1
|
Joshi G, Basu A. Epigenetic control of cell signalling in cancer stem cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 383:67-88. [PMID: 38359971 DOI: 10.1016/bs.ircmb.2023.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
The self-renewing cancer stem cells (CSCs) represent one of the distinct cell populations occurring in a tumour that can differentiate into multiple lineages. This group of sparsely abundant cells play a vital role in tumour survival and resistance to different treatments during cancer. The lack of exclusive markers associated with CSCs makes diagnosis and prognosis in cancer patients extremely difficult. This calls for the identification of unique regulators and markers for CSCs. Various signalling pathways like the Wnt/β-catenin pathway, Hedgehog pathway, Notch pathway, and TGFβ/BMP play a major role in the regulation and maintenance of CSCs. Epigenetic regulatory mechanisms add another layer of complexity to control these signalling pathways. In this chapter, we discuss about the role of epigenetic mechanisms in regulating the cellular signalling pathways in CSCs. The epigenetic regulatory mechanisms such as DNA methylation, histone modification and microRNAs can modulate the diverse effectors of signalling pathways and consequently the growth, differentiation and tumorigenicity of CSCs. In the end, we briefly discuss the therapeutic potential of targeting these epigenetic regulators and their target genes in CSCs.
Collapse
Affiliation(s)
- Gaurav Joshi
- Institute of Molecular Biology (IMB), Mainz, Germany.
| | - Amitava Basu
- Institute of Molecular Biology (IMB), Mainz, Germany.
| |
Collapse
|
2
|
Srivastava N, Usmani SS, Subbarayan R, Saini R, Pandey PK. Hypoxia: syndicating triple negative breast cancer against various therapeutic regimens. Front Oncol 2023; 13:1199105. [PMID: 37492478 PMCID: PMC10363988 DOI: 10.3389/fonc.2023.1199105] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 06/05/2023] [Indexed: 07/27/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is one of the deadliest subtypes of breast cancer (BC) for its high aggressiveness, heterogeneity, and hypoxic nature. Based on biological and clinical observations the TNBC related mortality is very high worldwide. Emerging studies have clearly demonstrated that hypoxia regulates the critical metabolic, developmental, and survival pathways in TNBC, which include glycolysis and angiogenesis. Alterations to these pathways accelerate the cancer stem cells (CSCs) enrichment and immune escape, which further lead to tumor invasion, migration, and metastasis. Beside this, hypoxia also manipulates the epigenetic plasticity and DNA damage response (DDR) to syndicate TNBC survival and its progression. Hypoxia fundamentally creates the low oxygen condition responsible for the alteration in Hypoxia-Inducible Factor-1alpha (HIF-1α) signaling within the tumor microenvironment, allowing tumors to survive and making them resistant to various therapies. Therefore, there is an urgent need for society to establish target-based therapies that overcome the resistance and limitations of the current treatment plan for TNBC. In this review article, we have thoroughly discussed the plausible significance of HIF-1α as a target in various therapeutic regimens such as chemotherapy, radiotherapy, immunotherapy, anti-angiogenic therapy, adjuvant therapy photodynamic therapy, adoptive cell therapy, combination therapies, antibody drug conjugates and cancer vaccines. Further, we also reviewed here the intrinsic mechanism and existing issues in targeting HIF-1α while improvising the current therapeutic strategies. This review highlights and discusses the future perspectives and the major alternatives to overcome TNBC resistance by targeting hypoxia-induced signaling.
Collapse
Affiliation(s)
- Nityanand Srivastava
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Salman Sadullah Usmani
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Rajasekaran Subbarayan
- Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY, United States
- Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Educations, Chennai, India
| | - Rashmi Saini
- Department of Zoology, Gargi College, University of Delhi, New Delhi, India
| | - Pranav Kumar Pandey
- Dr. R.P. Centre for Opthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
3
|
Revisiting Epithelial Carcinogenesis. Int J Mol Sci 2022; 23:ijms23137437. [PMID: 35806442 PMCID: PMC9267463 DOI: 10.3390/ijms23137437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 12/04/2022] Open
Abstract
The origin of cancer remains one of the most important enigmas in modern biology. This paper presents a hypothesis for the origin of carcinomas in which cellular aging and inflammation enable the recovery of cellular plasticity, which may ultimately result in cancer. The hypothesis describes carcinogenesis as the result of the dedifferentiation undergone by epithelial cells in hyperplasia due to replicative senescence towards a mesenchymal cell state with potentially cancerous behavior. In support of this hypothesis, the molecular, cellular, and histopathological evidence was critically reviewed and reinterpreted when necessary to postulate a plausible generic series of mechanisms for the origin and progression of carcinomas. In addition, the implications of this theoretical framework for the current strategies of cancer treatment are discussed considering recent evidence of the molecular events underlying the epigenetic switches involved in the resistance of breast carcinomas. The hypothesis also proposes an epigenetic landscape for their progression and a potential mechanism for restraining the degree of dedifferentiation and malignant behavior. In addition, the manuscript revisits the gradual degeneration of the nonalcoholic fatty liver disease to propose an integrative generalized mechanistic explanation for the involution and carcinogenesis of tissues associated with aging. The presented hypothesis might serve to understand and structure new findings into a more encompassing view of the genesis of degenerative diseases and may inspire novel approaches for their study and therapy.
Collapse
|
4
|
Wan Kamarul Zaman WS, Nurul AA, Nordin F. Stem Cells and Cancer Stem Cells: The Jekyll and Hyde Scenario and Their Implications in Stem Cell Therapy. Biomedicines 2021; 9:biomedicines9091245. [PMID: 34572431 PMCID: PMC8468168 DOI: 10.3390/biomedicines9091245] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/31/2021] [Accepted: 09/04/2021] [Indexed: 12/12/2022] Open
Abstract
"Jekyll and Hyde" refers to persons with an unpredictably dual personality, who are battling between good and evil within themselves In this regard, even cells consist of good and evil counterparts. Normal stem cells (NSCs) and cancer stem cells (CSCs) are two types of cells that share some similar characteristics but have distinct functions that play a major role in physiological and pathophysiological development. In reality, NSCs such as the adult and embryonic stem cells, are the good cells and the ultimate treatment used in cell therapy. CSCs are the corrupted cells that are a subpopulation of cancer cells within the cancer microenvironment that grow into a massive tumour or malignancy that needs to be treated. Hence, understanding the connection between NSCs and CSCs is important not just in cancer development but also in their therapeutic implication, which is the focus of this review.
Collapse
Affiliation(s)
- Wan Safwani Wan Kamarul Zaman
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur 50603, Malaysia
- Centre for Innovation in Medical Engineering (CIME), Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur 50603, Malaysia
- Correspondence:
| | - Asma Abdullah Nurul
- School of Health Science, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia;
| | - Fazlina Nordin
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Universiti Kebangsaan Malaysia Medical Centre, UKM, Cheras, Kuala Lumpur 56000, Malaysia;
| |
Collapse
|
5
|
Farina AR, Cappabianca LA, Zelli V, Sebastiano M, Mackay AR. Mechanisms involved in selecting and maintaining neuroblastoma cancer stem cell populations, and perspectives for therapeutic targeting. World J Stem Cells 2021; 13:685-736. [PMID: 34367474 PMCID: PMC8316860 DOI: 10.4252/wjsc.v13.i7.685] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/09/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
Pediatric neuroblastomas (NBs) are heterogeneous, aggressive, therapy-resistant embryonal tumours that originate from cells of neural crest (NC) origin and in particular neuroblasts committed to the sympathoadrenal progenitor cell lineage. Therapeutic resistance, post-therapeutic relapse and subsequent metastatic NB progression are driven primarily by cancer stem cell (CSC)-like subpopulations, which through their self-renewing capacity, intermittent and slow cell cycles, drug-resistant and reversibly adaptive plastic phenotypes, represent the most important obstacle to improving therapeutic outcomes in unfavourable NBs. In this review, dedicated to NB CSCs and the prospects for their therapeutic eradication, we initiate with brief descriptions of the unique transient vertebrate embryonic NC structure and salient molecular protagonists involved NC induction, specification, epithelial to mesenchymal transition and migratory behaviour, in order to familiarise the reader with the embryonic cellular and molecular origins and background to NB. We follow this by introducing NB and the potential NC-derived stem/progenitor cell origins of NBs, before providing a comprehensive review of the salient molecules, signalling pathways, mechanisms, tumour microenvironmental and therapeutic conditions involved in promoting, selecting and maintaining NB CSC subpopulations, and that underpin their therapy-resistant, self-renewing metastatic behaviour. Finally, we review potential therapeutic strategies and future prospects for targeting and eradication of these bastions of NB therapeutic resistance, post-therapeutic relapse and metastatic progression.
