CD47-SIRPα Signaling Induces Epithelial-Mesenchymal Transition and Cancer Stemness and Links to a Poor Prognosis in Patients with Oral Squamous Cell Carcinoma
"> Figure 1
CD47 is aberrantly expressed in human oral squamous cell carcinoma and influence survival rate. (A) CD47 transcript expression profile across TCGA and GTEx paired normal-tumor tissue cohort. (B) The expression of CD47 in downloaded data for OSCC based on morphology, anatomic site, and sample type from the Genomic Data Commons-The Cancer Genome Atlas (GDC TGCA) HNSCC dataset. (C) Differential expression of CD47 in normal oral and cancer tissues in TCGA OSCC cohort (n = 412; p = 0.0009). (D) Kaplan–Meier curves showing the effect of low and high CD47 expression on the overall survival of the TGCA malignant OSCC cohort. OSCC: oral squamous cell carcinoma; GTEx: genotype-tissue expression; HNSCC: head and neck squamous cell carcinoma; GDC: genome data commons; TCGA: the cancer genome atlas.
"> Figure 2The aberrant expression of CD47 in oral squamous cell carcinoma positively correlates with disease progression. (A) Representative immunohistochemistry staining of CD47 in human normal oral and OSCC tissues. (B) Pie chart showing the distribution of patients in our OSCC cohort (n = 71) based on histological types. (C) Graphical representation of the histology-specific relative expression of CD47 in tissue samples from our OSCC cohort. CD47 tissue expression is relative to that in the normal group. ns, not significant; * p < 0.05, ** p < 0.01, *** p < 0.001.
"> Figure 3CD47 modulates the cancer stem cell-like phenotype in oral squamous cell carcinoma cells. (A) Box and whiskers chart showing the correlative differential expression of CD47 (upper), SOX2 (middle), and CD133 (lower) mRNA from analyses of the human OSCC genome U133A array from the Toruner head-neck cohort, n = 20. (B) Knockdown efficiency of shCD47-1 and shCD47-2 on the protein expression of CD47 in SAS and TW2.6 cell lines shown by western blot analysis. (C) Effect of CD47 knockdown on the expression level of CD47, SOX2, OCT4, and CD133 proteins in SAS Sp, shCD47-1, or shCD47-2 cells shown by western blot analysis. GAPDH served as a loading control. (D) Immunofluorescent staining showing the effect of shCD47 on the expression of CD47, OCT4, c-Myc, and SOX2 proteins in spheres formed by TW2.6 cells. TW2.6 and SAS cells transfected with shCD47-1 or shCD47-2 exhibited decreased orosphere size (left) and number (right) in both (E) primary and (F) secondary generation orospheres. (G) shCD47 attenuated OCT4 and SOX2 expression and inhibited their nuclear co-localization in TW2.6- or SAS-derived orospheres, as shown by immunofluorescent (IFC) staining. All assays are representative of experiments performed four times in triplicates. WT, wild type; Sp, orosphere; blue stain = DAPI, nuclear staining. All data are representative of experiment carried out four times in triplicate and are expressed as mean ± S.D. * p < 0.05, ** p < 0.01, *** p < 0.001.
"> Figure 4Downregulation of CD47 attenuates the EMT (epithelial-to-mesenchymal transition) and migration capacity of OSCC cells. (A) Representative image of scratch-wound migration assay shows the effect of shCD47 on the motility of SAS cells at 0, 3, 6, and 9-h time points (upper), and quantitative bar chart of the migrating cell fronts at indicated time points (lower). (B) Representative images of colony formed by WT, shCD47-1, or shCD47-2 transfected SAS cells in the culture plate using crystal violet solution and quantification of visible cells. (C) The inhibitory effect of shCD47 on the expression of CD47, vimentin, Slug, Snail, N-cadherin, and E-cadherin in SAS cells, as demonstrated by western blot analyses. (D) Photo-image, showing the fibroid/spindle shape of CD47-expressing WT cells, while shCD47 led to the loss of mesenchymal phenotype in SAS (upper) and TW2.6 (lower) cells. WT, wild type; GAPDH served as a loading control. All data are representative of experiment carried out four times in triplicate and are expressed as mean ± S.D. * p < 0.05, ** p < 0.01, *** p < 0.001.
