34781072 DOI: 10.1016/j.archoralbio.2021.105301] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To investigate claudin-17 (CLDN17) expression in oral cancer and its effect on epithelial-mesenchymal transition (EMT), invasion and migration in oral cancer cells. METHODS The GEO2R tool was used to analyze gene expression in two microarray datasets (GSE74530 and GSE146483) derived from the Gene Expression Omnibus (GEO) database. Gene Expression Profiling Interactive Analysis (GEPIA) verified CLDN17 expression in head and neck squamous cell carcinoma (HNSC) patients. Moreover, oral cancer cells were transfected with CLDN17 overexpression plasmid or CLDN17 shRNA to evaluate cell invasion and migration. Gene and protein expression was detected by qRT-PCR, immunohistochemistry and western blotting. RESULTS CLDN17 was one of the top 200 differentially expressed genes in the GSE74530 and GSE146483 datasets and was downregulated in oral cancer. CLDN17 expression was higher in HNSC tissues, and it was related to TNM staging. In HNSC tumors, CLDN17 expression was positively correlated with CDH1 but negatively related to VIM, SNAIL1, SNAIL2, and TWIST1. Meanwhile, we found that CLDN17 expression was lower in oral cancer tissues; it declined with higher T status, N status, M status and staging, lower differentiation grade, and a worse prognosis. Upregulation of CLDN17 inhibited the invasion and migration of oral cancer cells, with elevated CDH1 and reduced VIM, SNAIL1, SNAIL2, and TWIST1, while CLDN17 downregulation had the opposite effects. CONCLUSION CLDN17 may serve as a tumor suppressor in oral cancer since it could reduce the invasion and migration of cells by inhibiting the EMT process, thus becoming a potential therapeutic target in oral cancer.
Collapse
|
14
|
Integrative profiling analysis identifies the oncogenic long noncoding RNA DUXAP8 in oral cancer. Anticancer Drugs 2021; 31:792-798. [PMID: 32304409 DOI: 10.1097/cad.0000000000000936] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
A growing number of studies have revealed the critical roles of long noncoding RNAs (lncRNAs) in the tumorigenesis and cancer progression. Recently, next-generation sequencing technologies combined with bioinformatic have demonstrated that a great number of dysregulated lncRNAs are associated with diverse cancers. However, lots of lncRNAs' function and their underlying molecular mechanisms in oral carcinoma (OC) cancer remain unclear. In this study, we performed integrative lncRNA profiling analysis using the TCGA RNA sequencing data and gene microarray data from Gene Expression Omnibus to identify more OC associated lncRNAs. A total of 619 differentially expressed lncRNAs were identified between the five data sets, and only the double homeobox A pseudogene 8 (DUXAP8) was screened among the up-regulated lncRNAs in all the five groups. Meanwhile, univariate Cox regression analyses disclosed that some lncRNAs are associated with the outcome of OC patients, such as DUXAP8, LINC00152, MIR4435-2HG and LINC00582. Furthermore, we uncovered that silenced DUXAP8 expression exerted suppressive impact on the proliferation of OC cells through interacting with histone-lysine N-methyltransferase enzyme Enhancer of zeste homolog 2 (EZH2) and repressing KLF2 expression. In a word, we identified a lot of unreported OC associated lncRNAs, which may provide a useful resource of lncRNAs for other studies.
Collapse
|
15
|
Yang W, Zhou W, Zhao X, Wang X, Duan L, Li Y, Niu L, Chen J, Zhang Y, Han Y, Fan D, Hong L. Prognostic biomarkers and therapeutic targets in oral squamous cell carcinoma: a study based on cross-database analysis. Hereditas 2021; 158:15. [PMID: 33892811 PMCID: PMC8066950 DOI: 10.1186/s41065-021-00181-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 04/13/2021] [Indexed: 12/24/2022] Open
Abstract
Background Oral squamous cell carcinoma (OSCC) is a malignant cancer, the survival rate of patients is disappointing. Therefore, it is necessary to identify the driven-genes and prognostic biomarkers in OSCC. Methods Four Gene Expression Omnibus (GEO) datasets were integratedly analyzed using bioinformatics approaches, including identification of differentially expressed genes (DEGs), GO and KEGG analysis, construction of protein-protein interaction (PPI) network, selection of hub genes, analysis of prognostic information and genetic alterations of hub genes. ONCOMINE, The Cancer Genome Atlas (TCGA) and Human Protein Atlas databases were used to evaluate the expression and prognostic value of hub genes. Tumor immunity was assessed to investigate the functions of hub genes. Finally, Cox regression model was performed to construct a multiple-gene prognostic signature. Results Totally 261 genes were found to be dysregulated. 10 genes were considered to be the hub genes. The Kaplan-Meier analysis showed that upregulated SPP1, FN1, CXCL8, BIRC5, PLAUR, and AURKA were related to poor outcomes in OSCC patients. FOXM1 and TPX2 were considered as the potential immunotherapeutic targets with future clinical significance. Moreover, we constructed a nine-gene signature (TEX101, DSG2, SCG5, ADA, BOC, SCARA5, FST, SOCS1, and STC2), which can be utilized to predict prognosis of OSCC patients effectively. Conclusion These findings may provide new clues for exploring the molecular mechanisms and targeted therapy in OSCC. The hub genes and risk gene signature are helpful to the personalized treatment and prognostic judgement. Supplementary Information The online version contains supplementary material available at 10.1186/s41065-021-00181-1.