Collapse
Affiliation(s)
- Antonietta Rosella Farina
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Lucia Annamaria Cappabianca
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Veronica Zelli
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Michela Sebastiano
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Andrew Reay Mackay
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy.
| |
Collapse
|
6
|
Wang W, Fang F, Ozes A, Nephew KP. Targeting Ovarian Cancer Stem Cells by Dual Inhibition of HOTAIR and DNA Methylation. Mol Cancer Ther 2021; 20:1092-1101. [PMID: 33785648 DOI: 10.1158/1535-7163.mct-20-0826] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 01/11/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022]
Abstract
Ovarian cancer is a chemoresponsive tumor with very high initial response rates to standard therapy consisting of platinum/paclitaxel. However, most women eventually develop recurrence, which rapidly evolves into chemoresistant disease. Persistence of ovarian cancer stem cells (OCSCs) at the end of therapy has been shown to contribute to resistant tumors. In this study, we demonstrate that the long noncoding RNA HOTAIR is overexpressed in HGSOC cell lines. Furthermore, HOTAIR expression was upregulated in OCSCs compared with non-CSC, ectopic overexpression of HOTAIR enriched the ALDH+ cell population and HOTAIR overexpression increased spheroid formation and colony-forming ability. Targeting HOTAIR using peptide nucleic acid-PNA3, which acts by disrupting the interaction between HOTAIR and EZH2, in combination with a DNMT inhibitor inhibited OCSC spheroid formation and decreased the percentage of ALDH+ cells. Disrupting HOTAIR-EZH2 with PNA3 in combination with the DNMTi on the ability of OCSCs to initiate tumors in vivo as xenografts was examined. HGSOC OVCAR3 cells were treated with PNA3 in vitro and then implanted in nude mice. Tumor growth, initiation, and stem cell frequency were inhibited. Collectively, these results demonstrate that blocking HOTAIR-EZH2 interaction combined with inhibiting DNA methylation is a potential approach to eradicate OCSCs and block disease recurrence.
Collapse
Affiliation(s)
- Weini Wang
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana
| | - Fang Fang
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana
| | - Ali Ozes
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana
| | - Kenneth P Nephew
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana. .,Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, Indiana
| |
Collapse
|
7
|
Mukaida N, Tanabe Y, Baba T. Cancer non-stem cells as a potent regulator of tumor microenvironment: a lesson from chronic myeloid leukemia. MOLECULAR BIOMEDICINE 2021; 2:7. [PMID: 35006395 PMCID: PMC8607377 DOI: 10.1186/s43556-021-00030-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/17/2021] [Indexed: 01/10/2023] Open
Abstract
A limited subset of human leukemia cells has a self-renewal capacity and can propagate leukemia upon their transplantation into animals, and therefore, are named as leukemia stem cells, in the early 1990’s. Subsequently, cell subpopulations with similar characteristics were detected in various kinds of solid cancers and were denoted as cancer stem cells. Cancer stem cells are presently presumed to be crucially involved in malignant progression of solid cancer: chemoresitance, radioresistance, immune evasion, and metastasis. On the contrary, less attention has been paid to cancer non-stem cell population, which comprise most cancer cells in cancer tissues, due to the lack of suitable markers to discriminate cancer non-stem cells from cancer stem cells. Chronic myeloid leukemia stem cells generate a larger number of morphologically distinct non-stem cells. Moreover, accumulating evidence indicates that poor prognosis is associated with the increases in these non-stem cells including basophils and megakaryocytes. We will discuss the potential roles of cancer non-stem cells in fostering tumor microenvironment, by illustrating the roles of chronic myeloid leukemia non-stem cells including basophils and megakaryocytes in the pathogenesis of chronic myeloid leukemia, a typical malignant disorder arising from leukemic stem cells.
Collapse
Affiliation(s)
- Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan.
| | - Yamato Tanabe
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Tomohisa Baba
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan
| |
Collapse
|
8
|
Chen JB, Kong XF, Lv YY, Qin SC, Sun XJ, Mu F, Lu TY, Xu KC. "Real world survey" of hydrogen-controlled cancer: a follow-up report of 82 advanced cancer patients. Med Gas Res 2020; 9:115-121. [PMID: 31552873 PMCID: PMC6779007 DOI: 10.4103/2045-9912.266985] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Advanced cancer treatment is a huge challenge and new ideas and strategies are required. Hydrogen exerts antioxidant and anti-inflammatory effects that may be exploited to control cancer, the occurrence and progression of which is closely related to peroxidation and inflammation. We conducted a prospective follow-up study of 82 patients with stage III and IV cancer treated with hydrogen inhalation using the "real world evidence" method. After 3-46 months of follow-up, 12 patients died in stage IV. After 4 weeks of hydrogen inhalation, patients reported significant improvements in fatigue, insomnia, anorexia and pain. Furthermore, 41.5% of patients had improved physical status, with the best effect achieved in lung cancer patients and the poorest in patients with pancreatic and gynecologic cancers. Of the 58 cases with one or more abnormal tumor markers elevated, the markers were decreased at 13-45 days (median 23 days) after hydrogen inhalation in 36.2%. The greatest marker decrease was in achieved lung cancer and the lowest in pancreatic and hepatic malignancies. Of the 80 cases with tumors visible in imaging, the total disease control rate was 57.5%, with complete and partial remission appearing at 21-80 days (median 55 days) after hydrogen inhalation. The disease control rate was significantly higher in stage III patients than in stage IV patients (83.0% and 47.7%, respectively), with the lowest disease control rate in pancreatic cancer patients. No hematological toxicity was observed although minor adverse reactions that resolved spontaneously were seen in individual cases. In patients with advanced cancer, inhaled hydrogen can improve patients' quality-of-life and control cancer progression. Hydrogen inhalation is a simple, low-cost treatment with few adverse reactions that warrants further investigation as a strategy for clinical rehabilitation of patients with advanced cancer. The study protocol received ethical approval from the Ethics Committee of Fuda Cancer Hospital of Jinan University on December 7, 2018 (approval number: Fuda20181207).
Collapse
Affiliation(s)
- Ji-Bing Chen
- Department of Cancer Rehabilitation, Fuda Cancer Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Xiao-Feng Kong
- Research Center of Hydrogen Medicine, Xukecheng Health Care Studio of Guangdong Province, Guangzhou, Guangdong Province, China
| | - You-Yong Lv
- Molecular Biology Laboratory of Cancer Hospital, Peking University, Beijing, China
| | - Shu-Cun Qin
- Institute of Hydrogen Medicine, Shandong Medical University, Jinan, Shandong Province, China
| | - Xue-Jun Sun
- Institute of Diving Medicine, Navy Medical University, Shanghai, China
| | - Feng Mu
- Department of Cancer Rehabilitation, Fuda Cancer Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Tian-Yu Lu
- Department of Cancer Rehabilitation, Fuda Cancer Hospital of Jinan University; Research Center of Hydrogen Medicine, Xukecheng Health Care Studio of Guangdong Province, Guangzhou, Guangdong Province, China
| | - Ke-Cheng Xu
- Department of Cancer Rehabilitation, Fuda Cancer Hospital of Jinan University; Research Center of Hydrogen Medicine, Xukecheng Health Care Studio of Guangdong Province, Guangzhou, Guangdong Province, China
| |
Collapse
|
9
|
Basal-Type Breast Cancer Stem Cells Over-Express Chromosomal Passenger Complex Proteins. Cells 2020; 9:cells9030709. [PMID: 32183150 PMCID: PMC7140627 DOI: 10.3390/cells9030709] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 02/27/2020] [Accepted: 02/29/2020] [Indexed: 12/21/2022] Open
Abstract
(1) Aim: In the present paper we analyzed the transcriptome of CSCs (Cancer Stem Cells), in order to find defining molecular processes of breast cancer. (2) Methods: We performed RNA-Seq from CSCs isolated from the basal cell line MDA-MB-468. Enriched processes and networks were studied using the IPA (Ingenuity Pathway Analysis) tool. Validation was performed with qRT-PCR and the analysis of relevant genes was evaluated by overexpression, flow cytometry and in vivo zebrafish studies. Finally, the clinical relevance of these results was assessed using reported cohorts. (3) Results: We found that CSCs presented marked differences from the non-CSCs, including enrichment in transduction cascades related to stemness, cellular growth, proliferation and apoptosis. Interestingly, CSCs overexpressed a module of co-regulated Chromosomal Passenger Proteins including BIRC5 (survivin), INCENP and AURKB. Overexpression of BIRC5 increased the number of CSCs, as assessed by in vitro and in vivo zebrafish xenotransplant analyses. Analysis of previously published cohorts showed that this co-regulated module was not only overexpressed in basal breast tumors but also associated with relapse-free and overall survival in these patients. (4) Conclusions: These results underline the importance of Cancer Stem Cells in breast cancer progression and point toward the possible use of chromosomal passenger proteins as prognostic factors.
