201
|
The molecular mechanisms underlying the therapeutic resistance of cancer stem cells. Arch Pharm Res 2014; 38:389-401. [DOI: 10.1007/s12272-014-0531-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 12/08/2014] [Indexed: 12/19/2022]
|
202
|
Abstract
The cancer stem cell (CSC) hypothesis postulates that there is a hierarchy of cellular differentiation within cancers and that the bulk population of tumor cells is derived from a relatively small population of multi-potent neoplastic stem-like cells (CSCs). This tumor-initiating cell population plays an important role in maintaining tumor growth through their unlimited self-renewal, therapeutic resistance, and capacity to propagate tumors through asymmetric cell division. Recent findings from multiple laboratories show that cancer progenitor cells have the capacity to de-differentiate and acquire a stem-like phenotype in response to either genetic manipulation or environmental cues. These findings suggest that CSCs and relatively differentiated progenitors coexist in dynamic equilibrium and are subject to bidirectional conversion. In this review, we discuss emerging concepts regarding the stem-like phenotype, its acquisition by cancer progenitor cells, and the molecular mechanisms involved. Understanding the dynamic equilibrium between CSCs and cancer progenitor cells is critical for the development of novel therapeutic strategies that focus on depleting tumors of their tumor-propagating cell population.
Collapse
Affiliation(s)
| | - Yunqing Li
- Hugo W. Moser Research Institute at Kennedy Krieger, USA; Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - John Laterra
- Hugo W. Moser Research Institute at Kennedy Krieger, USA; Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
203
|
Chou MY, Hu FW, Yu CH, Yu CC. Sox2 expression involvement in the oncogenicity and radiochemoresistance of oral cancer stem cells. Oral Oncol 2014; 51:31-9. [PMID: 25456004 DOI: 10.1016/j.oraloncology.2014.10.002] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 09/17/2014] [Accepted: 10/06/2014] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Sox2, a high-mobility-group DNA binding protein, is part of the key set of transcription factors that are involved in the maintenance of pluripotency and self-renewal in undifferentiated stem cells. A recent study has further suggested cancer stem cells (CSCs) are key contributors to radiochemoresistance and are responsible for oral squamous cell carcinoma (OSCC) progression. The aim of this study was to determine the emerging role of Sox2 in radiochemosensitivity of oral CSCs. METHODS We determined the function of Sox2 on oncogenicity and radiochemosensitivity of OSCC by overexpression or silencing Sox2 in vitro and in vivo. RESULTS Initially, Sox2 expression was increased in OSCC cell lines and OSCC specimens. Upregulated Sox2 is correlated with poor survival outcome of OSCC patients. Overexpression of Sox2 was demonstrated to enhance invasiveness, anchorage-independent growth, xenotransplantation tumourigenicity in OSCC cells. Targeting Sox2 to spheroid cells (SC) and ALDH1+CD44+ cells from OSCC significantly inhibited their CSCs and tumorigenic abilities. Down regulation of SOX2 in OSCC-SC was found to repress invasiveness and diminish epithelail-mesenchymal transition (EMT) traits. Furthermore, silencing Sox2 effectively suppressed the expression of drug-resistance and anti-apoptotic genes and increased the sensitivity of the cells to radiation combined cisplatin treatment. Finally, the in vivo therapeutic efficacy of targeting Sox2 synergistically suppressed tumorigenesis and improved the survival rate when used in combination with radiotherapy and cisplatin in OSCC-SC-transplanted immunocompromised mice. CONCLUSION Sox2-mediated CSCs property is associated with the regulation of EMT and Sox2 s as therapeutic target in OSCC.
Collapse
Affiliation(s)
- Ming-Yung Chou
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan; Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan; Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Fang-Wei Hu
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan; Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chuan-Hang Yu
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan; Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Cheng-Chia Yu
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan; Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan; Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan.
| |
Collapse
|
204
|
Tamura R, Yokoyama Y, Yoshida H, Imaizumi T, Mizunuma H. 4-Methylumbelliferone inhibits ovarian cancer growth by suppressing thymidine phosphorylase expression. J Ovarian Res 2014; 7:94. [PMID: 25304388 PMCID: PMC4198731 DOI: 10.1186/s13048-014-0094-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Accepted: 10/01/2014] [Indexed: 01/09/2023] Open
Abstract
Background 4-Methylumbelliferone (4-MU), a hyaluronan (HA) synthesis inhibitor, has antitumor activity in cancer cells. However, few studies have focused on its effects on ovarian cancer. The aim of this study was to investigate the effects of 4-MU on ovarian cancer and to elucidate its mechanism of action. Methods The HRA human ovarian serous adenocarcinoma cell line was used in this study. The effects of 4-MU on cell proliferation, migration, and invasion were determined by using in vitro assays as well as an in vivo rat peritoneal carcinomatosis model. The expression of HA synthase (HAS), CD44 HA receptor, vascular endothelial growth factor (VEGF), and thymidine phosphorylase (TP) mRNA in HRA cells was analyzed by quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR). Results 4-MU administration inhibited the growth of peritoneal tumors and significantly prolonged survival. In vitro experiments showed that 4-MU inhibited HRA cell proliferation in a dose-dependent manner, while it did not affect HRA cell invasion and migration. 4-MU significantly decreased TP mRNA expression in HRA cells. On the other hand, since HAS2, CD44, and VEGF endogenous mRNA expression levels were very low in HRA cells, it was impossible to evaluate the effect of 4-MU treatment. Conclusions These results suggest that 4-MU exerts its antitumor effect on ovarian cancer through suppressing TP expression.
Collapse
Affiliation(s)
- Ryosuke Tamura
- Department of Obstetrics and Gynecology, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, 036-8562, Japan.
| | - Yoshihito Yokoyama
- Department of Obstetrics and Gynecology, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, 036-8562, Japan.
| | - Hidemi Yoshida
- Department of Vascular Biology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, 036-8562, Japan.
| | - Tadaatsu Imaizumi
- Department of Vascular Biology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, 036-8562, Japan.
| | - Hideki Mizunuma
- Department of Obstetrics and Gynecology, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, 036-8562, Japan.
| |
Collapse
|
205
|
Lu H, Yan C, Quan XX, Yang X, Zhang J, Bian Y, Chen Z, Van Waes C. CK2 phosphorylates and inhibits TAp73 tumor suppressor function to promote expression of cancer stem cell genes and phenotype in head and neck cancer. Neoplasia 2014; 16:789-800. [PMID: 25379016 PMCID: PMC4212254 DOI: 10.1016/j.neo.2014.08.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Revised: 08/21/2014] [Accepted: 08/22/2014] [Indexed: 01/03/2023]
Abstract
Cancer stem cells (CSC) and genes have been linked to cancer development and therapeutic resistance, but the signaling mechanisms regulating CSC genes and phenotype are incompletely understood. CK2 has emerged as a key signal serine/threonine kinase that modulates diverse signal cascades regulating cell fate and growth. We previously showed that CK2 is often aberrantly expressed and activated in head and neck squamous cell carcinomas (HNSCC), concomitantly with mutant (mt) tumor suppressor TP53, and inactivation of its family member, TAp73. Unexpectedly, we observed that classical stem cell genes Nanog, Sox2, and Oct4, are overexpressed in HNSCC with inactivated TAp73 and mtTP53. However, the potential relationship between CK2, TAp73 inactivation, and CSC phenotype is unknown. We reveal that inhibition of CK2 by pharmacologic inhibitors or siRNA inhibits the expression of CSC genes and side population (SP), while enhancing TAp73 mRNA and protein expression. Conversely, CK2 inhibitor attenuation of CSC protein expression and the SP by was abrogated by TAp73 siRNA. Bioinformatic analysis uncovered a single predicted CK2 threonine phosphorylation site (T27) within the N-terminal transactivation domain of TAp73. Nuclear CK2 and TAp73 interaction, confirmed by co-immunoprecipitation, was attenuated by CK2 inhibitor, or a T27A point-mutation of this predicted CK2 threonine phospho-acceptor site of TAp73. Further, T27A mutation attenuated phosphorylation, while enhancing TAp73 function in repressing CSC gene expression and SP cells. A new CK2 inhibitor, CX-4945, inhibited CSC related SP cells, clonogenic survival, and spheroid formation. Our study unveils a novel regulatory mechanism whereby aberrant CK2 signaling inhibits TAp73 to promote the expression of CSC genes and phenotype.