Collapse
Affiliation(s)
- Wanli Yang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, No.127, Changle West Road, Xi'an, 710032, Shaanxi Province, China
| | - Wei Zhou
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, No.127, Changle West Road, Xi'an, 710032, Shaanxi Province, China
| | - Xinhui Zhao
- Department of Thyroid and Breast Surgery, The Affiliated Hospital of Northwest University & Xi'an No.3 Hospital, Northwest University, Xi'an, 710018, Shaanxi Province, China
| | - Xiaoqian Wang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, No.127, Changle West Road, Xi'an, 710032, Shaanxi Province, China
| | - Lili Duan
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, No.127, Changle West Road, Xi'an, 710032, Shaanxi Province, China
| | - Yiding Li
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, No.127, Changle West Road, Xi'an, 710032, Shaanxi Province, China
| | - Liaoran Niu
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, No.127, Changle West Road, Xi'an, 710032, Shaanxi Province, China
| | - Junfeng Chen
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, No.127, Changle West Road, Xi'an, 710032, Shaanxi Province, China
| | - Yujie Zhang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, No.127, Changle West Road, Xi'an, 710032, Shaanxi Province, China
| | - Yu Han
- Department of Otolaryngology, Xijing Hospital, Fourth Military Medical University, No.127, Changle West Road, Xi'an, 710032, Shaanxi Province, China.
| | - Daiming Fan
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, No.127, Changle West Road, Xi'an, 710032, Shaanxi Province, China.
| | - Liu Hong
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, No.127, Changle West Road, Xi'an, 710032, Shaanxi Province, China.
| |
Collapse
|
16
|
Inhibition of PI3K Isoform p110γ Increases Both Anti-Tumor and Immunosuppressive Responses to Aggressive Murine Head and Neck Squamous Cell Carcinoma with Low Immunogenicity. Cancers (Basel) 2021; 13:cancers13050953. [PMID: 33668795 PMCID: PMC7956466 DOI: 10.3390/cancers13050953] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 02/08/2023] Open
Abstract
Simple Summary Poorly immunogenic head and neck squamous carcinomas (HNSCC) remain difficult to treat due to poor response rates to immunotherapy. Inhibition of the PI3K catalytic subunit p110γ, which is expressed in leukocytes and some HNSCCs, has shown promise in treating HNSCC; with clinical trials underway to gauge its effectiveness. However, the effect of PI3K p110γ inhibition on the host immune system in poorly immunogenic HNSCC has not been fully described. In this study, our group characterized the immune response to poorly immunogenic HNSCC in the absence of PI3K p110γ using an orthotopic mouse model with the MOC2 cell line. We found that mice lacking p110γ did not demonstrate significantly different tumor growth or metastasis, though we observed substantial elevation in both anti-tumor and immunosuppressive activity at the primary tumor site. Our results indicate that PI3K p110γ inhibition may potentially enhance anti-tumor immunity against poorly immunogenic HNSCC if administered with checkpoint inhibitors. Abstract HNSCC is the sixth most common cancer, with around 650,000 new cases yearly. Gain of function mutations in the PI3K pathway are common in HNSCC, and inhibition of the PI3K p110γ subunit has shown promise in HNSCC treatment. However, given that PI3K p110γ plays an important role in myeloid and lymphoid immune cell function, it is essential to understand how PI3K p110γ inhibition affects the anti-tumor immune response independent of tumor cells. To elucidate PI3K p110γ function in HNSCC, we employed an orthotopic mouse model using poorly immunogenic and aggressive cell line MOC2 on Pik3cg−/− mice. We observed that wild-type and Pik3cg−/− mice displayed similar rates of HNSCC tumor growth and metastasis after 20 days following tumor injection. T-cell infiltration and intrinsic T-cell responses to MOC2 oral tumors were comparable between wild-type and Pik3cg−/− mice. Interestingly, the immune response of tumor-bearing Pik3cg−/− mice was marked by increased anti-tumor cytotoxic molecules (IFN-γ, IL-17)) by T-cells and immune checkpoint marker (PD-L1, PD-1) expression by myeloid cells and T-cells compared to tumor-bearing wild-type mice. Taken together, our findings demonstrate that inhibition of PI3K p110γ modulates tumor-associated immune cells, which likely potentiates HNSCC treatment when used in combination with selective checkpoint inhibitors.