Collapse
|
10
|
Caglar HO, Biray Avci C. Alterations of cell cycle genes in cancer: unmasking the role of cancer stem cells. Mol Biol Rep 2020; 47:3065-3076. [DOI: 10.1007/s11033-020-05341-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/22/2020] [Indexed: 02/07/2023]
|
11
|
Sato K, Shimokawa T, Imai T. Difference in Acquired Radioresistance Induction Between Repeated Photon and Particle Irradiation. Front Oncol 2019; 9:1213. [PMID: 31799186 PMCID: PMC6863406 DOI: 10.3389/fonc.2019.01213] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/23/2019] [Indexed: 12/21/2022] Open
Abstract
In recent years, advanced radiation therapy techniques, including stereotactic body radiotherapy and carbon–ion radiotherapy, have progressed to such an extent that certain types of cancer can be treated with radiotherapy alone. The therapeutic outcomes are particularly promising for early stage lung cancer, with results matching those of surgical resection. Nevertheless, patients may still experience local tumor recurrence, which might be exacerbated by the acquisition of radioresistance after primary radiotherapy. Notwithstanding the risk of tumors acquiring radioresistance, secondary radiotherapy is increasingly used to treat recurrent tumors. In this context, it appears essential to comprehend the radiobiological effects of repeated photon and particle irradiation and their underlying cellular and molecular mechanisms in order to achieve the most favorable therapeutic outcome. However, to date, the mechanisms of acquisition of radioresistance in cancer cells have mainly been studied after repeated in vitro X-ray irradiation. By contrast, other critical aspects of radioresistance remain mostly unexplored, including the response to carbon-ion irradiation of X-ray radioresistant cancer cells, the mechanisms of acquisition of carbon-ion resistance, and the consequences of repeated in vivo X-ray or carbon-ion irradiation. In this review, we discuss the underlying mechanisms of acquisition of X-ray and carbon-ion resistance in cancer cells, as well as the phenotypic differences between X-ray and carbon-ion-resistant cancer cells, the biological implications of repeated in vivo X-ray or carbon-ion irradiation, and the main open questions in the field.
Collapse
Affiliation(s)
- Katsutoshi Sato
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, The Tisch Cancer Institute, New York, NY, United States
| | - Takashi Shimokawa
- Department of Accelerator and Medical Physics, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Sciences and Technology, Chiba, Japan
| | - Takashi Imai
- Medical Databank, Department of Radiation Medicine, QST Hospital, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| |
Collapse
|
12
|
Lian YF, Huang YL, Wang JL, Deng MH, Xia TL, Zeng MS, Chen MS, Wang HB, Huang YH. Anillin is required for tumor growth and regulated by miR-15a/miR-16-1 in HBV-related hepatocellular carcinoma. Aging (Albany NY) 2019; 10:1884-1901. [PMID: 30103211 PMCID: PMC6128427 DOI: 10.18632/aging.101510] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/27/2018] [Indexed: 12/26/2022]
Abstract
Anillin (ANLN) is an actin-binding protein essential for assembly of cleavage furrow during cytokinesis. Although reportedly overexpressed in various human cancers, its role in hepatocellular carcinoma (HCC) is unclear. To address this issue, we confirmed that in 436 liver samples obtained from surgically removed HCC tissues, higher ANLN expression was detected in tumor tissues than in adjacent non-tumor tissues of HCC as measured by immunohistochemistry, quantitative real-time PCR and western blotting. Correlation and Kaplan-Meier analysis revealed that patients with higher ANLN expression were associated with worse clinical outcomes and a shorter survival time, respectively. Moreover, ANLN inhibition resulted in growth restraint, reduced colony formation, and a lower sphere number in suspension culture. Mechanistically, ANLN deficiency induced an increasing number of multinucleated cells along with the activation of apoptosis signaling and DNA damage checkpoints. Furthermore, HBV infection increased ANLN expression by inhibiting the expression of microRNA (miR)-15a and miR-16-1, both of which were identified as ANLN upstream repressors by targeting its 3’ untranslated region. Thus, we conclude that ANLN promotes tumor growth by ways of decreased apoptosis and DNA damage. Expression level of ANLN significantly influences the survival probability of HCC patients and may represent a promising prognostic biomarker.
Collapse
Affiliation(s)
- Yi-Fan Lian
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yan-Lin Huang
- Department of Infectious Diseases, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jia-Liang Wang
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Mei-Hai Deng
- Department of Hepatobiliary Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tian-Liang Xia
- State Key Laboratory of Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Mu-Sheng Zeng
- State Key Laboratory of Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Min-Shan Chen
- Department of Hepatobiliary Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hong-Bo Wang
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yue-Hua Huang
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Infectious Diseases, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
13
|
Atashzar MR, Baharlou R, Karami J, Abdollahi H, Rezaei R, Pourramezan F, Zoljalali Moghaddam SH. Cancer stem cells: A review from origin to therapeutic implications. J Cell Physiol 2019; 235:790-803. [PMID: 31286518 DOI: 10.1002/jcp.29044] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/09/2019] [Accepted: 06/11/2019] [Indexed: 02/06/2023]
Abstract
Cancer stem cells (CSCs), also known as tumor-initiating cells (TICs), are elucidated as cells that can perpetuate themselves via autorestoration. These cells are highly resistant to current therapeutic approaches and are the main reason for cancer recurrence. Radiotherapy has made a lot of contributions to cancer treatment. However, despite continuous achievements, therapy resistance and tumor recurrence are still prevalent in most patients. This resistance might be partly related to the existence of CSCs. In the present study, recent advances in the investigation of different biological properties of CSCs, such as their origin, markers, characteristics, and targeting have been reviewed. We have also focused our discussion on radioresistance and adaptive responses of CSCs and their related extrinsic and intrinsic influential factors. In summary, we suggest CSCs as the prime therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Mohammad Reza Atashzar
- Department of Immunology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Rasoul Baharlou
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran.,Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Jafar Karami
- Student Research Committee, Iran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamid Abdollahi
- Department of Radiologic Sciences and Medical Physics, School of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Ramazan Rezaei
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Pourramezan
- Department of Immunology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | | |
Collapse
|
14
|
Kim Y, Choi JW, Lee JH, Kim YS. Spindle assembly checkpoint MAD2 and CDC20 overexpressions and cell-in-cell formation in gastric cancer and its precursor lesions. Hum Pathol 2019; 85:174-183. [DOI: 10.1016/j.humpath.2018.10.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/24/2018] [Accepted: 10/31/2018] [Indexed: 02/07/2023]
|
15
|
Katsura Y, Ohara T, Noma K, Ninomiya T, Kashima H, Kato T, Sato H, Komoto S, Narusaka T, Tomono Y, Xing B, Chen Y, Tazawa H, Kagawa S, Shirakawa Y, Kasai T, Seno M, Matsukawa A, Fujiwara T. A Novel Combination Cancer Therapy with Iron Chelator Targeting Cancer Stem Cells via Suppressing Stemness. Cancers (Basel) 2019; 11:cancers11020177. [PMID: 30717462 PMCID: PMC6406536 DOI: 10.3390/cancers11020177] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 01/28/2019] [Accepted: 01/31/2019] [Indexed: 02/07/2023] Open
Abstract
Excess iron causes cancer and is thought to be related to carcinogenesis and cancer progression including stemness, but the details remain unclear. Here, we hypothesized that stemness in cancer is related to iron metabolism and that regulating iron metabolism in cancer stem cells (CSCs) may be a novel therapy. In this study, we used murine induced pluripotent stem cells that expressed specific stem cell genes such as Nanog, Oct3/4, Sox2, Klf4, and c-Myc, and two human cancer cell lines with similar stem cell gene expression. Deferasirox, an orally available iron chelator, suppressed expression of stemness markers and spherogenesis of cells with high stemness status in vitro. Combination therapy had a marked antitumor effect compared with deferasirox or cisplatin alone. Iron metabolism appears important for maintenance of stemness in CSCs. An iron chelator combined with chemotherapy may be a novel approach via suppressing stemness for CSC targeted therapy.