Collapse
Key Words
- CK2, Casein Kinase 2
- CSC, Cancer Stem Cells
- DMAT, 2-Dimethylamino-4,5,6,7-tetrabromo-1H-benzimidazole
- HEKA, Human epidermal keratinocytes
- HNSCC, Head and neck squamous cell carcinoma
- HOK, Human oral keratinocytes
- SP, Side population
- TAp73, Transactivating p73
- TP53, Transforming Protein p53
- UM-SCC, University of Michigan Squamous Cell Carcinoma
- mt, Mutant
- wt, Wild-type
Collapse
Affiliation(s)
- Hai Lu
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892
- Orthopaedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Carol Yan
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892
- Howard Hughes Medical Institute-NIH Research Scholars Program, Bethesda, MD 20892, USA
| | - Xin Xin Quan
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892
| | - Xinping Yang
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892
| | - Jialing Zhang
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892
| | - Yansong Bian
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892
| | - Zhong Chen
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892
| | - Carter Van Waes
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
206
|
Afonso J, Santos LL, Miranda-Gonçalves V, Morais A, Amaro T, Longatto-Filho A, Baltazar F. CD147 and MCT1-potential partners in bladder cancer aggressiveness and cisplatin resistance. Mol Carcinog 2014; 54:1451-66. [PMID: 25263481 DOI: 10.1002/mc.22222] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 07/04/2014] [Accepted: 08/05/2014] [Indexed: 11/08/2022]
Abstract
The relapsing and progressive nature of bladder tumors, and the heterogeneity in the response to cisplatin-containing regimens, are the major concerns in the care of urothelial bladder carcinoma (UBC) patients. The metabolic adaptations that alter the tumor microenvironment and thus contribute to chemoresistance have been poorly explored in UBC setting. We found significant associations between the immunoexpressions of the microenvironment-related molecules CD147, monocarboxylate transporters (MCTs) 1 and 4, CD44 and CAIX in tumor tissue sections from 114 UBC patients. The presence of MCT1 and/or MCT4 expressions was significantly associated with unfavorable clinicopathological parameters. The incidence of CD147 positive staining significantly increased with advancing stage, grade and type of lesion, and occurrence of lymphovascular invasion. Similar associations were observed when considering the concurrent expression of CD147 and MCT1. This expression profile lowered significantly the 5-year disease-free and overall survival rates. Moreover, when selecting patients who received platinum-based chemotherapy, the prognosis was significantly worse for those with MCT1 and CD147 positive tumors. CD147 specific silencing by small interfering RNAs (siRNAs) in UBC cells was accompanied by a decrease in MCT1 and MCT4 expressions and, importantly, an increase in chemosensitivity to cisplatin. Our results provide novel insights for the involvement of CD147 and MCTs in bladder cancer progression and resistance to cisplatin-based chemotherapy. We consider that the possible cooperative role of CD147 and MCT1 in determining cisplatin resistance should be further explored as a potential theranostics biomarker.
Collapse
Affiliation(s)
- Julieta Afonso
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Lúcio L Santos
- Department of Surgical Oncology, Portuguese Institute of Oncology (IPO), Porto, Portugal.,Faculty of Health Sciences, University Fernando Pessoa (UFP), Porto, Portugal
| | - Vera Miranda-Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - António Morais
- Department of Urology, Portuguese Institute of Oncology (IPO), Porto, Portugal
| | - Teresina Amaro
- Experimental Pathology and Therapeutics Research Center, Portuguese Institute of Oncology (IPO), Porto, Portugal
| | - Adhemar Longatto-Filho
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.,Laboratory of Medical Investigation (LIM 14), Faculty of Medicine, São Paulo State University, São Paulo, Brazil.,Molecular Oncology Research Center, Barretos Cancer Hospital, São Paulo, Brazil
| | - Fátima Baltazar
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
207
|
Shigeishi H, Biddle A, Gammon L, Emich H, Rodini CO, Gemenetzidis E, Fazil B, Sugiyama M, Kamata N, Mackenzie IC. Maintenance of stem cell self-renewal in head and neck cancers requires actions of GSK3β influenced by CD44 and RHAMM. Stem Cells 2014; 31:2073-83. [PMID: 23649588 DOI: 10.1002/stem.1418] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 04/02/2013] [Indexed: 01/08/2023]
Abstract
Cells sorted from head and neck cancers on the basis of their high expression of CD44 have high potency for tumor initiation. These cells are also involved in epithelial to mesenchymal transition (EMT) and we have previously reported that cancer stem cells (CSCs) exist as two biologically distinct phenotypes. Both phenotypes are CD44(high) but one is also ESA(high) and maintains epithelial characteristics, the other is ESA(low) , has mesenchymal characteristics and is migratory. Examining CD44-regulated signal pathways in these cells we show that CD44, and also RHAMM, act to inhibit phosphorylation of glycogen synthase kinase 3β (GSK3β). We show that inhibitory phosphorylation reduces the formation of both "tumor spheres" and "holoclone" colonies, functional indicators of stemness. GSK3β inhibition also reduces the expression of stem cell markers such as Oct4, Sox2, and Nanog and upregulates expression of the differentiation markers Calgranulin B and Involucrin in the CD44(high) /ESA(high) cell fraction. Transition of CSCs out of EMT and back to the epithelial CSC phenotype is induced by GSK3β knockdown. These results indicate that GSK3β plays a central role in determining and maintaining the phenotypes and behavior of CSCs in vitro and are likely to be involved in controlling the growth and spread of tumors in vivo.
Collapse
Affiliation(s)
- Hideo Shigeishi
- Blizard Institute Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London United Kingdom; Department of Oral and Maxillofacial Surgery, Division of Cervico-Gnathostomatology, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
208
|
Spiegelberg D, Kuku G, Selvaraju R, Nestor M. Characterization of CD44 variant expression in head and neck squamous cell carcinomas. Tumour Biol 2014; 35:2053-62. [PMID: 24122205 PMCID: PMC3967078 DOI: 10.1007/s13277-013-1272-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Accepted: 09/25/2013] [Indexed: 01/03/2023] Open
Abstract
CD44 is a complex family of molecules, associated with aggressive malignancies and cancer stem cells. However, the role of CD44 variants in tumor progression and treatment resistance is not clear. In this study, the expression of CD44 and its variants was assessed in head and neck squamous cell carcinomas (HNSCC). Furthermore, subpopulations of cells expressing high amounts of CD44 variants were identified and characterized, for e.g., cell cycle phase and radioresistance. Results revealed high and homogenous CD44 and CD44v7 expression in four cell lines and CD44v4 and CD44v6 in three cell lines. CD44v3 was highly expressed in two cell lines, whereas CD44v5, CD44v7/8, CD44v10, CD133, and CD24 demonstrated no or moderate expression. Moreover, a subpopulation of very high CD44v4 expression was identified, which is independent of cell phase, demonstrating increased proliferation and radioresistance. In cell starvation experiments designed to enrich for cancer stem cells, a large population with dramatically increased expression of CD44, CD44v3, CD44v6, and CD44v7 was formed. Expression was independent of cell phase, and cells demonstrated increased radioresistance and migration rate. Our results demonstrate that the heterogeneity of tumor cells has important clinical implications for the treatment of HNSCC and that some of the CD44 variants may be associated with increased radioresistance. Highly expressed CD44 variants could make interesting candidates for selective cancer targeting.
Collapse
Affiliation(s)
- D. Spiegelberg
- Unit of Biomedical Radiation Sciences, Department Radiology, Oncology and Radiation Sciences, Uppsala University, Uppsala, Sweden
- Unit of Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - G. Kuku
- Unit of Biomedical Radiation Sciences, Department Radiology, Oncology and Radiation Sciences, Uppsala University, Uppsala, Sweden
| | - R. Selvaraju
- Unit of Biomedical Radiation Sciences, Department Radiology, Oncology and Radiation Sciences, Uppsala University, Uppsala, Sweden
| | - M. Nestor
- Unit of Biomedical Radiation Sciences, Department Radiology, Oncology and Radiation Sciences, Uppsala University, Uppsala, Sweden
- Unit of Otolaryngology and Head & Neck Surgery, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
209
|
Chen L, Jiang K, Jiang H, Wei P. miR-155 mediates drug resistance in osteosarcoma cells via inducing autophagy. Exp Ther Med 2014; 8:527-532. [PMID: 25009614 PMCID: PMC4079430 DOI: 10.3892/etm.2014.1752] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 05/02/2014] [Indexed: 12/19/2022] Open
Abstract
Frequent acquisition of drug resistance is often associated with the chemotherapy of malignant tumors, including osteosarcoma. A number of studies have demonstrated a critical role for autophagy in osteosarcoma development, therapy and drug resistance. However, the molecular mechanisms underlying the autophagy-mediated chemotherapy resistance of osteosarcoma cells remain largely unknown. In the present study, we determined the autophagy and microRNA-155 (miR-155) expression induced by chemotherapeutic drugs in osteosarcoma cells. Then we determined the promotory role of miR-155 to the chemotherapy-induced autophagy. Our results demonstrated that microRNA-155 (miR-155) expression was highly induced during chemotherapy of osteosarcoma cells, and this was accompanied by upregulated autophagy. The increased miR-155 expression levels upregulated anticancer drug-induced autophagy in osteosarcoma cells and ameliorated the anticancer drug-induced cell proliferation and viability decrease. Therefore, the results of the present study demonstrated that miR-155 mediated drug-resistance in osteosarcoma cells by inducing autophagy. The present study recognized a novel mechanism of chemoresistance in osteosarcoma cancers.