Collapse
|
17
|
Abstract
Initially identified as a T lymphocyte-elicited inhibitor of macrophage motility, macrophage migration inhibitory factor (MIF) has since been found to be expressed by nearly every immune cell type examined and overexpressed in most solid and hematogenous malignant cancers. It is localized to both extracellular and intracellular compartments and physically interacts with more than a dozen different cell surface and intracellular proteins. Although classically associated with and characterized as a mediator of pro-inflammatory innate immune responses, more recent studies demonstrate that, in malignant disease settings, MIF contributes to anti-inflammatory, immune evasive, and immune tolerant phenotypes in both innate and adaptive immune cell types. This review will summarize the studies describing MIF in tumor-specific innate and adaptive immune responses and attempt to reconcile these various pleiotropic functions in normal physiology.
Collapse
Affiliation(s)
- Jordan T. Noe
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY, United States
- J.G. Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Robert A. Mitchell
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY, United States
- J.G. Brown Cancer Center, University of Louisville, Louisville, KY, United States
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, United States
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States
| |
Collapse
|
18
|
Yin J, Zeng X, Ai Z, Yu M, Wu Y, Li S. Construction and analysis of a lncRNA-miRNA-mRNA network based on competitive endogenous RNA reveal functional lncRNAs in oral cancer. BMC Med Genomics 2020; 13:84. [PMID: 32571304 PMCID: PMC7310129 DOI: 10.1186/s12920-020-00741-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 06/15/2020] [Indexed: 12/24/2022] Open
Abstract
Background A growing evidence suggests that long non-coding RNAs (lncRNAs) can function as a microRNA (miRNA) sponge in various diseases including oral cancer. However, the pathophysiological function of lncRNAs remains unclear. Methods Based on the competitive endogenous RNA (ceRNA) theory, we constructed a lncRNA-miRNA-mRNA network in oral cancer with the human expression profiles GSE74530 from the Gene Expression Omnibus (GEO) database. We used topological analysis to determine the hub lncRNAs in the regulatory ceRNA network. Then, function enrichment analysis was performed using the clusterProfiler R package. Clinical information was downloaded from The Cancer Genome Atlas (TCGA) database and survival analysis was performed with Kaplan-Meier analysis. Results A total of 238 potential co-dysregulated competing triples were obtained in the lncRNA-associated ceRNA network in oral cancer, which consisted of 10 lncRNA nodes, 41 miRNA nodes and 122 mRNA nodes. Additionally, we found lncRNA HCG22 exhibiting superior potential as a diagnostic and prognostic marker of oral cancer. Conclusions Our findings provide novel insights to understand the ceRNA regulation in oral cancer and identify a novel lncRNA as a potential molecular biomarker.
Collapse
Affiliation(s)
- Junhao Yin
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Tongji University, Shanghai, 200072, China.,Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, China
| | - Xiaoli Zeng
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Tongji University, Shanghai, 200072, China.,Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, China
| | - Zexin Ai
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Tongji University, Shanghai, 200072, China.,Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, China
| | - Miao Yu
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Tongji University, Shanghai, 200072, China.,Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, China
| | - Yang'ou Wu
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Tongji University, Shanghai, 200072, China.,Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, China
| | - Shengjiao Li
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Tongji University, Shanghai, 200072, China. .,Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, China.
| |
Collapse
|
19
|
Multiple Functions of Fubp1 in Cell Cycle Progression and Cell Survival. Cells 2020; 9:cells9061347. [PMID: 32481602 PMCID: PMC7349734 DOI: 10.3390/cells9061347] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 12/11/2022] Open
Abstract
The discovery of novel and critical genes implicated in malignant development is a topic of high interest in cancer research. Intriguingly, a group of genes named “double-agent” genes were reported to have both oncogenic and tumor-suppressive functions. To date, less than 100 “double-agent” genes have been documented. Fubp1 is a master transcriptional regulator of a subset of genes by interacting with a far upstream element (FUSE). Mounting evidence has collectively demonstrated both the oncogenic and tumor suppressive roles of Fubp1 and the debate regarding its roles in tumorigenesis has been around for several years. Therefore, the detailed molecular mechanisms of Fubp1 need to be determined in each context. In the present study, we showed that the Fubp1 protein level was enriched in the S phase and we identified that Fubp1 deficiency altered cell cycle progression, especially in the S phase, by downregulating the mRNA expression levels of Ccna genes encoding cyclin A. Although this Fubp1-cyclin A axis appears to exist in several types of tumors, Fubp1 showed heterogeneous expression patterns among various cancer tissues, suggesting it exhibits multiple and complicated functions in cancer development. In addition, we showed that Fubp1 deficiency confers survival advantages to cells against metabolic stress and anti-cancer drugs, suggesting that Fubp1 may play both positive and negative roles in malignant development.