Collapse
Affiliation(s)
- Yuki Katsura
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
| | - Toshiaki Ohara
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
- Department of Pathology and Experimental Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Kazuhiro Noma
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
| | - Takayuki Ninomiya
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
| | - Hajime Kashima
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
| | - Takuya Kato
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
| | - Hiroaki Sato
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
| | - Satoshi Komoto
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
| | - Toru Narusaka
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
| | - Yasuko Tomono
- Shigei Medical Research Institute, Okayama 701-0202, Japan.
| | - Boyi Xing
- Department of Pathology and Experimental Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Yuehua Chen
- Department of Pathology and Experimental Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Hiroshi Tazawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama 700-8558, Japan.
| | - Shunsuke Kagawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
| | - Yasuhiro Shirakawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
| | - Tomonari Kasai
- School of Bioscience and Biotechnology, Tokyo University of Technology, Tokyo 192-0914, Japan.
| | - Masaharu Seno
- Laboratory of Nano-Biotechnology, Okayama University Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama 700-8530, Japan.
| | - Akihiro Matsukawa
- Department of Pathology and Experimental Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
| |
Collapse
|
16
|
Kim SW, Kim IK, Ha JH, Yeo CD, Kang HH, Kim JW, Lee SH. Normobaric hyperoxia inhibits the progression of lung cancer by inducing apoptosis. Exp Biol Med (Maywood) 2019; 243:739-748. [PMID: 29763371 DOI: 10.1177/1535370218774737] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Hypoxia is a critical characteristic of solid tumors with respect to cancer cell survival, angiogenesis, and metastasis. Hyperoxic treatment has been attempted to reverse hypoxia by enhancing the amount of dissolved oxygen in the plasma. In this study, we evaluated the effects of normobaric hyperoxia on the progression of lung cancer to determine whether oxygen toxicity can be used in cancer therapy. Following a tail vein injection of the Lewis lung carcinoma cells, C57BL/6J mice were exposed to a 24-h normobaric hyperoxia/normoxia cycle for two weeks. In addition, A549 lung cancer cells were incubated in a normobaric hyperoxia chamber for a 24-h period. As a result, the size and number of tumors in the lung decreased significantly with exposure to normobaric hyperoxia in the mouse model. Cell viability, colony-forming ability, migration, and invasion all decreased significantly in A549 cells exposed to normobaric hyperoxia and the normal control group exposed to normobaric hyperoxia showed no significant damage. Oxidative stress was more prominent with exposure to normobaric hyperoxia in cancer cells. A549 cells exposed to normobaric hyperoxia showed a significantly higher cell apoptosis ratio compared with A549 cells without normobaric hyperoxia exposure and normal human lung cells (BEAS-2B cells). The Bax/Bcl-2 mRNA expression ratio also increased significantly. Changes in the key regulators of apoptosis were similar between in vivo and in vitro conditions. The p-ERK level decreased, while the p-JNK level increased, after normobaric hyperoxia exposure in A549 cells. This study demonstrated the role of normobaric hyperoxia in inhibiting lung cancer. Normal tissue and cells showed no significant hyperoxic damage in our experimental setting. The anti-tumor effect of normobaric hyperoxia may due to the increased reactive oxygen species activity and apoptosis, which is related to the mitogen-activated protein kinase pathway. Impact statement Normobaric hyperoxia (NBO) is a feasible therapy for cancer with a low complication rate. Although NBO may be beneficial in cancer treatment, very few studies have been conducted; thus, the evidence is thin. This is the first study to clearly demonstrate morphological changes in lung cancer with NBO exposure and to investigate the underlying mechanisms both in vivo and in vitro. This study will arouse interest in NBO treatment and promote further research.
Collapse
Affiliation(s)
- Sei Won Kim
- 1 Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, St. Paul's Hospital, College of Medicine, The Catholic University of Korea, Seoul 02559, Republic of Korea
| | - In Kyoung Kim
- 1 Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, St. Paul's Hospital, College of Medicine, The Catholic University of Korea, Seoul 02559, Republic of Korea
| | - Jick Hwan Ha
- 2 Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon 21431, Republic of Korea
| | - Chang Dong Yeo
- 3 Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu 11765, Republic of Korea
| | - Hyeon Hui Kang
- 1 Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, St. Paul's Hospital, College of Medicine, The Catholic University of Korea, Seoul 02559, Republic of Korea
| | - Jin Woo Kim
- 3 Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu 11765, Republic of Korea
| | - Sang Haak Lee
- 1 Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, St. Paul's Hospital, College of Medicine, The Catholic University of Korea, Seoul 02559, Republic of Korea.,4 Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
17
|
Araldi RP, Sant’Ana TA, Módolo DG, de Melo TC, Spadacci-Morena DD, de Cassia Stocco R, Cerutti JM, de Souza EB. The human papillomavirus (HPV)-related cancer biology: An overview. Biomed Pharmacother 2018; 106:1537-1556. [DOI: 10.1016/j.biopha.2018.06.149] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 06/24/2018] [Accepted: 06/27/2018] [Indexed: 02/07/2023] Open
|
18
|
Jiménez G, Hackenberg M, Catalina P, Boulaiz H, Griñán-Lisón C, García MÁ, Perán M, López-Ruiz E, Ramírez A, Morata-Tarifa C, Carrasco E, Aguilera M, Marchal JA. Mesenchymal stem cell's secretome promotes selective enrichment of cancer stem-like cells with specific cytogenetic profile. Cancer Lett 2018; 429:78-88. [DOI: 10.1016/j.canlet.2018.04.042] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 04/27/2018] [Accepted: 04/27/2018] [Indexed: 12/15/2022]
|
19
|
Deng C, Lin YX, Qi XK, He GP, Zhang Y, Zhang HJ, Xu M, Feng QS, Bei JX, Zeng YX, Feng L. TNFRSF19 Inhibits TGFβ Signaling through Interaction with TGFβ Receptor Type I to Promote Tumorigenesis. Cancer Res 2018; 78:3469-3483. [PMID: 29735548 DOI: 10.1158/0008-5472.can-17-3205] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 03/27/2018] [Accepted: 04/26/2018] [Indexed: 11/16/2022]
Abstract
Genetic susceptibility underlies the pathogenesis of cancer. We and others have previously identified a novel susceptibility gene TNFRSF19, which encodes an orphan member of the TNF receptor superfamily known to be associated with nasopharyngeal carcinoma (NPC) and lung cancer risk. Here, we show that TNFRSF19 is highly expressed in NPC and is required for cell proliferation and NPC development. However, unlike most of the TNF receptors, TNFRSF19 was not involved in NFκB activation or associated with TRAF proteins. We identified TGFβ receptor type I (TβRI) as a specific binding partner for TNFRSF19. TNFRSF19 bound the kinase domain of TβRI in the cytoplasm, thereby blocking Smad2/3 association with TβRI and subsequent signal transduction. Ectopic expression of TNFRSF19 in normal epithelial cells conferred resistance to the cell-cycle block induced by TGFβ, whereas knockout of TNFRSF19 in NPC cells unleashed a potent TGFβ response characterized by upregulation of Smad2/3 phosphorylation and TGFβ target gene transcription. Furthermore, elevated TNFRSF19 expression correlated with reduced TGFβ activity and poor prognosis in patients with NPC. Our data reveal that gain of function of TNFRSF19 in NPC represents a mechanism by which tumor cells evade the growth-inhibitory action of TGFβ.Significance:TNFRSF19, a susceptibility gene for nasopharyngeal carcinoma and other cancers, functions as a potent inhibitor of the TGFβ signaling pathway.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/13/3469/F1.large.jpg Cancer Res; 78(13); 3469-83. ©2018 AACR.
Collapse
Affiliation(s)
- Chengcheng Deng
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yu-Xin Lin
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xue-Kang Qi
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Gui-Ping He
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yuchen Zhang
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Hao-Jiong Zhang
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Miao Xu
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Qi-Sheng Feng
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jin-Xin Bei
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yi-Xin Zeng
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Lin Feng
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| |
Collapse
|
20
|
Baicalin hydrate inhibits cancer progression in nasopharyngeal carcinoma by affecting genome instability and splicing. Oncotarget 2017; 9:901-914. [PMID: 29416665 PMCID: PMC5787522 DOI: 10.18632/oncotarget.22868] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 09/20/2017] [Indexed: 01/08/2023] Open
Abstract
Baicalin hydrate (BH), a natural compound, has been investigated for many years because of its traditional medicinal properties. However, the anti-tumor activities of BH and its epigenetic role in NPC have not been elucidated. In this study, we identified that BH inhibits NPC cell growth in vivo and in vitro by inducing apoptosis and cell cycle arrest. BH epigenetically regulated genome instability by up-regulating the expression of satellite 2 (Sat2), alpha satellite (α-Sat), and major satellite (Major-Sat). BH also increased the level of IKKα, Suv39H1, and H3K9me3 and decreased LSH expression. Interestingly, BH promoted the splicing of Suv39H1 via the enhancement of m6A RNA methylation, rather than DNA methylation. Taken together, our results demonstrated that BH has an anti-tumor role in NPC and revealed a unique role of BH in genome instability and splicing in response to DNA damage.