Collapse
Affiliation(s)
- Lu Chen
- Orthopedics Department, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Ke Jiang
- Orthopedics Department, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Hua Jiang
- Orthopedics Department, Second People's Hospital of Chengdu, Chengdu, Sichuan 610017, P.R. China
| | - Peng Wei
- Orthopedics Department, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
210
|
Dai W, Tan X, Sun C, Zhou Q. High expression of SOX2 is associated with poor prognosis in patients with salivary gland adenoid cystic carcinoma. Int J Mol Sci 2014; 15:8393-406. [PMID: 24828201 PMCID: PMC4057738 DOI: 10.3390/ijms15058393] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 02/24/2014] [Accepted: 03/11/2014] [Indexed: 02/07/2023] Open
Abstract
Sex determining region Y-BOX2 (SOX2), one of the key members of the SOX family, is a transcription factor that is involved in the maintenance of embryonic stem cell pluripotency and in multiple developmental processes. Recent studies have shown that SOX2 is aberrantly expressed in several types of tumors. The present study aimed to investigate the clinicopathological and prognostic significance of SOX2 in adenoid cystic carcinoma (ACC) of salivary gland. In this study, the expression of SOX2 in ACC tissues and matched adjacent non-cancerous tissues was measured by immunohistochemistry, western blot, and quantitative polymerase chain reaction. High SOX2 expression occurred in approximately 62.6% of primary ACC. In addition, high expression of SOX2 was significantly associated with T classification (p=0.003) and distant metastasis (p=0.002). The 5-year overall survival (OS) and disease-free survival (DFS) in patients with high SOX2 expression is poorer than those with low SOX2 expression. When adjusted by multivariate analysis, high SOX2 expression, together with distant metastasis, was an independent prognostic factor. The findings of the present study provide evidence that SOX2 represents a potential novel prognostic biomarker for ACC patients.
Collapse
Affiliation(s)
- Wei Dai
- Department of Oromaxillofacial-Head and Neck Surgery & Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, Shenyang 110002, China.
| | - Xuexin Tan
- Department of Oromaxillofacial-Head and Neck Surgery & Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, Shenyang 110002, China.
| | - Changfu Sun
- Department of Oromaxillofacial-Head and Neck Surgery & Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, Shenyang 110002, China.
| | - Qing Zhou
- Department of Oromaxillofacial-Head and Neck Surgery & Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, Shenyang 110002, China.
| |
Collapse
|
211
|
CD163+ tumor-associated macrophages correlated with poor prognosis and cancer stem cells in oral squamous cell carcinoma. BIOMED RESEARCH INTERNATIONAL 2014; 2014:838632. [PMID: 24883329 PMCID: PMC4032721 DOI: 10.1155/2014/838632] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 04/05/2014] [Accepted: 04/06/2014] [Indexed: 01/23/2023]
Abstract
Tumor-associated macrophages (TAMs) play an important role in the progression and prognostication of numerous cancers. However, the role and clinical significance of TAM markers in oral squamous cell carcinoma (OSCC) has not been elucidated. The present study was designed to investigate the correlation between the expression of TAM markers and pathological features in OSCC by tissue microarray. Tissue microarrays containing 16 normal oral mucosa, 6 oral epithelial dysplasia, and 43 OSCC specimens were studied by immunohistochemistry. We observed that the protein expression of the TAM markers CD68 and CD163 as well as the cancer stem cell (CSC) markers ALDH1, CD44, and SOX2 increased successively from the normal oral mucosa to OSCC. The expressions of CD68 and CD163 were significantly associated with lymph node status, and SOX2 was significantly correlated with pathological grade and lymph node status, whereas ALDH1 was correlated with tumor stage. Furthermore, CD68 was significantly correlated with CD163, SOX2, and ALDH1 (P < 0.05). Kaplan-Meier analysis revealed that OSCC patients overexpressing CD163 had significantly worse overall survival (P < 0.05). TAM markers are associated with cancer stem cell marker and OSCC overall survival, suggesting their potential prognostic value in OSCC.
Collapse
|
212
|
Abstract
Protein kinase C (PKC) is a family of phospholipid-dependent serine/threonine kinases, which can be further classified into three PKC isozymes subfamilies: conventional or classic, novel or nonclassic, and atypical. PKC isozymes are known to be involved in cell proliferation, survival, invasion, migration, apoptosis, angiogenesis, and drug resistance. Because of their key roles in cell signaling, PKC isozymes also have the potential to be promising therapeutic targets for several diseases, such as cardiovascular diseases, immune and inflammatory diseases, neurological diseases, metabolic disorders, and multiple types of cancer. This review primarily focuses on the activation, mechanism, and function of PKC isozymes during cancer development and progression.
Collapse
|
213
|
Kovalszky I, Hjerpe A, Dobra K. Nuclear translocation of heparan sulfate proteoglycans and their functional significance. Biochim Biophys Acta Gen Subj 2014; 1840:2491-7. [PMID: 24780644 DOI: 10.1016/j.bbagen.2014.04.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 04/17/2014] [Accepted: 04/18/2014] [Indexed: 12/25/2022]
Abstract
BACKGROUND Heparan sulfate proteoglycans (HSPGs) are important constituents of the cell membrane and they act as co-receptors for cellular signaling. Syndecan-1, glypican and perlecan also translocate to the nucleus in a regulated manner. Similar nuclear transport of growth factors and heparanase indicate a possible co-regulation and functional significance. SCOPE OF REVIEW In this review we dissect the structural requirement for the nuclear translocation of HSPGs and their functional implications.s MAJOR CONCLUSIONS The functions of the nuclear HSPGs are still incompletely understood. Evidence point to possible functions in hampering cell proliferation, inhibition of DNA topoisomerase I activity and inhibition of gene transcription. GENERAL SIGNIFICANCE HSPGs influence the behavior of malignant tumors in many different ways. Modulating their functions may offer powerful tools to control fundamental biological processes and provide the basis for subsequent targeted therapies in cancer. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.
Collapse
Affiliation(s)
- Ilona Kovalszky
- First Department of Pathology & Experimental Cancer Research Semmelweis University, Üllői street 26, Budapest 1085, Hungary
| | - Anders Hjerpe
- Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital F46, SE-141 86 Stockholm Sweden
| | - Katalin Dobra
- Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital F46, SE-141 86 Stockholm Sweden.
| |
Collapse
|
214
|
Piccinini AM, Midwood KS. Illustrating the interplay between the extracellular matrix and microRNAs. Int J Exp Pathol 2014; 95:158-80. [PMID: 24761792 DOI: 10.1111/iep.12079] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 02/18/2014] [Indexed: 12/19/2022] Open
Abstract
The discovery of cell surface receptors that bind to extracellular matrix (ECM) components marked a new era in biological research. Since then there has been an increasing appreciation of the importance of studying cells in the context of their extracellular environment. Cell behaviour is profoundly affected by the ECM, whose synthesis and turnover must be finely balanced in order to maintain normal function and prevent disease. In the last decade, microRNAs (miRNAs) have emerged as key regulators of ECM gene expression. As new technologies for the identification and validation of miRNA targets continue to be developed, a growing body of data supporting the role of miRNAs in regulating the ECM biology has arisen from a variety of cell and animal models along with clinical studies. However, more recent findings suggest an intriguing interplay between the ECM and miRNAs: not only can miRNAs control the composition of the ECM, but also the ECM can affect the expression of specific miRNAs. Here we discuss how miRNAs contribute to the synthesis, maintenance and remodelling of the ECM during development and disease. Furthermore, we bring to light evidence that points to a role for the ECM in regulating miRNA expression and function.