Collapse
|
20
|
Li Y, Cao X, Li H. Identification and Validation of Novel Long Non-coding RNA Biomarkers for Early Diagnosis of Oral Squamous Cell Carcinoma. Front Bioeng Biotechnol 2020; 8:256. [PMID: 32351944 PMCID: PMC7174591 DOI: 10.3389/fbioe.2020.00256] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 03/12/2020] [Indexed: 12/12/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are recently emerging as a novel promising biomarker for cancer diagnosis and prognosis. Despite these previous investigations, the expression pattern and diagnostic role of lncRNAs in oral squamous cell carcinoma (OSCC) remain unclear. In this study, we identified six novel lncRNA biomarkers (LINC01697, LINC02487, LOC105376575, AC005083.1, SLC8A1-AS1, and U62317.1) from a list of 29 differentially expressed lncRNAs using the least absolute shrinkage and selection operator (LASSO) method in the discovery dataset of 167 OSCC samples and 45 normal oral tissues. Using the logistic regression method, we constructed a six lncRNAs-based diagnostic risk model (6lncRNAScore) which was able to differentiate between OSCC samples and control samples with high performance with AUC of 0.995 and high diagnostic specificity of 88.9% and sensitivity of 98.2% in the discovery dataset. The diagnostic performance of the 6lncRNAScore was further validated in another two independent OSCC dataset with AUC of 0.968 and 1.0. Functional enrichment analysis for lncRNA biomarkers-related mRNAs suggested that lncRNAs biomarkers tended to be involved in the lipid metabolic process. Together, our study highlighted the importance of lncRNAs in OSCC and demonstrated the utility of lncRNA expression as a promising biomarker for early diagnosis of OSCC.
Collapse
Affiliation(s)
- Yue Li
- Department of Orthodontics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaofang Cao
- Department of Endodontics, The Second Affiliated Hospital of Endodontics, Harbin, China
| | - Hao Li
- Department of Stomatology, The Fourth Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| |
Collapse
|
21
|
Anderson K, Ryan N, Volpedo G, Varikuti S, Satoskar AR, Oghumu S. Immune Suppression Mediated by STAT4 Deficiency Promotes Lymphatic Metastasis in HNSCC. Front Immunol 2020; 10:3095. [PMID: 32010142 PMCID: PMC6974475 DOI: 10.3389/fimmu.2019.03095] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 12/18/2019] [Indexed: 12/13/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a prevalent form of cancer with 5-years survival rates around 57%, and metastasis is a leading cause of mortality. Host-derived immunological factors that affect HNSCC tumor development and metastasis are not completely understood. We investigated the role of host-derived signal transducer and activator of transcription 4 (STAT4) during experimental HNSCC using an aggressive and metastatic HNSCC cell line, LY2, which was orthotopically injected into the buccal sulcus of wild type (WT) and STAT4 deficient (Stat4-/-) BALB/c mice. Necropsies performed at terminal sacrifice revealed that Stat4-/- mice displayed comparable primary tumor growth to the WT mice. However, the rate and extent of lymph node and lung metastasis among Stat4-/- mice was significantly higher. Downstream analyses performed on primary tumors, draining lymph nodes, spleens and bone marrow revealed significant upregulation of lymphocytic immunosuppressive biomarkers as well as an accumulation of granulocytic MDSC subpopulations in draining lymph nodes of metastatic Stat4-/- mice. Further, we observed a significant decrease in TH1, TH17, and cytotoxic activity in tumor bearing Stat4-/- compared to WT mice. Our results demonstrate that STAT4 mediates resistance to HNSCC metastasis, and activation of STAT4 could potentially mitigate lymphatic metastasis in HNSCC patients.
Collapse
Affiliation(s)
- Kelvin Anderson
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Nathan Ryan
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Greta Volpedo
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Microbiology, The Ohio State University, Columbus, OH, United States
| | - Sanjay Varikuti
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Abhay R. Satoskar
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Microbiology, The Ohio State University, Columbus, OH, United States
| | - Steve Oghumu
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
22
|
Li Q, Dong H, Yang G, Song Y, Mou Y, Ni Y. Mouse Tumor-Bearing Models as Preclinical Study Platforms for Oral Squamous Cell Carcinoma. Front Oncol 2020; 10:212. [PMID: 32158692 PMCID: PMC7052016 DOI: 10.3389/fonc.2020.00212] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 02/06/2020] [Indexed: 12/16/2022] Open
Abstract
Preclinical animal models of oral squamous cell carcinoma (OSCC) have been extensively studied in recent years. Investigating the pathogenesis and potential therapeutic strategies of OSCC is required to further progress in this field, and a suitable research animal model that reflects the intricacies of cancer biology is crucial. Of the animal models established for the study of cancers, mouse tumor-bearing models are among the most popular and widely deployed for their high fertility, low cost, and molecular and physiological similarity to humans, as well as the ease of rearing experimental mice. Currently, the different methods of establishing OSCC mouse models can be divided into three categories: chemical carcinogen-induced, transplanted and genetically engineered mouse models. Each of these methods has unique advantages and limitations, and the appropriate application of these techniques in OSCC research deserves our attention. Therefore, this review comprehensively investigates and summarizes the tumorigenesis mechanisms, characteristics, establishment methods, and current applications of OSCC mouse models in published papers. The objective of this review is to provide foundations and considerations for choosing suitable model establishment methods to study the relevant pathogenesis, early diagnosis, and clinical treatment of OSCC.