Collapse
|
21
|
Qin YY, Zhou CZ, Zhu Z, Guan WJ, Lin XQ, Liu CL, Zhou YM, Wu H, Gu YY, Zhang DH, Ou-Yang M, Zhong NS, Li SY. Case report: dermatomyositis associated with lung cancer with heterogeneous morphology. J Thorac Dis 2017; 9:E1110-E1117. [PMID: 29312776 DOI: 10.21037/jtd.2017.11.107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Dermatomyositis (DM) complicated with non-small cell lung cancer (NSCLC) is not rare, and could rapidly develop into severe lung cancer [performance-status score (PS) between 2 and 4]. Moreover, tumor has remarkable heterogeneity, and it is not possible to properly target treatments in cases of relapse without knowing pathological diagnosis. We retrospectively analyzed the diagnosis and treatment of a patient with DM complicated with NSCLC, which developed into severe lung cancer with heterogeneity of the tumor during chemotherapy. In this report, we addressed that in patients with severe lung cancer, both the cancer and factors associated with exacerbation should be simultaneously managed to reduce the PS score and avoid unnecessary delay. A second biopsy is important for proper management of the tumor with heterogeneity.
Collapse
Affiliation(s)
- Yin-Yin Qin
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong 510120, China
| | - Cheng-Zhi Zhou
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong 510120, China
| | - Zheng Zhu
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong 510120, China
| | - Wei-Jie Guan
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong 510120, China
| | - Xin-Qing Lin
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong 510120, China
| | - Chun-Li Liu
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong 510120, China
| | - Yu-Ming Zhou
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong 510120, China
| | - Hua Wu
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong 510120, China
| | - Ying-Ying Gu
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong 510120, China
| | - De-Hua Zhang
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong 510120, China
| | - Ming Ou-Yang
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong 510120, China
| | - Nan-Shan Zhong
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong 510120, China
| | - Shi-Yue Li
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong 510120, China
| |
Collapse
|
22
|
Liang Y, Ahmed M, Guo H, Soares F, Hua JT, Gao S, Lu C, Poon C, Han W, Langstein J, Ekram MB, Li B, Davicioni E, Takhar M, Erho N, Karnes RJ, Chadwick D, van der Kwast T, Boutros PC, Arrowsmith CH, Feng FY, Joshua AM, Zoubeidi A, Cai C, He HH. LSD1-Mediated Epigenetic Reprogramming Drives CENPE Expression and Prostate Cancer Progression. Cancer Res 2017; 77:5479-5490. [PMID: 28916652 DOI: 10.1158/0008-5472.can-17-0496] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 06/28/2017] [Accepted: 08/17/2017] [Indexed: 12/31/2022]
Abstract
Androgen receptor (AR) signaling is a key driver of prostate cancer, and androgen-deprivation therapy (ADT) is a standard treatment for patients with advanced and metastatic disease. However, patients receiving ADT eventually develop incurable castration-resistant prostate cancer (CRPC). Here, we report that the chromatin modifier LSD1, an important regulator of AR transcriptional activity, undergoes epigenetic reprogramming in CRPC. LSD1 reprogramming in this setting activated a subset of cell-cycle genes, including CENPE, a centromere binding protein and mitotic kinesin. CENPE was regulated by the co-binding of LSD1 and AR to its promoter, which was associated with loss of RB1 in CRPC. Notably, genetic deletion or pharmacological inhibition of CENPE significantly decreases tumor growth. Our findings show how LSD1-mediated epigenetic reprogramming drives CRPC, and they offer a mechanistic rationale for its therapeutic targeting in this disease. Cancer Res; 77(20); 5479-90. ©2017 AACR.
Collapse
Affiliation(s)
- Yi Liang
- Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario, Canada
| | - Musaddeque Ahmed
- Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario, Canada
| | - Haiyang Guo
- Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario, Canada
| | - Fraser Soares
- Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario, Canada
| | - Junjie T Hua
- Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Shuai Gao
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
| | - Catherine Lu
- Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario, Canada
| | - Christine Poon
- Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario, Canada
| | - Wanting Han
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
| | - Jens Langstein
- Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario, Canada
- German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Muhammad B Ekram
- Department of Medical Oncology, The Center for Functional Cancer Epigenetics, Dana Farber Cancer Institute, Boston, Massachusetts
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Brian Li
- Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario, Canada
| | - Elai Davicioni
- Research & Development, GenomeDx Biosciences Inc., Vancouver BC, Canada
| | - Mandeep Takhar
- Research & Development, GenomeDx Biosciences Inc., Vancouver BC, Canada
| | - Nicholas Erho
- Research & Development, GenomeDx Biosciences Inc., Vancouver BC, Canada
| | | | - Dianne Chadwick
- UHN Program in BioSpecimen Sciences, Department of Pathology, University Health Network, Toronto, Ontario, Canada
| | - Theodorus van der Kwast
- Department of Pathology and Laboratory Medicine, Toronto General Hospital/University Health Network, Toronto, Canada
| | - Paul C Boutros
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Cheryl H Arrowsmith
- Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario, Canada
- Structural Genomics Consortium, University of Toronto, Toronto, Ontario, Canada
| | - Felix Y Feng
- Department of Radiation Oncology, University of California at San Francisco, San Francisco, California
- Department of Urology, University of California at San Francisco, San Francisco, California
- Department of Medicine, University of California at San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California
| | - Anthony M Joshua
- Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario, Canada
- Kinghorn Cancer Centre, St Vincent's Hospital, Sydney, Australia
| | - Amina Zoubeidi
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Changmeng Cai
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
| | - Housheng H He
- Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
23
|
Wang W, Zhang Y, Chen R, Tian Z, Zhai Y, Janz S, Gu C, Yang Y. Chromosomal instability and acquired drug resistance in multiple myeloma. Oncotarget 2017; 8:78234-78244. [PMID: 29100463 PMCID: PMC5652852 DOI: 10.18632/oncotarget.20829] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 08/26/2017] [Indexed: 12/29/2022] Open
Abstract
Chromosomal instability (CIN) is an important hallmark of human cancer. CIN not only contributes to all stages of tumor development (initiation, promotion and progression) but also drives, in large measure, the acquisition of drug resistance by cancer cells. Although CIN is a cornerstone of the complex mutational architecture that underlies neoplastic cell development and tumor heterogeneity and has been tightly associated with treatment responses and survival of cancer patients, it may be one of the least understood features of the malignant phenotype in terms of genetic pathways and molecular mechanisms. Here we review new insights into the type of CIN seen in multiple myeloma (MM), a blood cancer of terminally differentiated, immunoglobulin-producing B-lymphocytes called plasma cells that remains incurable in the great majority of cases. We will consider bona fide myeloma CIN genes, methods for measuring CIN in myeloma cells, and novel approaches to CIN-targeted treatments of patients with myeloma. The new findings generate optimism that enhanced understanding of CIN will lead to the design and testing of new therapeutic strategies to overcome drug resistance in MM in the not-so-distant future.
Collapse
Affiliation(s)
- Wang Wang
- The Third Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yi Zhang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Ruini Chen
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhidan Tian
- Department of Pathology, Nanjing First Hospital, Nanjing, 210006, China
| | - Yongpin Zhai
- Department of Hematology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210002, China
| | - Siegfried Janz
- Department of Pathology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, 52242, USA
| | - Chunyan Gu
- The Third Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ye Yang
- The Third Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| |
Collapse
|
24
|
Chen X, Liao R, Li D, Sun J. Induced cancer stem cells generated by radiochemotherapy and their therapeutic implications. Oncotarget 2017; 8:17301-17312. [PMID: 28038467 PMCID: PMC5370042 DOI: 10.18632/oncotarget.14230] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 12/13/2016] [Indexed: 12/26/2022] Open
Abstract
Local and distant recurrence of malignant tumors following radio- and/or chemotherapy correlates with poor prognosis of patients. Among the reasons for cancer recurrence, preexisting cancer stem cells (CSCs) are considered the most likely cause due to their properties of self-renewal, pluripotency, plasticity and tumorigenicity. It has been demonstrated that preexisting cancer stem cells derive from normal stem cells and differentiated somatic cells that undergo transformation and dedifferentiation respectively under certain conditions. However, recent studies have revealed that cancer stem cells can also be induced from non-stem cancer cells by radiochemotherapy, constituting the subpopulation of induced cancer stem cells (iCSCs). These findings suggest that radiochemotherapy has the side effect of directly transforming non-stem cancer cells into induced cancer stem cells, possibly contributing to tumor recurrence and metastasis. Therefore, drugs targeting cancer stem cells or preventing dedifferentiation of non-stem cancer cells can be combined with radiochemotherapy to improve its antitumor efficacy. The current review is to investigate the mechanisms by which induced cancer stem cells are generated by radiochemotherapy and hence provide new strategies for cancer treatment.