"> Figure 5CD47 modulates the expression and subcellular localization of mesenchymal and epithelial factors in OSCC. (A,B) Immunofluorescent staining images, showing the expression of CD47, vimentin, and E-cadherin in SAS WT, and shCD47-1 or shCD47-2 cells. Representative dot plot of the correlative expression of (C) CD47 mRNA versus vimentin mRNA or (D) CD47 mRNA versus CDH1 mRNA in OSCC patients from the Roepman cohort, n = 220 using the R2 online genomic analysis and visualization software. (E) The associative network of the interaction between CD47 and molecular moieties involved in stem cell development, stem cell differentiation, stem cell maintenance, positive regulation of cell migration, and positive regulation of cell motility. Sp, orosphere; DAPI, nuclear staining.
"> Figure 6Suppression of CD47 expression enhances the sensitivity of OSCC-SCs to radiation therapy. (A) Bar chart of the inhibitory effect of exposure to 0 Gy–15 Gy radiation on the viability of SAS or TW2.6 cells. (B) shCD47 with or without 5 Gy–15 Gy radiation decreased the viability of SAS cells dose-dependently. (C) Transwell migration assay images show reduced migration in 5 Gy-exposed shCD47 SAS cells, compared to their SAS WT counterparts. (D) Transwell invasion assay images show reduced invasion in 5 Gy-exposed shCD47 SAS cells, compared to their SAS WT counterparts. (E) shCD47-transfected cells yielded smaller and fewer tumorspheres compared to their WT or shCD47 scramble counterparts. (F) shCD47-1 or shCD47-2 SAS cells formed fewer colonies when exposed to 5 Gy, compared to the SAS WT cells. Data represent mean ± SD from three independent experiments performed in triplicates. * p < 0.05, ** p < 0.01, and *** p < 0.001.
"> Figure 7Schema showing CD47-related molecular network and how the suppression of the ”don’t-eat-me” signal CD47 enhances radiosensitivity by downregulating cancer stem cells-associated pluripotency factors and deactivating epithelial-to-mesenchymal transition in oral squamous cell carcinoma cells.
">
Abstract
Background: Oral squamous cell carcinoma (OSCC), with high mortality rates, is one of the most diagnosed head and neck cancers. Epithelial-to-mesenchymal transition (EMT) and the generation of cancer stem cells (CSCs) are two keys for therapy-resistance, relapse, and distant metastasis. Accumulating evidence indicates that aberrantly expressed cluster of differentiation (CD)47 is associated with cell-death evasion and metastasis; however, the role of CD47 in the generation of CSCs in OSCC is not clear. Methods: We investigated the functional roles of CD47 in OSCC cell lines SAS, TW2. 6, HSC-3, and FaDu using the bioinformatics approach, immunoblotting, immunofluorescence staining, and assays for cellular migration, invasion, colony, and orosphere formation, as well as radiosensitivity. Results: We demonstrated increased expression of CD47 in OSCC patients was associated with an estimated poorly survival disadvantage (p = 0. 0391) and positively correlated with the expression of pluripotency factors. Silencing CD47 significantly suppressed cell viability and orosphere formation, accompanied by a downregulated expression of CD133, SRY-Box transcription factor 2 (SOX2), octamer-binding transcription factor 4 (OCT4), and c-Myc. In addition, CD47-silenced OSCC cells showed reduced EMT, migration, and clonogenicity reflected by increased E-cadherin and decreased vimentin, Slug, Snail, and N-cadherin expression V体育官网入口. Conclusion: Of therapeutic relevance, CD47 knockdown enhanced the anti-OSCC effect of radiotherapy. Collectively, we showed an increased CD47 expression promoted the generation of CSCs and malignant OSCC phenotypes. Silencing CD47, in combination with radiation, could provide an alternative and improved therapeutic efficacy for OSCC patients. Keywords: chemoradiation; oral cancer stem cells; CD47; radiotherapy techniques; radioresistance .1. Introduction