Collapse
Affiliation(s)
- Qiang Li
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Heng Dong
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
- Department of Oral Implantology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Guangwen Yang
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yuxian Song
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yongbin Mou
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
- Department of Oral Implantology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
- *Correspondence: Yongbin Mou
| | - Yanhong Ni
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
- Yanhong Ni
| |
Collapse
|
23
|
Ryan N, Anderson K, Volpedo G, Hamza O, Varikuti S, Satoskar AR, Oghumu S. STAT1 inhibits T-cell exhaustion and myeloid derived suppressor cell accumulation to promote antitumor immune responses in head and neck squamous cell carcinoma. Int J Cancer 2019; 146:1717-1729. [PMID: 31709529 DOI: 10.1002/ijc.32781] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 10/21/2019] [Accepted: 10/31/2019] [Indexed: 12/16/2022]
Abstract
Cancers of the oral cavity remain the sixth most diagnosed cancer worldwide, with high rates of recurrence and mortality. We determined the role of STAT1 during oral carcinogenesis using two orthotopic models in mice genetically deficient for Stat1. Metastatic (LY2) and nonmetastatic (B4B8) head and neck squamous cell carcinoma (HNSCC) cell lines were injected into the oral cavity of Stat1 deficient (Stat1-/- ) and Stat1 competent (Stat1+/+ ) mice. Stat1-/- mice displayed increased tumor growth and metastasis compared to Stat1+/+ mice. Mechanistically, Stat1-/- mice displayed impaired CD4+ and CD8+ T-cell expansion compared to Stat1+/+ mice. This was associated with enhanced T-cell exhaustion, and severely attenuated T-cell antitumor effector responses including reduced expression of IFN-γ and perforin at the tumor site. Interestingly, tumor necrosis factor (TNF)-α production by T cells in tumor-bearing mice was suppressed by Stat1 deficiency. This deficiency in T-cell expansion and functional responses in mice was linked to PD-1 and CD69 overexpression in T cells of Stat1-/- mice. In contrast, we observed increased accumulation of CD11b+ Ly6G+ myeloid derived suppressor cells in tumors, draining lymph nodes, spleens and bone marrow of tumor-bearing Stat1-/- mice, resulting in a protumorigenic microenvironment. Our data demonstrates that STAT1 is an essential mediator of the antitumor response through inhibition of myeloid derived suppressor cell accumulation and promotion of T-cell mediated immune responses in murine head and neck squamous cell carcinoma. Selective induction of STAT1 phosphorylation in HNSCC patients could potentially improve oral tumor outcomes and response to therapy.
Collapse
Affiliation(s)
- Nathan Ryan
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Kelvin Anderson
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Greta Volpedo
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH.,Department of Microbiology, The Ohio State University, Columbus, OH
| | - Omar Hamza
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Sanjay Varikuti
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Abhay R Satoskar
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH.,Department of Microbiology, The Ohio State University, Columbus, OH
| | - Steve Oghumu
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH
| |
Collapse
|
24
|
Wang S, Li F, Qiang D, Hu Z, Meng Y, Shi L, Zhao E, Niu Y. Impact of immunodeficiency on lingual carcinogenesis and lymph node metastasis in mice. J Oral Pathol Med 2019; 48:826-831. [PMID: 31206766 DOI: 10.1111/jop.12916] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 05/19/2019] [Accepted: 05/21/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Shan Wang
- Department of Oral Pathology, Hospital of Stomatology, the First Affiliated Hospital Harbin Medical University Harbin China
- Institute of Oral Biomedicine Heilongjiang Academy of Medical Science Harbin China
| | - Fang Li
- Department of Oral and Maxillofacial Surgery Heilongjiang Provincial Hospital Harbin China
| | - Dongxia Qiang
- Department of Oral Pathology, Hospital of Stomatology, the First Affiliated Hospital Harbin Medical University Harbin China
| | - Zheng Hu
- Laboratory of Sono‐ and Photo‐theranostic Technologies Harbin Institute of Technology Harbin China
| | - Yan Meng
- Department of Oral Pathology, Hospital of Stomatology, the First Affiliated Hospital Harbin Medical University Harbin China
| | - Lei Shi
- Department of Oral Pathology, Hospital of Stomatology, the First Affiliated Hospital Harbin Medical University Harbin China
| | - Eryang Zhao
- Department of Oral Pathology, Hospital of Stomatology, the First Affiliated Hospital Harbin Medical University Harbin China
| | - Yumei Niu
- Institute of Oral Biomedicine Heilongjiang Academy of Medical Science Harbin China
- Department of Oral Medicine, Hospital of Stomatology, the First Affiliated Hospital Harbin Medical University Harbin China
| |
Collapse
|
25
|
Metabolic Regulation of Glycolysis and AMP Activated Protein Kinase Pathways during Black Raspberry-Mediated Oral Cancer Chemoprevention. Metabolites 2019; 9:metabo9070140. [PMID: 31336728 PMCID: PMC6680978 DOI: 10.3390/metabo9070140] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 06/15/2019] [Accepted: 07/08/2019] [Indexed: 12/26/2022] Open
Abstract
Oral cancer is a public health problem with an incidence of almost 50,000 and a mortality of 10,000 each year in the USA alone. Black raspberries (BRBs) have been shown to inhibit oral carcinogenesis in several preclinical models, but our understanding of how BRB phytochemicals affect the metabolic pathways during oral carcinogenesis remains incomplete. We used a well-established rat oral cancer model to determine potential metabolic pathways impacted by BRBs during oral carcinogenesis. F344 rats were exposed to the oral carcinogen 4-nitroquinoline-1-oxide in drinking water for 14 weeks, then regular drinking water for six weeks. Carcinogen exposed rats were fed a 5% or 10% BRB supplemented diet or control diet for six weeks after carcinogen exposure. RNA-Seq transcriptome analysis on rat tongue, and mass spectrometry and NMR metabolomics analysis on rat urine were performed. We tentatively identified 57 differentially or uniquely expressed metabolites and over 662 modulated genes in rats being fed with BRB. Glycolysis and AMPK pathways were modulated during BRB-mediated oral cancer chemoprevention. Glycolytic enzymes Aldoa, Hk2, Tpi1, Pgam2, Pfkl, and Pkm2 as well as the PKA-AMPK pathway genes Prkaa2, Pde4a, Pde10a, Ywhag, and Crebbp were downregulated by BRBs during oral cancer chemoprevention. Furthermore, the glycolysis metabolite glucose-6-phosphate decreased in BRB-administered rats. Our data reveal the novel metabolic pathways modulated by BRB phytochemicals that can be targeted during the chemoprevention of oral cancer.