Collapse
Affiliation(s)
- Xiewan Chen
- Medical English Department, College of Basic Medicine, Third Military Medical University, Chongqing, China.,Cancer Institute of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Rongxia Liao
- Medical English Department, College of Basic Medicine, Third Military Medical University, Chongqing, China
| | - Dezhi Li
- Cancer Institute of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Jianguo Sun
- Cancer Institute of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
25
|
Wang P, Wan WW, Xiong SL, Feng H, Wu N. Cancer stem-like cells can be induced through dedifferentiation under hypoxic conditions in glioma, hepatoma and lung cancer. Cell Death Discov 2017; 3:16105. [PMID: 28179999 PMCID: PMC5253691 DOI: 10.1038/cddiscovery.2016.105] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/15/2016] [Accepted: 12/17/2016] [Indexed: 12/11/2022] Open
Abstract
Traditional studies have shown that transcription factors, including SOX-2, OCT-4, KLF-4, Nanog and Lin-28A, contribute to the dedifferentiation and reprogramming process in normal tissues. Hypoxia is a physiological phenomenon that exists in tumors and promotes the expression of SOX-2, OCT-4, KLF-4, Nanog and Lin-28A. Therefore, an interesting question is whether hypoxia as a stimulating factor promotes the process of dedifferentiation and induces the formation of cancer stem-like cells. Studies have shown that OCT-4 and Nanog overexpression induced the formation of cancer stem cell-like cells through dedifferentiation and enhanced malignancy in lung adenocarcinoma, and reprogramming SOX-2 in pancreatic cancer cells also promoted the dedifferentiation process. Therefore, we investigated this phenomenon in glioma, lung cancer and hepatoma cells and found that the transcription factors mentioned above were highly expressed under hypoxic conditions and induced the formation of spheres, which exhibited asymmetric division and cell cycle arrest. The dedifferentiation process induced by hypoxia highlights a new pattern of cancer development and recurrence, demonstrating that all kinds of cancer cells and the hypoxic microenvironment should be taken into consideration when developing tumor therapies.
Collapse
Affiliation(s)
- Pan Wang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University , Chongqing 400038, China
| | - Wen-Wu Wan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University , Chongqing 400038, China
| | | | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University , Chongqing 400038, China
| | - Nan Wu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University , Chongqing 400038, China
| |
Collapse
|
26
|
Amaro A, Chiara S, Pfeffer U. Molecular evolution of colorectal cancer: from multistep carcinogenesis to the big bang. Cancer Metastasis Rev 2016; 35:63-74. [PMID: 26947218 DOI: 10.1007/s10555-016-9606-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Colorectal cancer is characterized by exquisite genomic instability either in the form of microsatellite instability or chromosomal instability. Microsatellite instability is the result of mutation of mismatch repair genes or their silencing through promoter methylation as a consequence of the CpG island methylator phenotype. The molecular causes of chromosomal instability are less well characterized. Genomic instability and field cancerization lead to a high degree of intratumoral heterogeneity and determine the formation of cancer stem cells and epithelial-mesenchymal transition mediated by the TGF-β and APC pathways. Recent analyses using integrated genomics reveal different phases of colorectal cancer evolution. An initial phase of genomic instability that yields many clones with different mutations (big bang) is followed by an important, previously not detected phase of cancer evolution that consists in the stabilization of several clones and a relatively flat outgrowth. The big bang model can best explain the coexistence of several stable clones and is compatible with the fact that the analysis of the bulk of the primary tumor yields prognostic information.
Collapse
Affiliation(s)
- Adriana Amaro
- Molecular Pathology, IRCCS AOU San Martino-IST-Istituto Nazionale per la Ricerca sul Cancro, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Silvana Chiara
- Medical Oncology, IRCCS AOU San Martino-IST-Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Ulrich Pfeffer
- Molecular Pathology, IRCCS AOU San Martino-IST-Istituto Nazionale per la Ricerca sul Cancro, Largo Rosanna Benzi 10, 16132, Genoa, Italy.
| |
Collapse
|
27
|
Hsu CL, Chung FH, Chen CH, Hsu TT, Liu SM, Chung DS, Hsu YF, Chen CL, Ma N, Lee HC. Genotypes of cancer stem cells characterized by epithelial-to-mesenchymal transition and proliferation related functions. Sci Rep 2016; 6:32523. [PMID: 27597445 PMCID: PMC5011650 DOI: 10.1038/srep32523] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 08/08/2016] [Indexed: 12/13/2022] Open
Abstract
Cancer stem cells (CSCs), or cancer cells with stem cell-like properties, generally exhibit drug resistance and have highly potent cancer inducing capabilities. Genome-wide expression data collected at public repositories over the last few years provide excellent material for studies that can lead to insights concerning the molecular and functional characteristics of CSCs. Here, we conducted functional genomic studies of CSC based on fourteen PCA-screened high quality public CSC whole genome gene expression datasets and, as control, four high quality non-stem-like cancer cell and non-cancerous stem cell datasets from the Gene Expression Omnibus database. A total of 6,002 molecular signatures were taken from the Molecular Signatures Database and used to characterize the datasets, which, under two-way hierarchical clustering, formed three genotypes. Type 1, consisting of mainly glia CSCs, had significantly enhanced proliferation, and significantly suppressed epithelial-mesenchymal transition (EMT), related functions. Type 2, mainly breast CSCs, had significantly enhanced EMT, but not proliferation, related functions. Type 3, composed of ovarian, prostate, and colon CSCs, had significantly suppressed proliferation related functions and mixed expressions on EMT related functions.
Collapse
Affiliation(s)
- Chueh-Lin Hsu
- Institute of Systems Biology and Bioinformatics, Department of Biomedical Science and Engineering, National Central University, Zhongli, 32001, Taiwan
| | - Feng-Hsiang Chung
- Institute of Systems Biology and Bioinformatics, Department of Biomedical Science and Engineering, National Central University, Zhongli, 32001, Taiwan
| | - Chih-Hao Chen
- Institute of Systems Biology and Bioinformatics, Department of Biomedical Science and Engineering, National Central University, Zhongli, 32001, Taiwan
| | - Tzu-Ting Hsu
- Institute of Systems Biology and Bioinformatics, Department of Biomedical Science and Engineering, National Central University, Zhongli, 32001, Taiwan
| | - Szu-Mam Liu
- Institute of Systems Biology and Bioinformatics, Department of Biomedical Science and Engineering, National Central University, Zhongli, 32001, Taiwan
| | - Dao-Sheng Chung
- Department of Radiation Oncology, Landseed Hospital, Taoyuan, 324, Taiwan
| | - Ya-Fen Hsu
- Department of Surgery, Landseed Hospital, Taoyuan, 324, Taiwan
| | - Chien-Lung Chen
- Department of Nephrology, Landseed Hospital, Taoyuan, 324, Taiwan
| | - Nianhan Ma
- Institute of Systems Biology and Bioinformatics, Department of Biomedical Science and Engineering, National Central University, Zhongli, 32001, Taiwan
| | - Hoong-Chien Lee
- Institute of Systems Biology and Bioinformatics, Department of Biomedical Science and Engineering, National Central University, Zhongli, 32001, Taiwan.,Department of Physics, Chung Yuan Christian University, Zhongli, 32023, Taiwan.,Center for Dynamical Biomarkers and Translational Medicine, National Central University, Zhongli, 32001, Taiwan
| |
Collapse
|
28
|
Schneider N, Hallin M, Thway K. STAT6 Loss in Dedifferentiated Solitary Fibrous Tumor. Int J Surg Pathol 2016; 25:58-60. [PMID: 27189111 DOI: 10.1177/1066896916650257] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
29
|
Abstract
NOP14, which is functionally conserved among eukaryotes, has been implicated in cancer development. Here, we show that NOP14 is poorly expressed in breast cancer cells and invasive breast cancer tissues. In vivo and in vitro studies indicated that NOP14 suppressed the tumorigenesis and metastasis of breast cancer cells. Further investigations revealed that NOP14 enhanced ERα expression and inhibited the Wnt/β-catenin pathway by up-regulating NRIP1 expression. Survival analysis indicated that low NOP14 expression was significantly associated with poor overall survival (P = 0.0006) and disease-free survival (P = 0.0007), suggesting that NOP14 is a potential prognostic factor in breast cancer. Taken together, our findings reveal that NOP14 may suppress breast cancer progression and provide new insights into the development of targeted therapeutic agents for breast cancer.