2. Materials and Methods
2.1. Patient Samples
"VSports注册入口" 2.2. Immunohistochemical (IHC) Staining
2.3. Reagents
"V体育平台登录" 2.4. Cell Lines and Culture
"V体育官网" 2.5. shRNA Transfection of OSCC Cell Lines
"VSports" 2.6. Orosphere Formation and Self-Renewal Assay
2.7. Colony Formation Assay
2.8. Radiation and Cell Viability Assay
2.9. Western Blot Analysis
2.10. Wound Healing Migration Assay
2.11. Matrigel Invasion and Migration Assay
2.12. Immunofluorescence Staining
2.13. Statistical Analysis
3. Results
3.1. CD47 Is Aberrantly Expressed in Human Oral Squamous Cell Carcinoma and Influence Survival Rate
"V体育安卓版" 3.2. The Aberrant Expression of CD47 in Human Oral Squamous Cell Carcinoma Tissue Positively Correlates with Disease Progression
3.3. CD47 Modulates the Cancer Stem Cell-Like Phenotype and Self-Renewal in Oral Squamous Cell Carcinoma Cells
3.4. Downregulation of CD47 Attenuate the EMT and Migration Capacity of OSCC Cells (V体育官网入口)
3.5. CD47 Modulates the Expression and Subcellular Localization of Mesenchymal and Epithelial Factors in OSCC
3.6. Suppression of CD47 Expression Enhances the Sensitivity of OSCC-SCs to Radiation Therapy
4. Discussion
V体育ios版 - Supplementary Materials
Author Contributions
V体育官网入口 - Funding
Conflicts of Interest (VSports在线直播)
"VSports手机版" References
- Shield, K.D.; Ferlay, J.; Jemal, A.; Sankaranarayanan, R.; Chaturvedi, A.K.; Bray, F.; Soerjomataram, I. The global incidence of lip, oral cavity, and pharyngeal cancers by subsite in 2012. CA Cancer J. Clin. 2017, 67, 51–64. [Google Scholar] [CrossRef] [PubMed]
- Ferlay, J.; Soerjomataram, I.; Dikshit, R.; Eser, S.; Mathers, C.; Rebelo, M.; Parkin, D.M.; Forman, D.; Bray, F. Cancer incidence and mortality world-wide: Sources, methods and major patterns in GLOBOCAN 2012. Int. J. Cancer 2015, 136, E359–E386. [Google Scholar] [CrossRef] [PubMed]
- Ou, D.; Blanchard, P.; El Khoury, C.; De Felice, F.; Even, C.; Levy, A.; Nguyen, F.; Janot, F.; Gorphe, P.; Deutsch, E.; et al. Induction chemotherapy with docetaxel, cisplatin and fluorouracil followed by concurrent chemoradiotherapy or chemoradiotherapy alone in locally advanced non-endemic nasopharyngeal carcinoma. Oral Oncol. 2016, 62, 114–121. [Google Scholar] [CrossRef] [PubMed]
- Tangthongkum, M.; Kirtsreesakul, V.; Supanimitjaroenporn, P.; Leelasawatsuk, P. Treatment outcome of advance staged oral cavity cancer: Concurrent chemoradiotherapy compared with primary surgery. Eur. Arch. Otorhinolaryngol. 2017. ["V体育安卓版" Google Scholar] [CrossRef]
- Palve, V.C.; Teni, T.R. Association of anti-apoptotic Mcl-1L isoform expression with radioresistance of oral squamous carcinoma cells. Radiat. Oncol. 2012, 7, 135. ["VSports app下载" Google Scholar] [CrossRef]
- Maccalli, C.; de Maria, R. Cancer stem cells: Perspectives for therapeutic targeting. Cancer Immunol. Immunother. CII 2015, 64, 91–97. [Google Scholar] [CrossRef]
- Ajani, J.A.; Song, S.; Hochster, H.S.; Steinberg, I.B. Cancer stem cells: The promise and the potential. Semin. Oncol. 2015, 42 (Suppl. 1), S3–S17. [Google Scholar] [CrossRef]