Collapse
Affiliation(s)
- Anna M Piccinini
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington, Oxford, UK
| | | |
Collapse
|
215
|
miR-22 inhibits osteosarcoma cell proliferation and migration by targeting HMGB1 and inhibiting HMGB1-mediated autophagy. Tumour Biol 2014; 35:7025-34. [PMID: 24752578 DOI: 10.1007/s13277-014-1965-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 04/10/2014] [Indexed: 12/19/2022] Open
Abstract
Acquisition of drug-resistant phenotypes is often associated with chemotherapy in osteosarcoma. Studies show that high-mobility group box 1 (HMGB1) plays an important role in facilitating autophagy and promotes drug resistance in osteosarcoma cells. In this study, we determined the targeting role of miR-22 to HMGB1 and the regulation of miR-22 on the HMGB1-mediated cell autophagy and on the cell proliferation, migration, and invasion of osteosarcoma cells. Results demonstrated that miR-22 well paired with the 3'-UTR of HMGB1 downregulated the HMGB1 expression and blocked the HMGB1-mediated autophagy during chemotherapy in osteosarcoma cells in vitro. Further study showed that the blockage of autophagy by miR-22 inhibited the osteosarcoma cell proliferation, migration, and invasion. In summary, this study implied the negative regulation of miR-22 on the HMGB1-mediated autophagy in osteosarcoma cells.
Collapse
|
216
|
Wang R, Liu W, Helfer CM, Bradner JE, Hornick JL, Janicki SM, French CA, You J. Activation of SOX2 expression by BRD4-NUT oncogenic fusion drives neoplastic transformation in NUT midline carcinoma. Cancer Res 2014; 74:3332-43. [PMID: 24736545 DOI: 10.1158/0008-5472.can-13-2658] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BRD4 is implicated in the pathogenesis of a number of different cancers. It is also the target of translocation t(15;19) that accounts for the highly aggressive NUT midline carcinoma (NMC). We discovered that t(15;19) NMC cells display the ability to grow into stem cell-like spheres and express an exceptionally high level of the stem cell marker, SOX2. The BRD4-NUT fusion oncogene resulting from t(15;19) translocation is required for the abnormal activation of SOX2, which drives the stem cell-like proliferation and cellular transformation in NMC cells. SOX2 knockdown phenocopies the effects of BRD4-NUT inhibition, whereas ectopic SOX2 expression rescues the phenotype. The BRD4-NUT-induced abnormal SOX2 activation was observed in multiple NMC cell lines as well as in NMC primary tumors. We further demonstrate that BRD4-NUT oncoprotein recruits p300 to stimulate transcription activation and that inhibition of p300 represses SOX2 transcription in NMC cells. These studies identify this stem cell marker as a novel BRD4-NUT target that supports the highly aggressive transforming activity of t(15;19) carcinomas. Our study provides new mechanistic insights for understanding how alteration of BRD4 function by BRD4-NUT oncogene leads to the highly malignant NMC carcinoma. Because abnormal stem cell self-renewal is frequently observed during tumor formation and metastasis, the aberrant stem cell-like proliferation associated with BRD4 dysregulation observed in NMC carcinoma may have implications for studying the oncogenic mechanism of other BRD4-associated tumors.
Collapse
Affiliation(s)
- Ranran Wang
- Authors' Affiliations: Department of Microbiology, University of Pennsylvania Perelman School of Medicine; and Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, Pennsylvania; Department of Medical Oncology, Dana-Farber Cancer Institute, Dana 510D; and Department of Pathology, Brigham and Women's Hospital/Harvard Medical School, Boston, Massachusetts
| | - Wei Liu
- Authors' Affiliations: Department of Microbiology, University of Pennsylvania Perelman School of Medicine; and Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, Pennsylvania; Department of Medical Oncology, Dana-Farber Cancer Institute, Dana 510D; and Department of Pathology, Brigham and Women's Hospital/Harvard Medical School, Boston, Massachusetts
| | - Christine M Helfer
- Authors' Affiliations: Department of Microbiology, University of Pennsylvania Perelman School of Medicine; and Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, Pennsylvania; Department of Medical Oncology, Dana-Farber Cancer Institute, Dana 510D; and Department of Pathology, Brigham and Women's Hospital/Harvard Medical School, Boston, Massachusetts
| | - James E Bradner
- Authors' Affiliations: Department of Microbiology, University of Pennsylvania Perelman School of Medicine; and Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, Pennsylvania; Department of Medical Oncology, Dana-Farber Cancer Institute, Dana 510D; and Department of Pathology, Brigham and Women's Hospital/Harvard Medical School, Boston, Massachusetts
| | - Jason L Hornick
- Authors' Affiliations: Department of Microbiology, University of Pennsylvania Perelman School of Medicine; and Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, Pennsylvania; Department of Medical Oncology, Dana-Farber Cancer Institute, Dana 510D; and Department of Pathology, Brigham and Women's Hospital/Harvard Medical School, Boston, Massachusetts
| | - Susan M Janicki
- Authors' Affiliations: Department of Microbiology, University of Pennsylvania Perelman School of Medicine; and Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, Pennsylvania; Department of Medical Oncology, Dana-Farber Cancer Institute, Dana 510D; and Department of Pathology, Brigham and Women's Hospital/Harvard Medical School, Boston, Massachusetts
| | - Christopher A French
- Authors' Affiliations: Department of Microbiology, University of Pennsylvania Perelman School of Medicine; and Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, Pennsylvania; Department of Medical Oncology, Dana-Farber Cancer Institute, Dana 510D; and Department of Pathology, Brigham and Women's Hospital/Harvard Medical School, Boston, Massachusetts
| | - Jianxin You
- Authors' Affiliations: Department of Microbiology, University of Pennsylvania Perelman School of Medicine; and Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, Pennsylvania; Department of Medical Oncology, Dana-Farber Cancer Institute, Dana 510D; and Department of Pathology, Brigham and Women's Hospital/Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
217
|
Liu L, Lian J, Zhang H, Tian H, Liang M, Yin M, Sun F. MicroRNA-302a sensitizes testicular embryonal carcinoma cells to cisplatin-induced cell death. J Cell Physiol 2014; 228:2294-304. [PMID: 23625774 DOI: 10.1002/jcp.24394] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 04/18/2013] [Indexed: 11/09/2022]
Abstract
Cisplatin is a commonly used chemotherapeutic agent for the treatment of several human malignancies, such as testicular germ cell tumors (TGCT). The toxic effects persist and those that are present long after chemotherapy affect the overall quality of life of patients. MicroRNAs (miRNAs) play important roles in the responses of cancer cells to chemotherapy and have been shown to modulate cell sensitivity to chemotherapeutic drugs. However, the relationship between miRNA expression and cisplatin sensitivity of TGCT has not been fully explored. In this study, the effects of miR-302a on cisplatin cytotoxicity in TGCT-derived cell line NTERA-2 (NT2) were evaluated. We found that expression levels of miR-302a were increased in cisplatin-treated NT2 cells. Up-regulation of miR-302a significantly increased the sensitivity of NT2 cells to cisplatin by enhancing cisplatin-induced G2/M phase arrest and the subsequent progression to apoptosis. MiR-302a also increased the killing effects of cisplatin by lowering the apoptotic threshold; the same result was also observed in another TGCT-derived cell line, NCCIT. Furthermore, miR-302a-enhanced cisplatin sensitivity was partially mediated through the down-regulation of p21 in NT2 cells. MiR-302a induced apoptosis was further enhanced by silencing of p53 in NT2 cells. p53 levels were inversely associated with the expression of Oct4, Sox2, and Nanog in response to cisplatin. Thus, targeting miR-302a may offer new therapeutic interventions in TGCT.