Collapse
|
26
|
Rossa C, D'Silva NJ. Immune-relevant aspects of murine models of head and neck cancer. Oncogene 2019; 38:3973-3988. [PMID: 30696955 PMCID: PMC6533118 DOI: 10.1038/s41388-019-0686-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/26/2018] [Accepted: 12/05/2018] [Indexed: 12/19/2022]
Abstract
Head and neck cancers (HNCs) cause significant mortality and morbidity. There have been few advances in therapeutic management of HNC in the past 4 to 5 decades, which support the need for studies focusing on HNC biology. In recent years, increased recognition of the relevance of the host response in cancer progression has led to novel therapeutic strategies and putative biomarkers of tumor aggressiveness. However, tumor-immune interactions are highly complex and vary with cancer type. Pre-clinical, in vivo models represent an important and necessary step in understanding biological processes involved in development, progression and treatment of HNC. Rodents (mice, rats, hamsters) are the most frequently used animal models in HNC research. The relevance and utility of information generated by studies in murine models is unquestionable, but it is also limited in application to tumor-immune interactions. In this review, we present information regarding the immune-specific characteristics of the murine models most commonly used in HNC research, including immunocompromised and immunocompetent animals. The particular characteristics of xenograft, chemically induced, syngeneic, transgenic, and humanized models are discussed in order to provide context and insight for researchers interested in the in vivo study of tumor-immune interactions in HNC.
Collapse
Affiliation(s)
- Carlos Rossa
- Department of Diagnosis and Surgery, UNESP-State University of Sao Paulo, School of Dentistry at Araraquara, Araraquara - SP, Brazil. .,Department of Periodontics and Oral Medicine, School of Dentistry, Ann Arbor, MI, 48109, USA.
| | - Nisha J D'Silva
- Department of Periodontics and Oral Medicine, School of Dentistry, Ann Arbor, MI, 48109, USA. .,Department of Pathology, Medical School, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
27
|
Zhao C, Zou H, Zhang J, Wang J, Liu H. An integrated methylation and gene expression microarray analysis reveals significant prognostic biomarkers in oral squamous cell carcinoma. Oncol Rep 2018; 40:2637-2647. [PMID: 30226546 PMCID: PMC6151890 DOI: 10.3892/or.2018.6702] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 09/05/2018] [Indexed: 12/01/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a life-threatening disease with a poor prognosis. Although previous studies have reported that the methylation of certain genes is associated with the pathogenesis of OSCC, the methylation of genes that have relevance to OSCC progression is not clearly documented. The present study aimed to gain insights into the mechanisms underlying DNA methylation regulation associated with OSCC progression and to identify potential prognostic markers for OSCC treatment. DNA methylation dataset GSE41114 and gene expression dataset GSE74530 were downloaded from the Gene Expression Omnibus database. The global methylation status of OSCC tumor samples and normal control samples was determined, and differentially methylated genes (DMGs) in OSCC samples compared with control samples were identified. The mRNA expression data were then integrated to identify differentially expressed genes (DEGs) in OSCC samples compared with control samples. Overlapping genes between DEGs and DMGs were identified, and functional enrichment analysis was performed. In addition, survival analysis of the overlapping genes was performed to screen genes with prognostic significance in OSCC. A total of 40,115 differential methylation CpG sites spanning 3,360 DMGs were identified; CpG sites in the promoter, gene body and intergenic regions were generally highly hypermethylated or hypomethylated. Additionally, 508 DEGs in OSCC samples were identified, including 332 upregulated and 176 downregulated genes. A total of 82 overlapping genes between DEGs and DMGs were found, which were mainly involved in protein metabolism, regulation of the metabolic process and the immune system. Additionally, differential methylation or expression of several genes, including fibroblast activation protein α (FAP), interferon α inducible protein 27 (IFI27), laminin subunit γ2 (LAMC2), matrix metallopeptidase 1 (MMP1), serine peptidase inhibitor Kazal-type 5 (SPINK5) and zinc finger protein 662 (ZNF662), was significantly associated with the survival of OSCC patients, and their differential expression in OSCC patients was further confirmed by reverse transcription-quantitative polymerase chain reaction in OSCC and normal oral cell lines. Overall, FAP, IFI27, LAMC2, MMP1, SPINK5 and ZNF662 genes caused by epigenetic changes via DNA methylation may be associated with the development and progression of OSCC, and should be valuable OSCC therapeutic biomarkers.