- Lin, C.S.; Lin, Y.C.; Bamodu, O.A.; Wu, A.; Chen, J.H.; Peng, Y.J.; Cheng, M.F.; Lee, W.H.; Hsiao, M.; Chao, T.Y.; et al. Silencing JARID1B suppresses oncogenicity, stemness and increases radiation sensitivity in human oral carcinoma. Cancer Lett. 2015, 368, 36–45. [Google Scholar] [CrossRef]
- Liu, S.C.; Huang, C.M.; Bamodu, O.A.; Lin, C.S.; Liu, B.L.; Tzeng, Y.M.; Tsai, J.T.; Lee, W.H.; Chen, T.M. Ovatodiolide suppresses nasopharyngeal cancer by targeting stem cell-like population, inducing apoptosis, inhibiting EMT and dysregulating JAK/STAT signaling pathway. Phytomedicine 2018. [VSports在线直播 - Google Scholar] [CrossRef]
- Liu, X.; Fan, D. The epithelial-mesenchymal transition and cancer stem cells: Functional and mechanistic links. Curr. Pharm. Des. 2015, 21, 1279–1291. [Google Scholar] [CrossRef]
- Sato, R.; Semba, T.; Saya, H.; Arima, Y. Concise Review: Stem Cells and Epithelial-Mesenchymal Transition in Cancer: Biological Implications and Therapeutic Targets. Stem Cells 2016, 34, 1997–2007. [Google Scholar] [CrossRef] [PubMed]
- Ye, X.; Weinberg, R.A. Epithelial-Mesenchymal Plasticity: A Central Regulator of Cancer Progression. Trends Cell Biol. 2015, 25, 675–686. [Google Scholar] [CrossRef] [PubMed]
- Shibue, T.; Weinberg, R.A. EMT, CSCs, and drug resistance: The mechanistic link and clinical implications. Nat. Rev. Clin. Oncol. 2017, 14, 611–629. [V体育安卓版 - Google Scholar] [CrossRef] [PubMed]
- Zhao, H.; Wang, J.; Kong, X.; Li, E.; Liu, Y.; Du, X.; Kang, Z.; Tang, Y.; Kuang, Y.; Yang, Z.; et al. CD47 Promotes Tumor Invasion and Metastasis in Non-small Cell Lung Cancer. Sci. Rep. 2016, 6, 29719. [Google Scholar] [CrossRef] [PubMed]
- Huang, Y.; Ma, Y.; Gao, P.; Yao, Z. Targeting CD47: The achievements and concerns of current studies on cancer immunotherapy. J. Thorac. Dis. 2017, 9, E168–E174. [VSports手机版 - Google Scholar] [CrossRef]
- Poole, C.J.; van Riggelen, J. MYC—Master Regulator of the Cancer Epigenome and Transcriptome. Genes 2017, 8, 142. ["VSports手机版" Google Scholar] [CrossRef]
- Casey, S.C.; Baylot, V.; Felsher, D.W. MYC: Master Regulator of Immune Privilege. Trends Immunol. 2017, 38, 298–305. [Google Scholar] [CrossRef]
- Baillie, R.; Tan, S.T.; Itinteang, T. Cancer Stem Cells in Oral Cavity Squamous Cell Carcinoma: A Review. Front. Oncol. 2017, 7, 112. ["VSports最新版本" Google Scholar] [CrossRef]
- Sawant, S.; Gokulan, R.; Dongre, H.; Vaidya, M.; Chaukar, D.; Prabhash, K.; Ingle, A.; Joshi, S.; Dange, P.; Joshi, S.; et al. Prognostic role of Oct4, CD44 and c-Myc in radio-chemo-resistant oral cancer patients and their tumourigenic potential in immunodeficient mice. Clin. Oral Investig. 2016, 20, 43–56. [Google Scholar] [CrossRef]
- Shrivastava, S.; Steele, R.; Sowadski, M.; Crawford, S.E.; Varvares, M.; Ray, R.B. Identification of molecular signature of head and neck cancer stem-like cells. Sci. Rep. 2015, 5, 7819. ["V体育官网入口" Google Scholar] [CrossRef]
- Felthaus, O.; Ettl, T.; Gosau, M.; Driemel, O.; Brockhoff, G.; Reck, A.; Zeitler, K.; Hautmann, M.; Reichert, T.E.; Schmalz, G.; et al. Cancer stem cell-like cells from a single cell of oral squamous carcinoma cell lines. Biochem. Biophys. Res. Commun. 2011, 407, 28–33. [Google Scholar] [CrossRef] [PubMed]
- Ye, X.; Wang, X.; Lu, R.; Zhang, J.; Chen, X.; Zhou, G. CD47 as a potential prognostic marker for oral leukoplakia and oral squamous cell carcinoma. Oncol. Lett. 2018, 15, 9075–9080. ["V体育安卓版" Google Scholar] [CrossRef] [PubMed]