Collapse
Affiliation(s)
- Lin Liu
- Hefei National Laboratory for Physical Sciences at Microscale and Department of Cell and Developmental Biology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, P.R. China
| | | | | | | | | | | | | |
Collapse
|
218
|
Li X, Wang S, Chen Y, Liu G, Yang X. miR-22 targets the 3' UTR of HMGB1 and inhibits the HMGB1-associated autophagy in osteosarcoma cells during chemotherapy. Tumour Biol 2014; 35:6021-8. [PMID: 24609901 DOI: 10.1007/s13277-014-1797-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 02/25/2014] [Indexed: 12/19/2022] Open
Abstract
Osteosarcoma is the most common malignant bone tumor for children and adolescents, and the frequent acquisition of drug-resistant phenotypes and the occurrence of "secondary malignancies" are often associated with chemotherapy and are significant obstacles to achieving favorable outcomes. Thus, it is urgent to identify the molecular mechanisms underlying the chemoresistance of osteosarcoma. In this study, we showed that miR-22 and high-mobility group box 1 (HMGB1) were deregulated in osteosarcoma cells, post-chemotherapy; the upregulated HMGB1 mediated autophagy and contributed to chemotherapy resistance in osteosarcoma in vitro. However, possibly as a compensatory effect, miR-22 was also upregulated during the chemotherapy, and the overexpressed miR-22 targeted the 3' UTR of HMGB1 and inhibits the HMGB1-promoted autophagy. Our study suggests a complexity in the regulation of autophagy by miR-22 and HMGB1 during chemotherapy resistance in osteosarcoma. These results reveal novel potential role of miR-22 against chemotherapy resistance during the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Xuefeng Li
- Department of Anesthesiology, China-Japan Union Hospital, Jilin University, Changchun, China
| | | | | | | | | |
Collapse
|
219
|
Chen L, Bourguignon LYW. Hyaluronan-CD44 interaction promotes c-Jun signaling and miRNA21 expression leading to Bcl-2 expression and chemoresistance in breast cancer cells. Mol Cancer 2014; 13:52. [PMID: 24606718 PMCID: PMC3975292 DOI: 10.1186/1476-4598-13-52] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 02/25/2014] [Indexed: 11/10/2022] Open
Abstract
MicroRNA-21 (miR-21) is associated with the development of solid tumors progression including breast cancer. In this study we investigated matrix hyaluronan (HA)-CD44 (a primary HA receptor) interaction with c-Jun N-Terminal Kinase (JNK)/c-Jun signaling in MDA-MB-468 breast cancer cells [a triple-negative (estrogen receptor-negative/progesterone receptor-negative/HER2-negative) breast cancer cell line]. Our results indicated that HA binding to CD44 promotes c-Jun nuclear translocation and transcriptional activation. Further analyses revealed that miR-21 is regulated by an upstream promoter containing AP1 binding site(s), and chromatin immunoprecipitation (CHIP) assays demonstrated that stimulation of miR-21 expression by HA/CD44 interaction is c-Jun-dependent in these breast cancer cells. This process results in an increase of the anti-apoptosis protein Bcl-2 and upregulation of inhibitors of the apoptosis family of proteins (IAPs) as well as chemoresistance in MDA-MB-468 cells. Treatment with c-Jun specific small interfering RNAs effectively blocks HA-mediated c-Jun signaling and abrogates miR-21 production as well as causes downregulation of survival proteins (Bcl-2 and IAPs) and enhancement of chemosensitivity. In addition, our results demonstrated that anti-miR-21 inhibitor not only downregulates Bcl-2/IAP expression but also increases chemosensitivity in HA-treated breast cancer cells. Together, these findings suggest that the HA/CD44-induced c-Jun signaling plays a pivotal role in miR-21 production leading to survival protein (Bcl-2/IAP) upregulation and chemoresistance in triple negative breast cancer cells such as MDA-MB-468 cell line. This novel HA/CD44-mediated c-Jun signaling pathway and miR-21 production provide a new drug target for the future intervention strategies to treat breast cancer.
Collapse
Affiliation(s)
| | - Lilly Y W Bourguignon
- San Francisco Veterans Affairs Medical Center and Department of Medicine, University of California at San Francisco & Endocrine Unit (111N2), 4150 Clement Street, San Francisco, CA 94121, USA.
| |
Collapse
|
220
|
Xiao S, Zhou Y, Jiang J, Yuan L, Xue M. CD44 affects the expression level of FOS‑like antigen 1 in cervical cancer tissues. Mol Med Rep 2014; 9:1667-74. [PMID: 24604526 DOI: 10.3892/mmr.2014.2010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Accepted: 02/02/2014] [Indexed: 12/19/2022] Open
Abstract
Cervical carcinoma is the second most prevalent type of malignancy in females worldwide. The crucial etiological factors involved in the development of cervical carcinoma include infection with the papillomavirus, and the structural or functional mutation of oncogenes and tumor suppressor genes. CD44 refers to a multifunctional family of type I transmembrane proteins. These proteins have been implicated in numerous biological processes, including cell adhesion, cell migration and metastasis. The present study examined the differences in the expression levels of ATP-binding cassette sub-family G member 2, CD24, CD44, CD133, cytokeratin (CK) 14 and CK19 between cervical cancer tissues and corresponding normal non-tumor tissues by flow cytometry. Then, the CD44+ or CD44‑ cells from cervical cancer tissues were sorted for identification and confirmation of differential expression by flow cytometry. The results demonstrated that the expression level of CD44 in cervical cancer tissues was higher than in the corresponding non-tumor normal tissues (t=3.12; P=0.0102). Compared with the CD44‑ cells, the FOS-like antigen 1 (Fra-1), nestin, nuclear receptor subfamily 4, group A, member 2, OCT4 and p63 genes were highly expressed in CD44+ cells. The fold changes were 3.55, 3.55, 2.46, 2.87 and 2.56, respectively (P<0.05). However, BMI1 polycomb ring finger oncogene, ck5, tumor protein p53 and lactotransferrin genes exhibited low expression levels in CD44+ cells. It was verified by western blot analysis and flow cytometry that Fra-1 was highly expressed in CD44+ cells. Fra-1 was a potential target of miR-19a and miR-19b. The expression of miR-19a and miR-19b was downregulated by ~50% in CD44+ cells compared with CD44‑ cells. These findings suggested that CD44 dysregulated the activation of the Fra‑1 gene. The interaction of Fra-1 and CD44 may therefore be important in cervical carcinoma.
Collapse
Affiliation(s)
- Songshu Xiao
- Department of Gynecology and Obstetrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Yanhong Zhou
- Molecular Genetics Laboratory, Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Jianfa Jiang
- Department of Gynecology and Obstetrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Le Yuan
- Department of Gynecology and Obstetrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Min Xue
- Department of Gynecology and Obstetrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
221
|
MicroRNAs: master regulators of drug resistance, stemness, and metastasis. J Mol Med (Berl) 2014; 92:321-36. [PMID: 24509937 DOI: 10.1007/s00109-014-1129-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 01/21/2014] [Accepted: 01/23/2014] [Indexed: 12/13/2022]
Abstract
MicroRNAs (miRNAs) are 20-22 nucleotides long small non-coding RNAs that regulate gene expression post-transcriptionally. Last decade has witnessed emerging evidences of active roles of miRNAs in tumor development, progression, metastasis, and drug resistance. Many factors contribute to their dysregulation in cancer, such as chromosomal aberrations, differential methylation of their own or host genes' promoters and alterations in miRNA biogenesis pathways. miRNAs have been shown to act as tumor suppressors or oncogenes depending on the targets they regulate and the tissue where they are expressed. Because miRNAs can regulate dozens of genes simultaneously and they can function as tumor suppressors or oncogenes, they have been proposed as promising targets for cancer therapy. In this review, we focus on the role of miRNAs in driving drug resistance and metastasis which are associated with stem cell properties of cancer cells. Furthermore, we discuss systems biology approaches to combine experimental and computational methods to study effects of miRNAs on gene or protein networks regulating these processes. Finally, we describe methods to target oncogenic or replace tumor suppressor miRNAs and current delivery strategies to sensitize refractory cells and to prevent metastasis. A holistic understanding of miRNAs' functions in drug resistance and metastasis, which are major causes of cancer-related deaths, and the development of novel strategies to target them efficiently will pave the way towards better translation of miRNAs into clinics and management of cancer therapy.
Collapse
|
222
|
Ibrahim SA, Hassan H, Götte M. MicroRNA-dependent targeting of the extracellular matrix as a mechanism of regulating cell behavior. Biochim Biophys Acta Gen Subj 2014; 1840:2609-20. [PMID: 24462576 DOI: 10.1016/j.bbagen.2014.01.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 01/13/2014] [Accepted: 01/15/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND MicroRNAs are small noncoding RNAs which regulate gene expression at the posttranscriptional level by inducing mRNA degradation or translational repression. MicroRNA-dependent modulation of the extracellular matrix and its cellular receptors has emerged as a novel mechanism of regulating numerous matrix-dependent processes, including cell proliferation and apoptosis, cell adhesion and migration, cell differentiation and stem cell properties. SCOPE OF REVIEW In this review, we will present different mechanisms by which microRNAs and extracellular matrix constituents mutually regulate their expression, and we will demonstrate how these expression changes affect cell behavior. We will also highlight the importance of dysregulated matrix-related microRNA expression for the pathogenesis of inflammatory and malignant disease, and discuss the potential for diagnostic and therapeutic applications. MAJOR CONCLUSIONS MicroRNAs and matrix-dependent signal transduction processes form novel regulatory circuits, which profoundly affect cell behavior. As misexpression of microRNAs targeting extracellular matrix constituents is observed in a variety of diseases, a pharmacological intervention with these processes has therapeutic potential, as successfully demonstrated in vitro and in advanced animal models. However, a deeper mechanistic understanding is required to address potential side effects prior to clinical applications in humans. GENERAL SIGNIFICANCE A full understanding of the role and function of microRNA-dependent regulation of the extracellular matrix may lead to new targeted therapies and new diagnostics for malignant and inflammatory diseases in humans. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.