Collapse
Affiliation(s)
- Chenguang Zhao
- Department of Emergency, Tianjin Stomatological Hospital, Tianjin 300041, P.R. China
| | - Huiru Zou
- Central Laboratory, Tianjin Stomatological Hospital, Tianjin 300041, P.R. China
| | - Jun Zhang
- Department of Oral and Maxillofacial Surgery, Tianjin Stomatological Hospital, Tianjin 300041, P.R. China
| | - Jinhui Wang
- Department of Emergency, Tianjin Stomatological Hospital, Tianjin 300041, P.R. China
| | - Hao Liu
- Department of Oral and Maxillofacial Surgery, Tianjin Stomatological Hospital, Tianjin 300041, P.R. China
| |
Collapse
|
28
|
Luo JJ, Young CD, Zhou HM, Wang XJ. Mouse Models for Studying Oral Cancer: Impact in the Era of Cancer Immunotherapy. J Dent Res 2018; 97:683-690. [PMID: 29649368 DOI: 10.1177/0022034518767635] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Model systems for oral cancer research have progressed from tumor epithelial cell cultures to in vivo systems that mimic oral cancer genetics, pathological characteristics, and tumor-stroma interactions of oral cancer patients. In the era of cancer immunotherapy, it is imperative to use model systems to test oral cancer prevention and therapeutic interventions in the presence of an immune system and to discover mechanisms of stromal contributions to oral cancer carcinogenesis. Here, we review in vivo mouse model systems commonly used for studying oral cancer and discuss the impact these models are having in advancing basic mechanisms, chemoprevention, and therapeutic intervention of oral cancer while highlighting recent discoveries concerning the role of immune cells in oral cancer. Improvements to in vivo model systems that highly recapitulate human oral cancer hold the key to identifying features of oral cancer initiation, progression, and invasion as well as molecular and cellular targets for prevention, therapeutic response, and immunotherapy development.
Collapse
Affiliation(s)
- J J Luo
- 1 State Key Laboratory of Oral Diseases, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,2 Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - C D Young
- 2 Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - H M Zhou
- 1 State Key Laboratory of Oral Diseases, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - X J Wang
- 2 Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
29
|
Oballe HJR, Muniz FWMG, Bueno CC, Klein IP, Carrard VC, Rösing CK, Gaio EJ. Spontaneous alveolar bone loss after 4NQO exposure in Wistar rats. Arch Oral Biol 2018; 89:44-48. [PMID: 29448184 DOI: 10.1016/j.archoralbio.2018.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 01/22/2018] [Accepted: 02/04/2018] [Indexed: 11/24/2022]
Abstract
OBJECTIVE This study evaluated the effect of an experimental carcinogenic, 4-Nitroquinoline 1-oxide (4NQO), in the spontaneous alveolar bone loss (ABL) in an animal model. DESIGN Twenty-two male Wistar rats were included in this study. They were randomly divided into two groups: the control group (n = 10) received food and water ad libitum, and the test group (n = 12) receive the same food; however, 25 ppm of 4NQO was diluted in the drinking water. All animals were euthanized after 20 weeks, and the tongues were removed and analyzed macroscopically to determine the presence of oral mucosal lesions. All specimens were paraffin-embedded and histological sections were obtained. The microscopic analysis was based on routine procedure (haematoxylin and eosin stain). The analysis of spontaneous ABL was performed by a calibrated examiner using standardized photographs and imaging software. Differences in spontaneous ABL were assessed among the three resulting groups: control, 4NQO with oral squamous cell carcinoma (OSCC), and 4NQO without OSCC. RESULTS In the 4NQO-treated group, nine animals developed OSCC. The animals in the 4NQO with OSCC group presented significantly more spontaneous ABL (0.65 ± 0.21 mm) than the control group (0.34 ± 0.05) (p < 0.001). The animals in the 4NQO without OSCC group showed a mean spontaneous ABL of 0.47 ± 0.13 mm, which was not statistically significant different when compared to the control group (p = 0.096). CONCLUSIONS It was concluded that the presence of OSCC enhanced spontaneous ABL in Wistar rats when compared to control animals. Additionally, it was shown that, solely, administration of 4NQO may not be considered responsible for alveolar bone destruction.