Collapse
|
30
|
Tsukamoto Y, Ohtsu N, Echizenya S, Otsuguro S, Ogura R, Natsumeda M, Isogawa M, Aoki H, Ichikawa S, Sakaitani M, Matsuda A, Maenaka K, Fujii Y, Kondo T. Chemical Screening Identifies EUrd as a Novel Inhibitor Against Temozolomide-Resistant Glioblastoma-Initiating Cells. Stem Cells 2016; 34:2016-25. [DOI: 10.1002/stem.2380] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 03/29/2016] [Accepted: 04/04/2016] [Indexed: 11/09/2022]
Affiliation(s)
- Yoshihiro Tsukamoto
- Division of Stem Cell Biology; Institute for Genetic Medicine, Hokkaido University; Sapporo Japan
- Department of Neurosurgery; Brain Research Institute, Niigata University; Niigata Japan
| | - Naoki Ohtsu
- Division of Stem Cell Biology; Institute for Genetic Medicine, Hokkaido University; Sapporo Japan
| | - Smile Echizenya
- Division of Stem Cell Biology; Institute for Genetic Medicine, Hokkaido University; Sapporo Japan
- Department of Neurosurgery; Hokkaido University Graduate School of Medicine; Sapporo Japan
| | - Satoko Otsuguro
- Center for Research and Education on Drug Discovery; Hokkaido University; Sapporo Japan
| | - Ryosuke Ogura
- Department of Neurosurgery; Brain Research Institute, Niigata University; Niigata Japan
| | - Manabu Natsumeda
- Department of Neurosurgery; Brain Research Institute, Niigata University; Niigata Japan
| | - Mizuho Isogawa
- Department of Neurosurgery; Brain Research Institute, Niigata University; Niigata Japan
| | - Hiroshi Aoki
- Department of Neurosurgery; Brain Research Institute, Niigata University; Niigata Japan
| | - Satoshi Ichikawa
- Center for Research and Education on Drug Discovery; Hokkaido University; Sapporo Japan
| | - Masahiro Sakaitani
- Center for Research and Education on Drug Discovery; Hokkaido University; Sapporo Japan
| | - Akira Matsuda
- Center for Research and Education on Drug Discovery; Hokkaido University; Sapporo Japan
| | - Katsumi Maenaka
- Center for Research and Education on Drug Discovery; Hokkaido University; Sapporo Japan
| | - Yukihiko Fujii
- Department of Neurosurgery; Brain Research Institute, Niigata University; Niigata Japan
| | - Toru Kondo
- Division of Stem Cell Biology; Institute for Genetic Medicine, Hokkaido University; Sapporo Japan
| |
Collapse
|
31
|
Qin L, Yin YT, Zheng FJ, Peng LX, Yang CF, Bao YN, Liang YY, Li XJ, Xiang YQ, Sun R, Li AH, Zou RH, Pei XQ, Huang BJ, Kang TB, Liao DF, Zeng YX, Williams BO, Qian CN. WNT5A promotes stemness characteristics in nasopharyngeal carcinoma cells leading to metastasis and tumorigenesis. Oncotarget 2016; 6:10239-52. [PMID: 25823923 PMCID: PMC4496352 DOI: 10.18632/oncotarget.3518] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Accepted: 02/17/2015] [Indexed: 01/20/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) has the highest metastasis rate among head and neck cancers with unclear mechanism. WNT5A belongs to the WNT family of cysteine-rich secreted glycoproteins. Our previous high-throughput gene expression profiling revealed that WNT5A was up-regulated in highly metastatic cells. In the present study, we first confirmed the elevated expression of WNT5A in metastatic NPC tissues at both the mRNA and protein levels. We then found that WNT5A promoted epithelial-mesenchymal transition (EMT) in NPC cells, induced the accumulation of CD24-CD44+ cells and side population, which are believed to be cancer stem cell characteristics. Moreover, WNT5A promoted the migration and invasion of NPC cells in vitro, while in vivo treatment with recombinant WNT5A promoted lung metastasis. Knocking down WNT5A diminished NPC tumorigenesis in vivo. When elevated expression of WNT5A coincided with the elevated expression of vimentin in the primary NPC, the patients had a poorer prognosis. Among major signaling pathways, protein kinase C (PKC) signaling was activated by WNT5A in NPC cells. A positive feedback loop between WNT5A and phospho-PKC to promote EMT was also revealed. Taken together, these data suggest that WNT5A is an important molecule in promoting stem cell characteristics in NPC, leading to tumorigenesis and metastasis.
Collapse
Affiliation(s)
- Li Qin
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Hunan Province Engineering Research Center of Bioactive Substance Discovery of Traditional Chinese Medicine, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, China.,Division of Pharmacoproteomics, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan, China
| | - Yan-Tao Yin
- Division of Pharmacoproteomics, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan, China
| | - Fang-Jing Zheng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Radiotherapy, Ningde Municipal Hospital, Fujian Medical University Affiliated Hospital, Ningde, Fujian, China
| | - Li-Xia Peng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Chang-Fu Yang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ying-Na Bao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ying-Ying Liang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xin-Jian Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Brain Tumor Center and Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yan-Qun Xiang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Rui Sun
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - An-Hua Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Ultrasonography, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Ru-Hai Zou
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Ultrasonography, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xiao-Qing Pei
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Ultrasonography, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Bi-Jun Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Tie-Bang Kang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Duan-Fang Liao
- Division of Stem Cell Regulation and Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yi-Xin Zeng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Bart O Williams
- Laboratory of Cell Signaling and Carcinogenesis, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Chao-Nan Qian
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| |
Collapse
|
32
|
Han JY, Han YK, Park GY, Kim SD, Lee CG. Bub1 is required for maintaining cancer stem cells in breast cancer cell lines. Sci Rep 2015; 5:15993. [PMID: 26522589 PMCID: PMC4629164 DOI: 10.1038/srep15993] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 10/06/2015] [Indexed: 12/15/2022] Open
Abstract
Breast cancer is a leading cause of death among women worldwide due to therapeutic resistance and cancer recurrence. Cancer stem cells are believed to be responsible for resistance and recurrence. Many efforts to overcome resistance and recurrence by regulating cancer stem cells are ongoing. Bub1 (Budding uninhibited by benzimidazoles 1) is a mitotic checkpoint serine/threonine kinase that plays an important role in chromosome segregation. Bub1 expression is correlated with a poor clinical prognosis in patients with breast cancer. We identified that depleting Bub1 using shRNAs reduces cancer stem cell potential of the MDA-MB-231 breast cancer cell line, resulting in inhibited formation of xenografts in immunocompromised mice. These results suggest that Bub1 may be associated with cancer stem cell potential and could be a target for developing anti-breast cancer stem cell therapies.
Collapse
Affiliation(s)
- Jeong Yoon Han
- Research Center, Dongnam Institute of Radiological &Medical Sciences, Busan 619-953, ROK
| | - Yu Kyeong Han
- Research Center, Dongnam Institute of Radiological &Medical Sciences, Busan 619-953, ROK.,Research Center, Dongnam Institute of Radiological &Medical Sciences, Busan 619-953, ROK
| | - Ga-Young Park
- Research Center, Dongnam Institute of Radiological &Medical Sciences, Busan 619-953, ROK.,Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan 609-735, ROK
| | - Sung Dae Kim
- Research Center, Dongnam Institute of Radiological &Medical Sciences, Busan 619-953, ROK
| | - Chang Geun Lee
- Research Center, Dongnam Institute of Radiological &Medical Sciences, Busan 619-953, ROK
| |
Collapse
|
33
|
Deng CC, Xu M, Li J, Luo XL, Zhu YJ, Jiang R, Zhang MX, Lei JJ, Lian YF, Zou X, You R, Chen LZ, Feng QS, Bei JX, Chen MY, Zeng YX. Unconjugated Bilirubin Is a Novel Prognostic Biomarker for Nasopharyngeal Carcinoma and Inhibits Its Metastasis via Antioxidation Activity. Cancer Prev Res (Phila) 2015; 9:180-8. [DOI: 10.1158/1940-6207.capr-15-0257] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 09/16/2015] [Indexed: 11/16/2022]
|
34
|
Qiu H, Fang X, Luo Q, Ouyang G. Cancer stem cells: a potential target for cancer therapy. Cell Mol Life Sci 2015; 72:3411-24. [PMID: 25967289 PMCID: PMC11113644 DOI: 10.1007/s00018-015-1920-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 04/08/2015] [Accepted: 04/28/2015] [Indexed: 02/06/2023]
Abstract
Current evidence indicates that a subpopulation of cancer cells, named cancer stem cells (CSCs) or tumor-initiating cells, are responsible for the initiation, growth, metastasis, therapy resistance and recurrence of cancers. CSCs share core regulatory pathways with normal stem cells; however, CSCs rely on distinct reprogrammed pathways to maintain stemness and to contribute to the progression of cancers. The specific targeting of CSCs, together with conventional chemotherapy or radiotherapy, may achieve stable remission or cure cancer. Therefore, the identification of CSCs and a better understanding of the complex characteristics of CSCs will provide invaluable diagnostic, therapeutic and prognostic targets for clinical application. In this review, we will introduce the dysregulated properties of CSCs in cancers and discuss the possible challenges in targeting CSCs for cancer treatment.