Collapse
Affiliation(s)
| | - Hebatallah Hassan
- Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Campus 1, D11, 48149 Münster, Germany.
| |
Collapse
|
223
|
Huang CF, Xu XR, Wu TF, Sun ZJ, Zhang WF. Correlation of ALDH1, CD44, OCT4 and SOX2 in tongue squamous cell carcinoma and their association with disease progression and prognosis. J Oral Pathol Med 2014; 43:492-8. [PMID: 24450601 DOI: 10.1111/jop.12159] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2013] [Indexed: 01/25/2023]
Affiliation(s)
- Cong-Fa Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan China
- Department of Oral and Maxillofacial Surgery; School of Stomatology; Wuhan University; Wuhan China
| | - Xiao-Rong Xu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan China
| | - Tian-Fu Wu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan China
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan China
- Department of Oral and Maxillofacial Surgery; School of Stomatology; Wuhan University; Wuhan China
| | - Wen-Feng Zhang
- Department of Oral and Maxillofacial Surgery; School of Stomatology; Wuhan University; Wuhan China
| |
Collapse
|
224
|
Zhang M, Yang Q, Zhang L, Zhou S, Ye W, Yao Q, Li Z, Huang C, Wen Q, Wang J. miR-302b is a potential molecular marker of esophageal squamous cell carcinoma and functions as a tumor suppressor by targeting ErbB4. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2014; 33:10. [PMID: 24438167 PMCID: PMC3898821 DOI: 10.1186/1756-9966-33-10] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Accepted: 01/16/2014] [Indexed: 11/10/2022]
Abstract
Background ErbB4 expression has been noted in various tumors, but its regulatory mechanism in esophageal squamous cell carcinoma (ESCC) remains unclear. The aim of this study was to investigate whether miR-302b regulates the expression of ErbB4 at the post-transcriptional level and to determine its expression, significance, and function in ESCC. Methods We used real-time reverse transcriptase-polymerase chain reaction to quantify the expression of miR-302b in 50 ESCC tissues and analyzed its relationship with clinicopathological factors and survival. Then, we investigated the post-transcriptional regulation of ErbB4 expression using immunoblot analysis and luciferase reporter assays. Finally, the effects of miR-302b on proliferation, apoptosis, and invasion of ESCC cells was detected using MTT, flow cytometric analysis, and transwell invasion assays, respectively. Results miR-302b was significantly down-regulated and correlated with tumor differentiation and lymph node metastasis in ESCC. Univariate and multivariate analyses indicated that low miR-302b expression might be a poor prognostic factor. Further studies demonstrated that miR-302b post-transcriptionally down-regulated the expression of ErbB4 in vitro. Moreover, miR-302b inhibited proliferation by inducing apoptosis and repressed invasion in the ESCC cell lines. Conclusions miR-302b is a potential molecular marker of ESCC and functions as a tumor suppressor by post-transcriptionally regulating ErbB4.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Cheng Huang
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038 Shaanxi Province, China.
| | | | | |
Collapse
|
225
|
Iv Santaliz-Ruiz LE, Xie X, Old M, Teknos TN, Pan Q. Emerging role of nanog in tumorigenesis and cancer stem cells. Int J Cancer 2014; 135:2741-8. [PMID: 24375318 DOI: 10.1002/ijc.28690] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 12/10/2013] [Accepted: 12/16/2013] [Indexed: 12/16/2022]
Abstract
Nanog is a transcription factor that is well-established as a key regulator of embryonic stem cell (ESC) maintenance. Recent evidence demonstrates that Nanog is dysregulated and intimately involved in promoting tumorigenesis in part through regulation of the cancer stem cell (CSC) population. Elevated Nanog is associated with poorer outcome in numerous epithelial malignancies. Nanog is enriched in CSCs and ablation of Nanog is sufficient to reduce the CSC pool. Nanog has also been implicated to promote chemoresistance and epithelial-mesenchymal transition (EMT). Insight into the Nanog signaling cascade, upstream regulators and downstream effectors, is beginning to emerge but remains to be fully elucidated. This review highlights the current literature on the emerging role of Nanog in tumorigenesis and CSCs.
Collapse
Affiliation(s)
- Luis E Iv Santaliz-Ruiz
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University Wexner Medical Center, Columbus, OH; Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | | | | | | | | |
Collapse
|
226
|
Garg S, Madkaikar M, Ghosh K. Investigating cell surface markers on normal hematopoietic stem cells in three different niche conditions. Int J Stem Cells 2014; 6:129-33. [PMID: 24386557 DOI: 10.15283/ijsc.2013.6.2.129] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2013] [Indexed: 01/07/2023] Open
Abstract
Hematopoietic stem cells are of therapeutic interest to the clinicians and researchers due to their promising assistance in management of malignant and inherited hematological conditions. Evaluation of cell surface markers using multiparametric flow cytometry is a well adapted qualitative measure of cells in question for many years. An artillery of these markers has been studied in hematological malignancies and related disorders. However, their role and differential expression on normal hematopoietic stem cells from clinically available sources is not always described carefully. In the present study, we attempted to evaluate expression of CD44, CD90, CD96 and CD123 in three clinically available sources of normal HSCs (Hematopoietic stem cells). Sources of HSCs in the present study involved umbilical cord blood (UCB), normal bone marrow (NBM) and bone marrow from idiopathic thrombocytopenic purpura (ITP) patients (IBM). CD44 is an important homing receptor while CD90 is involved in maintaining stem cell quiescent. CD96 is known to be leukemia specific marker and CD123 is involved in stem cell differentiation and survival. We observed a significant difference in expression CD44, CD90 and CD123 on normal HSCs derived from umbilical cord and ITP marrow. CD96 was highly expressed on HSCs obtained from ITP marrow. Investigating expression of these markers on normal HSCs in different niches will be helpful in correlating their function with niche condition and delineating their 'abnormal' expression from the normal.
Collapse
Affiliation(s)
- Swati Garg
- National Institute of Immunohaemtology, Mumbai, India
| | | | | |
Collapse
|
227
|
Ahmad A, Li Y, Bao B, Kong D, Sarkar FH. Epigenetic regulation of miRNA-cancer stem cells nexus by nutraceuticals. Mol Nutr Food Res 2013; 58:79-86. [PMID: 24272883 DOI: 10.1002/mnfr.201300528] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Revised: 10/06/2013] [Accepted: 10/08/2013] [Indexed: 01/15/2023]
Abstract
Nutraceuticals, the bioactive food components represented by many naturally occurring dietary compounds, have been investigated for a few decades for their numerous beneficial effects, including their anticancer properties. The initial interest in the cancer-preventing/therapeutic ability of these agents was based on their ability to affect multiple signaling pathways that are deregulated in cancer cells. With a shift in the focus of cancer research to the emerging areas such as epigenetic regulation, microRNAs (miRNAs) and the cancer stem cells (CSCs), nutraceuticals initially appeared out of place. However, research investigations over the last several years have slowly but firmly presented evidence that supports a relevance of these agents in modern day research. While nutraceuticals are increasingly being realized to alter miRNA/CSCs expression and function, the molecular mechanism(s) are not very clearly understood. Epigenetic regulation is one mechanism by which these agents exert their anticancer effects. In this focused mini review, we summarize our current understanding of epigenetic regulation of miRNAs and CSCs by nutraceuticals. We discuss both direct and indirect evidences that support such an activity of these compounds.