Collapse
Affiliation(s)
- Harry J R Oballe
- Department of Periodontology, Faculty of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil; Rua Ramiro Barcelos, 2492, Porto Alegre, Rio Grande do Sul, Zip code: 90035-003, Brazil.
| | - Francisco Wilker M G Muniz
- Department of Periodontology, Faculty of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil; Rua Ramiro Barcelos, 2492, Porto Alegre, Rio Grande do Sul, Zip code: 90035-003, Brazil.
| | - Cheyenne C Bueno
- Rua Ramiro Barcelos, 2492, Porto Alegre, Rio Grande do Sul, Zip code: 90035-003, Brazil; Department of Oral Pathology, Faculty of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.
| | - Isadora P Klein
- Rua Ramiro Barcelos, 2492, Porto Alegre, Rio Grande do Sul, Zip code: 90035-003, Brazil; Department of Oral Pathology, Faculty of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.
| | - Vinicius C Carrard
- Rua Ramiro Barcelos, 2492, Porto Alegre, Rio Grande do Sul, Zip code: 90035-003, Brazil; Department of Oral Pathology, Faculty of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.
| | - Cassiano K Rösing
- Department of Periodontology, Faculty of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil; Rua Ramiro Barcelos, 2492, Porto Alegre, Rio Grande do Sul, Zip code: 90035-003, Brazil.
| | - Eduardo J Gaio
- Department of Periodontology, Faculty of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil; Rua Ramiro Barcelos, 2492, Porto Alegre, Rio Grande do Sul, Zip code: 90035-003, Brazil.
| |
Collapse
|
30
|
Oghumu S, Casto BC, Ahn-Jarvis J, Weghorst LC, Maloney J, Geuy P, Horvath KZ, Bollinger CE, Warner BM, Summersgill KF, Weghorst CM, Knobloch TJ. Inhibition of Pro-inflammatory and Anti-apoptotic Biomarkers during Experimental Oral Cancer Chemoprevention by Dietary Black Raspberries. Front Immunol 2017; 8:1325. [PMID: 29109723 PMCID: PMC5660285 DOI: 10.3389/fimmu.2017.01325] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 09/29/2017] [Indexed: 01/12/2023] Open
Abstract
Oral cancer continues to be a significant public health problem worldwide. Recently conducted clinical trials demonstrate the ability of black raspberries (BRBs) to modulate biomarkers of molecular efficacy that supports a chemopreventive strategy against oral cancer. However, it is essential that a preclinical animal model of black raspberry (BRB) chemoprevention which recapitulates human oral carcinogenesis be developed, so that we can validate biomarkers and evaluate potential mechanisms of action. We therefore established the ability of BRBs to inhibit oral lesion formation in a carcinogen-induced rat oral cancer model and examined potential mechanisms. F344 rats were administered 4-nitroquinoline 1-oxide (4NQO) (20 µg/ml) in drinking water for 14 weeks followed by regular drinking water for 6 weeks. At week 14, rats were fed a diet containing either 5 or 10% BRB, or 0.4% ellagic acid (EA), a BRB phytochemical. Dietary administration of 5 and 10% BRB reduced oral lesion incidence and multiplicity by 39.3 and 28.6%, respectively. Histopathological analyses demonstrate the ability of BRBs and, to a lesser extent EA, to inhibit the progression of oral cancer. Oral lesion inhibition by BRBs was associated with a reduction in the mRNA expression of pro-inflammatory biomarkers Cxcl1, Mif, and Nfe2l2 as well as the anti-apoptotic and cell cycle associated markers Birc5, Aurka, Ccna1, and Ccna2. Cellular proliferation (Ki-67 staining) in tongue lesions was inhibited by BRBs and EA. Our study demonstrates that, in the rat 4NQO oral cancer model, dietary administration of BRBs inhibits oral carcinogenesis via inhibition of pro-inflammatory and anti-apoptotic pathways.
Collapse
Affiliation(s)
- Steve Oghumu
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University Columbus, Columbus, OH, United States.,Comprehensive Cancer Center, The Ohio State University Columbus, Columbus, OH, United States
| | - Bruce C Casto
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University Columbus, Columbus, OH, United States.,Comprehensive Cancer Center, The Ohio State University Columbus, Columbus, OH, United States
| | - Jennifer Ahn-Jarvis
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University Columbus, Columbus, OH, United States
| | - Logan C Weghorst
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University Columbus, Columbus, OH, United States
| | - Jim Maloney
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University Columbus, Columbus, OH, United States
| | - Paul Geuy
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University Columbus, Columbus, OH, United States
| | - Kyle Z Horvath
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University Columbus, Columbus, OH, United States
| | - Claire E Bollinger
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University Columbus, Columbus, OH, United States
| | - Blake M Warner
- School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kurt F Summersgill
- School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Christopher M Weghorst
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University Columbus, Columbus, OH, United States.,Comprehensive Cancer Center, The Ohio State University Columbus, Columbus, OH, United States.,Department of Otolaryngology, College of Medicine, The Ohio State University Columbus, Columbus, OH, United States
| | - Thomas J Knobloch
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University Columbus, Columbus, OH, United States.,Comprehensive Cancer Center, The Ohio State University Columbus, Columbus, OH, United States
| |
Collapse
|