- Chao, M.P.; Weissman, I.L.; Majeti, R. The CD47-SIRPα pathway in cancer immune evasion and potential therapeutic implications. Curr. Opin. Immunol. 2012, 24, 225–232. [Google Scholar] [CrossRef] [PubMed]
- Smolle, M.A.; Pichler, M. Inflammation, phagocytosis and cancer: Another step in the CD47 act. J. Thorac. Dis. 2017, 9, 2279–2282. [Google Scholar] [CrossRef]
- Koh, E.; Lee, E.J.; Nam, G.H.; Hong, Y.; Cho, E.; Yang, Y.; Kim, I.S. Exosome-SIRPα, a CD47 blockade increases cancer cell phagocytosis. Biomaterials 2017, 121, 121–129. [Google Scholar] [CrossRef]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. ["V体育平台登录" Google Scholar] [CrossRef]
- Enderling, H.; Hahnfeldt, P. Cancer stem cells in solid tumors: Is ‘evading apoptosis’ a hallmark of cancer? Prog. Biophys. Mol. Biol. 2011, 106, 391–399. [Google Scholar] [CrossRef]
- Liu, L.; Zhang, L.; Yang, L.; Li, H.; Li, R.; Yu, J.; Yang, L.; Wei, F.; Yan, C.; Sun, Q.; et al. Anti-CD47 Antibody As a Targeted Therapeutic Agent for Human Lung Cancer and Cancer Stem Cells. Front. Immunol. 2017, 8, 404. ["V体育2025版" Google Scholar] [CrossRef]
- Liu, R.; Wei, H.; Gao, P.; Yu, H.; Wang, K.; Fu, Z.; Ju, B.; Zhao, M.; Dong, S.; Li, Z.; et al. CD47 promotes ovarian cancer progression by inhibiting macrophage phagocytosis. Oncotarget 2017, 8, 39021–39032. [Google Scholar (V体育2025版)] [CrossRef]
- Baranwal, S.; Alahari, S.K. Molecular mechanisms controlling E-cadherin expression in breast cancer. Biochem. Biophys. Res. Commun. 2009, 384, 6–11. ["VSports" Google Scholar] [CrossRef]
- Costa, L.C.M.C.; Leite, C.F.; Cardoso, S.V.; Loyola, A.M.; de Faria, P.R.; Souza, P.E.A.; Horta, M.C.R. Expression of epithelial-mesenchymal transition markers at the invasive front of oral squamous cell carcinoma. J. Appl. Oral Sci. 2015, 23, 169–178. [Google Scholar] [CrossRef] [PubMed]
- Kim, D.H.; Xing, T.; Yang, Z.; Dudek, R.; Lu, Q.; Chen, Y.H. Epithelial Mesenchymal Transition in Embryonic Development, Tissue Repair and Cancer: A Comprehensive Overview. J. Clin. Med. 2017, 7, 1. [Google Scholar] [CrossRef] [PubMed]
- Huang, S.H.; O’Sullivan, B. Oral cancer: Current role of radiotherapy and chemotherapy. Med. Oral Patol. Oral Cir. Bucal. 2013, 18, e233–e240. [Google Scholar (V体育官网)] [CrossRef] [PubMed]
- Glenny, A.M.; Furness, S.; Worthington, H.V.; Conway, D.I.; Oliver, R.; Clarkson, J.E.; Macluskey, M.; Pavitt, S.; Chan, K.K.; Brocklehurst, P.; et al. Interventions for the treatment of oral cavity and oropharyngeal cancer: Radiotherapy. Cochrane Database Syst. Rev. 2010, CD006387. [Google Scholar] [CrossRef] [PubMed]
- Gillison, M.L.; Koch, W.M.; Capone, R.B.; Spafford, M.; Westra, W.H.; Wu, L.; Zahurak, M.L.; Daniel, R.W.; Viglione, M.; Symer, D.E.; et al. Evidence for a causal association between human papillomavirus and a subset of head and neck cancers. J. Natl. Cancer Inst. 2000, 92, 709–720. [Google Scholar] [CrossRef]
- Van Houten, V.M.; Snijders, P.J.; van Den Brekel, M.W.; Kummer, J.A.; Meijer, C.J.; van Leeuwen, B.; Denkers, F.; Smeele, L.E.; Snow, G.B.; Brakenhoff, R.H. Biological evidence that human papillomaviruses are etiologically involved in a subgroup of head and neck squamous cell carcinomas. Int. J. Cancer 2001, 93, 232–235. [Google Scholar] [CrossRef]