Collapse
Affiliation(s)
- Aamir Ahmad
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | | | | | | | | |
Collapse
|
228
|
Langenfeld E, Deen M, Zachariah E, Langenfeld J. Small molecule antagonist of the bone morphogenetic protein type I receptors suppresses growth and expression of Id1 and Id3 in lung cancer cells expressing Oct4 or nestin. Mol Cancer 2013; 12:129. [PMID: 24160469 PMCID: PMC4176118 DOI: 10.1186/1476-4598-12-129] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 10/17/2013] [Indexed: 01/06/2023] Open
Abstract
Background Bone morphogenetic proteins (BMP) are embryonic morphogens that are aberrantly expressed in lung cancer. BMPs mediate cell fate decisions and self-renewal of stem cells, through transcription regulation of inhibitor of differentiation protein/DNA binding proteins (Id1-3). Inhibition of BMP signaling decreases growth and induces cell death of lung cancer cells lines by downregulating the expression of Id proteins. It is not known whether the BMP signaling cascade regulates growth and the expression of Id proteins of lung cancer cells expressing the stem cell markers Oct4 and/or nestin. Methods Lung cancer cells expressing Oct4 or nestin were isolated from lung cancer cell lines by stably transfecting the Oct4 promoter or nestin promoter expression vectors that induce expression of the green fluorescent protein reporter. Results Our studies suggest that lung cancer cells expressing Oct4 or nestin are different cell populations. Microarray and quantitative RT-PCR demonstrated that the expression of specific stem cell markers were different between isolated Oct4 and nestin cells. Both the Oct4 and nestin populations were more tumorigenic than controls but histologically they were quite different. The isolated Oct4 and nestin cells also responded differently to inhibition of BMP signaling. Blockade of BMP signaling with the BMP receptor antagonist DMH2 caused significant growth inhibition of both the Oct4 and nestin cell populations but only increased cell death in the nestin population. DMH2 also induced the expression of nestin in the Oct4 population but not in the nestin cells. We also show that BMP signaling is an important regulator of Id1 and Id3 in both the Oct4 and nestin cell populations. Furthermore, we show that NeuN is frequently expressed in NSCLC and provide evidence suggesting that Oct4 cells give rise to cancer cells expressing nestin and/or NeuN. Conclusion These studies show that although biologically different, BMP signaling is growth promoting in cancer cells expressing Oct4 or nestin. Inhibition of BMP signaling decreases expression of Id proteins and suppresses growth of cancer cells expressing Oct4 or Nestin. Small molecule antagonists of the BMP type I receptors represent potential novel drugs to target the population of cancer cells expressing stem cell markers.
Collapse
Affiliation(s)
| | | | | | - John Langenfeld
- Division of Thoracic Surgery, Rutgers-Robert Wood Johnson Medical School, One Robert Wood Johnson Place, P,O, Box 19, New Brunswick, NJ 08903-0019, USA.
| |
Collapse
|
229
|
Liu J, Xiao Z, Wong SKM, Tin VPC, Ho KY, Wang J, Sham MH, Wong MP. Lung cancer tumorigenicity and drug resistance are maintained through ALDH(hi)CD44(hi) tumor initiating cells. Oncotarget 2013; 4:1698-1711. [PMID: 24091605 PMCID: PMC3858556 DOI: 10.18632/oncotarget.1246] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 08/28/2013] [Indexed: 02/07/2023] Open
Abstract
Limited improvement in long term survival of lung cancer patients has been achieved by conventional chemotherapy or targeted therapy. To explore the potentials of tumor initiating cells (TIC)-directed therapy, it is essential to identify the cell targets and understand their maintenance mechanisms. We have analyzed the performance of ALDH/CD44 co-expression as TIC markers and treatment targets of lung cancer using well-validated in vitro and in vivo analyses in multiple established and patient-derived lung cancer cells. The ALDH(hi)CD44(hi) subset showed the highest enhancement of stem cell phenotypic properties compared to ALDH(hi)CD44(lo), ALDH(lo)CD44(hi), ALDH(lo)CD44(lo) cells and unsorted controls. They showed higher invasion capacities, pluripotency genes and epithelial-mesenchymal transition transcription factors expression, lower intercellular adhesion protein expression and higher G2/M phase cell cycle fraction. In immunosuppressed mice, the ALDH(hi)CD44(hi)xenografts showed the highest tumor induction frequency, serial transplantability, shortest latency, largest volume and highest growth rates. Inhibition of sonic Hedgehog and Notch developmental pathways reduced ALDH+CD44+ compartment. Chemotherapy and targeted therapy resulted in higher AALDH(hi)CD44(hi) subset viability and ALDH(lo)CD44(lo) subset apoptosis fraction. ALDH inhibition and CD44 knockdown led to reduced stemness gene expression and sensitization to drug treatment. In accordance, clinical lung cancers containing a higher abundance of ALDH and CD44-coexpressing cells was associated with lower recurrence-free survival. Together, results suggested theALDH(hi)CD44(hi)compartment was the cellular mediator of tumorigenicity and drug resistance. Further investigation of the regulatory mechanisms underlying ALDH(hi)CD44(hi)TIC maintenance would be beneficial for the development of long term lung cancer control.
Collapse
Affiliation(s)
- Jing Liu
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR China
| | - Zhijie Xiao
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR China
| | - Sunny Kit-Man Wong
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR China
| | - Vicky Pui-Chi Tin
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR China
| | - Ka-Yan Ho
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR China
| | - Junwen Wang
- Department of Biochemistry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR China
| | - Mai-Har Sham
- Department of Biochemistry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR China
| | - Maria Pik Wong
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR China
| |
Collapse
|
230
|
Rengaraj D, Park TS, Lee SI, Lee BR, Han BK, Song G, Han JY. Regulation of Glucose Phosphate Isomerase by the 3′UTR-Specific miRNAs miR-302b and miR-17-5p in Chicken Primordial Germ Cells1. Biol Reprod 2013; 89:33. [DOI: 10.1095/biolreprod.112.105692] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
231
|
Hu W, Zhao J, Pei G. Activation of aryl hydrocarbon receptor (ahr) by tranilast, an anti-allergy drug, promotes miR-302 expression and cell reprogramming. J Biol Chem 2013; 288:22972-84. [PMID: 23821545 DOI: 10.1074/jbc.m113.475624] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
MiR-302 has been shown to regulate pluripotency genes and help somatic cell reprogramming. Thus, promotion of endogenous miR-302 expression could be a desirable way to facilitate cell reprogramming. By using a luciferase reporter system of the miR-302 promoter, we screened and found that an anti-allergy drug, tranilast, could significantly promote miR-302 expression. Further experiments revealed that two aryl hydrocarbon receptor (AhR) binding motifs on the miR-302 promoter are critical and that activation of AhR is required for tranilast-induced miR-302 expression. Consistently, not only tranilast but other AhR agonists promoted miR-302 expression. Furthermore, the activation of AhR facilitated cell reprogramming in a miR-302-dependent way. These results elucidate that miR-302 expression can be regulated by AhR and thus provide a strategy for promoting somatic cell reprogramming by AhR ligands.
Collapse
Affiliation(s)
- Wenxiang Hu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | |
Collapse
|
232
|
Duan JJ, Qiu W, Xu SL, Wang B, Ye XZ, Ping YF, Zhang X, Bian XW, Yu SC. Strategies for isolating and enriching cancer stem cells: well begun is half done. Stem Cells Dev 2013; 22:2221-39. [PMID: 23540661 DOI: 10.1089/scd.2012.0613] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cancer stem cells (CSCs) constitute a subpopulation of cancer cells that have the potential for self-renewal, multipotent differentiation, and tumorigenicity. Studies on CSC biology and CSC-targeted therapies depend on CSC isolation and/or enrichment methodologies. Scientists have conducted extensive research in this field since John Dick's group successfully isolated CSCs based on the expression of the CD34 and CD38 surface markers. Progress in CSC research has been greatly facilitated by the enrichment and isolation of these cells. In this review, we summarize the current strategies used in our and other laboratories for CSC isolation and enrichment, including methods based on stem cell surface markers, intracellular enzyme activity, the concentration of reactive oxygen species, the mitochondrial membrane potential, promoter-driven fluorescent protein expression, autofluorescence, suspension/adherent culture, cell division, the identification of side population cells, resistance to cytotoxic compounds or hypoxia, invasiveness/adhesion, immunoselection, and physical property. Although many challenges remain to be overcome, it is reasonable to believe that more reliable, efficient, and convenient methods will be developed in the near future.
Collapse
Affiliation(s)
- Jiang-Jie Duan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
233
|
Kidwai F, Costea DE, Hutchison I, Mackenzie I. The effects of CD44 down-regulation on stem cell properties of head and neck cancer cell lines. J Oral Pathol Med 2013; 42:682-90. [DOI: 10.1111/jop.12076] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2013] [Indexed: 01/23/2023]
Affiliation(s)
- Fahad Kidwai
- Faculty of Dentistry; National University of Singapore; Singapore
| | - Daniela E. Costea
- Section for Pathology; Clinical Institute 1; University of Bergen; Bergen Norway
| | - Iain Hutchison
- Department of Oral and Maxillofacial Surgery; Barts and The London School of Medicine and Dentistry; London UK
| | - Ian Mackenzie
- Blizard Institute; Barts and The London School of Medicine and Dentistry; Queen Mary University of London; London UK
| |
Collapse
|
234
|
Di J, Massuger LFAG, Duiveman-de Boer T, Zusterzeel PLM, Figdor CG, Torensma R. Functional OCT4-specific CD4 + and CD8 + T cells in healthy controls and ovarian cancer patients. Oncoimmunology 2013; 2:e24271. [PMID: 23762805 PMCID: PMC3667911 DOI: 10.4161/onci.24271] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Revised: 03/11/2013] [Accepted: 03/12/2013] [Indexed: 01/17/2023] Open
Abstract
The identification of growth and differentiation pathways that are responsible for the proliferation and survival of cancer stem cells (CSCs) has opened avenues for the discovery of novel therapeutic targets. In the initial phase of an anticancer immune response, T cells specific for tumor-associated antigens develop in patients and, at least under selected circumstances, are able to eliminate malignant cells. However, it remains unknown whether CSC-specific T cells are also operational. We found naturally occurring multifunctional CD4+ and CD8+ T cells specific for the stem cell marker OCT4 among the peripheral blood mononuclear cells (PBMCs) of both healthy individuals and ovarian cancer patients. Moreover, lymphocytes isolated from the ascites of patients affected by ovarian malignancies also contained OCT4-specific T cells. OCT4-reactive CD4+ T cells did not produce interferon γ (IFNγ) and IFNγ-inducible protein 10 (IP-10) but were capable of proliferation upon stimulation with dendritic cells (DCs) loaded with an OCT4-derived peptide or OCT4 mRNA. OCT4-reactive CD8+ cells did not proliferate in response to a similar challenge, yet produced IP-10 as well as sufficient amounts of IFNγ to induce IP-10 . Furthermore, CD8+ cytotoxic T cells were able to release their lysosomal components, as indicated by the mobilization of CD107a. These results demonstrate the existence of anti-CSC specific T cells in ovarian cancer patients.
Collapse
Affiliation(s)
- Jiabo Di
- Department of Tumor Immunology; Nijmegen Centre for Molecular Life Sciences; Radboud University Nijmegen Medical Centre; Nijmegen, The Netherlands
| | | | | | | | | | | |
Collapse
|
235
|
Williams K, Motiani K, Giridhar PV, Kasper S. CD44 integrates signaling in normal stem cell, cancer stem cell and (pre)metastatic niches. Exp Biol Med (Maywood) 2013; 238:324-38. [PMID: 23598979 PMCID: PMC11037417 DOI: 10.1177/1535370213480714] [Citation(s) in RCA: 165] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The stem cell niche provides a regulatory microenvironment for cells as diverse as totipotent embryonic stem cells to cancer stem cells (CSCs) which exhibit stem cell-like characteristics and have the capability of regenerating the bulk of tumor cells while maintaining self-renewal potential. The transmembrane glycoprotein CD44 is a common component of the stem cell niche and exists as a standard isoform (CD44s) and a range of variant isoforms (CD44v) generated though alternative splicing. CD44 modulates signal transduction through post-translational modifications as well as interactions with hyaluronan, extracellular matrix molecules and growth factors and their cognate receptor tyrosine kinases. While the function of CD44 in hematopoietic stem cells has been studied in considerable detail, our knowledge of CD44 function in tissue-derived stem cell niches remains limited. Here we review CD44s and CD44v in both hematopoietic and tissue-derived stem cell niches, focusing on their roles in regulating stem cell behavior including self-renewal and differentiation in addition to cell-matrix interactions and signal transduction during cell migration and tumor progression. Determining the role of CD44 and CD44v in normal stem cell, CSC and (pre)metastatic niches and elucidating their unique functions could provide tools and therapeutic strategies for treating diseases as diverse as fibrosis during injury repair to cancer progression.
Collapse
Affiliation(s)
- Karin Williams
- Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, OH 45267
| | - Karan Motiani
- Division of Urology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267
| | | | - Susan Kasper
- Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, OH 45267
| |
Collapse
|
236
|
Liu K, Lin B, Zhao M, Yang X, Chen M, Gao A, Liu F, Que J, Lan X. The multiple roles for Sox2 in stem cell maintenance and tumorigenesis. Cell Signal 2013; 25:1264-71. [PMID: 23416461 DOI: 10.1016/j.cellsig.2013.02.013] [Citation(s) in RCA: 195] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2013] [Accepted: 02/10/2013] [Indexed: 12/11/2022]
Abstract
The Sry-containing protein Sox2 initially was known to regulate the self-renewal of the mouse and human embryonic stem cells (ESCs). It is also important for the maintenance of stem cells in multiple adult tissues including the brain and trachea, and it is one of the key transcription factors for establishing induced pluripotent stem cells. Recently, overexpression and gene amplification of Sox2 have been associated with the development of squamous cell carcinoma in multiple tissues such as the lung and esophagus. These different roles for Sox2 involve a complicated regulatory networks consisting of microRNAs, kinases and signaling molecules. While the levels of Sox2 are modulated transcriptionally and translationally, post-translational modification is also important for the various functions of Sox2. In clinics, high levels of Sox2 are correlated with poor prognosis and increased proliferation of cancer stem cells. Therefore targeting Sox2 can be potentially explored for a new therapeutic avenue to treat cancers. This review will focus on the different roles for Sox2 in stem cell maintenance and its oncogenic roles in the context of signal transcription and microRNA regulation. We will also review the main upstream and downstream targets of Sox2, which can be potentially used as therapeutic measures to treat cancer with abnormal levels of Sox2.
Collapse
Affiliation(s)
- Kuancan Liu
- Institute for laboratory medicine, Fuzhou General Hospital, PLA, Fuzhou, Fujian, 350025, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
237
|
Abstract
In recent years, many studies have shown that some types of tumors are characterized by the presence of cells with stem-like characteristics, called cancer stem cells (CSCs). These are considered cells that initiate the tumor and are probably responsible for tumor recurrence. CSCs have the capacity for self-renewal, the potential to give rise to one or more cell types within the tumor, and the ability to drive, in a continuous manner, the proliferation of malignant cells. The failure of current cancer therapies can be attributed to the relative ineffectiveness of drugs against CSCs, which remain viable while retaining their full ability to reproduce the tumor. The development of new strategies is currently hampered by the lack of reliable markers to identify CSCs. One promising surface marker of CSCs in head and neck cancer is the CD44 molecule, which has been shown in preliminary studies to have high specificity, although there are discrepant data because its prognostic value may depend on the specific tumor location. More rigorous studies are needed to investigate the usefulness of CD44 expression in head and neck tumors for possible clinical applicability.
Collapse
Affiliation(s)
- Serena Trapasso
- Otolaryngology - Head and Neck Surgery, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | | |
Collapse
|
238
|
Gao R, Li D, Xun J, Zhou W, Li J, Wang J, Liu C, Li X, Shen W, Qiao H, Stupack DG, Luo N. CD44ICD promotes breast cancer stemness via PFKFB4-mediated glucose metabolism. Endocrinology 1983; 8:6248-6262. [PMID: 30613295 PMCID: PMC6299690 DOI: 10.7150/thno.28721] [Citation(s) in RCA: 63] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 10/20/2018] [Indexed: 12/20/2022]
Abstract
CD44 is a single-pass cell surface glycoprotein that is distinguished as the first molecule used to identify cancer stem cells in solid tumors based on its expression. In this regard, the CD44high cell population demonstrates not only the ability to regenerate a heterogeneous tumor, but also the ability to self-regenerate when transplanted into immune-deficient mice. However, the exact role of CD44 in cancer stem cells remains unclear in part because CD44 exists in various isoforms due to alternative splicing. Methods: Gain- and loss-of-function methods in different models were used to investigate the effects of CD44 on breast cancer stemness. Cancer stemness was analyzed by detecting SOX2, OCT4 and NANOG expression, ALDH activity, side population (SP) and sphere formation. Glucose consumption, lactate secretion and reactive oxygen species (ROS) levels were detected to assess glycolysis. Western blot, immunohistochemical staining, ELISA and TCGA dataset analysis were performed to determine the association of CD44ICD and PFKFB4 with clinical cases. A PFKFB4 inhibitor, 5MPN, was used in a xenograft model to inhibit breast cancer development. Results: In this report, we found that the shortest CD44 isoform (CD44s) inhibits breast cancer stemness, whereas the cleaved product of CD44 (CD44ICD) promotes breast cancer stemness. Furthermore, CD44ICD interacts with CREB and binds to the promoter region of PFKFB4, thereby regulating PFKFB4 transcription and expression. The resultant PFKFB4 expression facilitates the glycolysis pathway (vis-à-vis oxidative phosphorylation) and promotes stemness of breast cancer. In addition, we found that CD44ICD and PFKFB4 expressions are generally up-regulated in the tumor portion of breast cancer patient samples. Most importantly, we found that 5MPN (a selective inhibitor of PFKFB4) suppresses CD44ICD-induced tumor development. Conclusion: CD44ICD promotes breast cancer stemness via PFKFB4-mediated glycolysis, and therapies that target PFKFB4 (e.g., 5MPN therapy) may lead to improved outcomes for cancer patients.
Collapse
|