- Wang, J.; Sun, H.; Zeng, Q.; Guo, X.-J.; Wang, H.; Liu, H.-H.; Dong, Z.-Y. HPV-positive status associated with inflamed immune microenvironment and improved response to anti-PD-1 therapy in head and neck squamous cell carcinoma. Sci. Rep. 2019, 9, 13404. [Google Scholar] [CrossRef]
- Ludwig, S.; Marczak, L.; Sharma, P.; Abramowicz, A.; Gawin, M.; Widlak, P.; Whiteside, T.L.; Pietrowska, M. Proteomes of exosomes from HPV(+) or HPV(−) head and neck cancer cells: Differential enrichment in immunoregulatory proteins. Oncoimmunology 2019, 8, 1593808. [Google Scholar (V体育平台登录)] [CrossRef]
- Vermeer, D.W.; Spanos, W.C.; Vermeer, P.D.; Bruns, A.M.; Lee, K.M.; Lee, J.H. Radiation-induced loss of cell surface CD47 enhances immune-mediated clearance of human papillomavirus-positive cancer. Int. J. Cancer 2013, 133, 120–129. [Google Scholar] [CrossRef]
- Golden, E.B.; Apetoh, L. Radiotherapy and immunogenic cell death. Semin. Radiat. Oncol. 2015, 25, 11–17. [Google Scholar] [CrossRef]







| Variables | N |
|---|---|
| Age [median (range)] | 48 (29–72) |
| Sex (M/F) | 62/9 |
| Total | 71 |
| Follow-up | |
| Range (days) | 161–3645 |
| AJCC* staging | |
| Early (I+II) | 31 |
| Late (III+IV) | 40 |
| Tumor | |
| T1 | 13 |
| T2 | 18 |
| T3 | 21 |
| T4 | 19 |
| Nodes | |
| N (−) | 34 |
| N (+) | 37 |
| Clinicopathological Variables | Univariate | Multivariate# | ||||
|---|---|---|---|---|---|---|
| HR | 95%CI | p-Value | HR | 95%CI | p-Value | |
| Age, years (≦mean vs. > mean) | 0.97 | 0.41–2.64 | 0.58 | |||
| Sex (Male vs. Female) | 0.55 | 0.03–1.78 | 0.42 | |||
| Tumor (T1 + T2 vs. T3 + T4) | 1.27 | 0.52–3.59 | 0.36 | |||
| Nodes (N1 + N2 + N3 vs. N0) | 3.25 | 1.27–5.16 | 0.001 | 1.73 | 0.51–3.19 | 0.817 |
| Pathological grade (grades 2 + 3 vs. grade 1) | 1.35 | 0.67–3.02 | 0.28 | |||
| Local recurrence (Yes vs. No) | 3.82 | 1.16–7.68 | 0.003 | 4.26 | 1.98–9.28 | 0.031 |
| CD47 expression (high vs. low) | 6.83 | 1.72–18.09 | 0.01 | 5.18 | 0.73–12.64 | 0.019 |
| AJCC* Staging (III + IV vs. I + II) | 7.67 | 1.90–15.37 | 0.002 | 3.42 | 1.09–9.85 | 0.048 |
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
"V体育官网" Share and Cite
Pai, S.; Bamodu, O.A.; Lin, Y.-K.; Lin, C.-S.; Chu, P.-Y.; Chien, M.-H.; Wang, L.-S.; Hsiao, M.; Yeh, C.-T.; Tsai, J.-T. CD47-SIRPα Signaling Induces Epithelial-Mesenchymal Transition and Cancer Stemness and Links to a Poor Prognosis in Patients with Oral Squamous Cell Carcinoma. Cells 2019, 8, 1658. https://doi.org/10.3390/cells8121658
Pai S, Bamodu OA, Lin Y-K, Lin C-S, Chu P-Y, Chien M-H, Wang L-S, Hsiao M, Yeh C-T, Tsai J-T. CD47-SIRPα Signaling Induces Epithelial-Mesenchymal Transition and Cancer Stemness and Links to a Poor Prognosis in Patients with Oral Squamous Cell Carcinoma. Cells. 2019; 8(12):1658. https://doi.org/10.3390/cells8121658
Chicago/Turabian StylePai, Shin, Oluwaseun Adebayo Bamodu, Yen-Kuang Lin, Chun-Shu Lin, Pei-Yi Chu, Ming-Hsien Chien, Liang-Shun Wang, Michael Hsiao, Chi-Tai Yeh, and Jo-Ting Tsai. 2019. "CD47-SIRPα Signaling Induces Epithelial-Mesenchymal Transition and Cancer Stemness and Links to a Poor Prognosis in Patients with Oral Squamous Cell Carcinoma" Cells 8, no. 12: 1658. https://doi.org/10.3390/cells8121658
APA StylePai, S., Bamodu, O. A., Lin, Y.-K., Lin, C.-S., Chu, P.-Y., Chien, M.-H., Wang, L.-S., Hsiao, M., Yeh, C.-T., & Tsai, J.-T. (2019). CD47-SIRPα Signaling Induces Epithelial-Mesenchymal Transition and Cancer Stemness and Links to a Poor Prognosis in Patients with Oral Squamous Cell Carcinoma. Cells, 8(12), 1658. https://doi.org/10.3390/cells8121658