Collapse
Affiliation(s)
- Hong Qiu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, 361102 China
| | - Xiaoguang Fang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, 361102 China
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195 USA
| | - Qi Luo
- Department of Surgical Oncology, First Affiliated Hospital of Xiamen University, Xiamen, 361003 China
| | - Gaoliang Ouyang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, 361102 China
| |
Collapse
|
35
|
Dagrada GP, Spagnuolo RD, Mauro V, Tamborini E, Cesana L, Gronchi A, Stacchiotti S, Pierotti MA, Negri T, Pilotti S. Solitary fibrous tumors: loss of chimeric protein expression and genomic instability mark dedifferentiation. Mod Pathol 2015; 28:1074-83. [PMID: 26022454 DOI: 10.1038/modpathol.2015.70] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/25/2015] [Indexed: 01/21/2023]
Abstract
Solitary fibrous tumors, which are characterized by their broad morphological spectrum and unpredictable behavior, are rare mesenchymal neoplasias that are currently divided into three main variants that have the NAB2-STAT6 gene fusion as their unifying molecular lesion: usual, malignant and dedifferentiated solitary fibrous tumors. The aims of this study were to validate molecular and immunohistochemical/biochemical approaches to diagnose the range of solitary fibrous tumors by focusing on the dedifferentiated variant, and to reveal the genetic events associated with dedifferentiation by integrating the findings of array comparative genomic hybridization. We studied 29 usual, malignant and dedifferentiated solitary fibrous tumors from 24 patients (including paired samples from five patients whose tumors progressed to the dedifferentiated form) by means of STAT6 immunohistochemistry and (when frozen material was available) reverse-transcriptase polymerase chain reaction and biochemistry. In addition, the array comparative genomic hybridization findings were used to profile 12 tumors from nine patients. The NAB2/STAT6 fusion was detected in all of the tumors, but immunohistochemistry and western blotting indicated that chimeric protein expression was atypical or absent in 9 out of 11 dedifferentiated tumors. The comparative genomic hybridization results revealed that the usual and malignant solitary fibrous tumors had a simple profile, whereas the genome of the dedifferentiated tumors was complex and unstable, and suggested that 13q and 17p deletions and TP53 mutations may be present in malignant lesions before the full expression of a dedifferentiated phenotype. Solitary fibrous tumor dedifferentiation is associated with the loss of chimeric oncoprotein expression, genomic instability, and cell decommitment and reprogramming. The assessment of dedifferentiated solitary fibrous tumors is based on the presence of the fusion transcripts and, in principle, negative STAT6 immunohistochemistry should not rule out a diagnosis of solitary fibrous tumor.
Collapse
Affiliation(s)
- Gian P Dagrada
- Laboratory of Experimental Molecular Pathology, Department of Diagnostic Pathology and Laboratory, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Rosalin D Spagnuolo
- Laboratory of Experimental Molecular Pathology, Department of Diagnostic Pathology and Laboratory, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Valentina Mauro
- Laboratory of Experimental Molecular Pathology, Department of Diagnostic Pathology and Laboratory, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elena Tamborini
- Department of Diagnostic Pathology and Laboratory, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Luca Cesana
- Department of Diagnostic Pathology and Laboratory, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessandro Gronchi
- Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvia Stacchiotti
- Adult Mesenchymal Tumor Medical Oncology Unit, Cancer Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marco A Pierotti
- Scientific Directorate, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Tiziana Negri
- Laboratory of Experimental Molecular Pathology, Department of Diagnostic Pathology and Laboratory, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvana Pilotti
- Laboratory of Experimental Molecular Pathology, Department of Diagnostic Pathology and Laboratory, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
36
|
Liao D, Zhong L, Duan T, Zhang RH, Wang X, Wang G, Hu K, Lv X, Kang T. Aspirin Suppresses the Growth and Metastasis of Osteosarcoma through the NF-κB Pathway. Clin Cancer Res 2015. [PMID: 26202947 DOI: 10.1158/1078-0432.ccr-15-0198] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Aspirin has recently been reported to reduce both the incidence and the risk of metastasis in colon cancer. However, there is no evidence at the cellular levels or in the animal models for such an effect of aspirin on cancer metastasis. EXPERIMENTAL DESIGN MTT assay, colony formation assay, and apoptosis assay were employed to analyze the effects of aspirin on the osteosarcoma cell viability in vitro. The NF-κB activity was measured by the NF-κB p65 luciferase reporter. Western blotting was used to analyze the proteins in cells. The migration and invasion abilities of osteosarcoma cells in vitro were measured by the Transwell assay. Xenograft-bearing mice were used to assess the roles of aspirin in both tumor growth and metastasis of osteosarcoma in vivo (n = 5-8 mice/group). An unpaired Student t test or ANOVA with the Bonferroni post hoc test were used for the statistical comparisons. RESULTS Aspirin reduced cell viability in a dose- and time-dependent manner in osteosarcoma cell lines, and aspirin synergistically sensitized osteosarcoma cells to cisplatin (DDP) in vitro and in vivo (P < 0.001). Moreover, aspirin markedly repressed the migration and invasion of osteosarcoma cells in vitro (P < 0.001), and dramatically diminished the occurrence of osteosarcoma xenograft metastases to the lungs in vivo (P < 0.001). Mechanistically, aspirin diminishes osteosarcoma migration, invasion, and metastasis through the NF-κB pathway. CONCLUSIONS Aspirin suppresses both the growth and metastasis of osteosarcoma through the NF-κB pathway at the cellular level and in the animal models.
Collapse
Affiliation(s)
- Dan Liao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Li Zhong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Tingmei Duan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Ru-Hua Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xin Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Gang Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Kaishun Hu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xiaobin Lv
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Tiebang Kang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
37
|
Zheng D, Zhu G, Liao S, Yi W, Luo G, He J, Pei Z, Li G, Zhou Y. Dysregulation of the PI3K/Akt signaling pathway affects cell cycle and apoptosis of side population cells in nasopharyngeal carcinoma. Oncol Lett 2015; 10:182-188. [PMID: 26170996 DOI: 10.3892/ol.2015.3218] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 04/08/2015] [Indexed: 01/15/2023] Open
Abstract
Increasing evidence has suggested that certain types of cancer possess their own stem-like cells, and that one subset of these cells, termed the side population (SP), may have an important role in tumorigenesis and cancer therapy. However, the molecular mechanisms underlying the modulation of SP cells in nasopharyngeal carcinoma (NPC) have remained elusive. In the present study, it was hypothesized that dysregulation of the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/Akt signaling pathway may influence SP and non-SP (NSP) phenotype. SP cells from the HK-1 NPC cell line were identified, and cancer stem cell markers were found to be highly expressed in SP cells compared with that of NSP cells. Freshly sorted SP cells demonstrated a significant increase in the proportion of cells in G0/G1 phase, while the majority of NSP cells were in the proliferative phase. Following 48 h of culture subsequent to cell sorting, the differences in cell cycle distribution between the SP and NSP cells converged. In addition, the apoptotic ratio of NSP cells was higher than that of SP cells at 24 h following sorting, but had no significant differences 48 h following sorting. To elucidate the potential mechanism mediating the cell cycle and apoptosis in SP cells, the expression levels of key molecules in the PI3K/Akt signaling pathway were evaluated. PI3K and Akt were upregulated, while 14-3-3σ protein was downregulated in SP cells when freshly sorted (0 h). However, there was no significant difference in the expression of these molecules between SP and NSP cells following 48 h of culture. These results suggested that dysregulation of the PI3K/Akt signaling pathway may be associated with the cell cycle and apoptosis of SP cells in NPC. However, further investigation is required to elucidate the detailed mechanisms underlying these effects.
Collapse
Affiliation(s)
- Danwei Zheng
- Medical Experimental Center, Hunan Provincial Tumor Hospital and The Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, P.R. China ; Molecular Genetics Laboratory, Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Changsha, Hunan 410078, P.R. China ; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Guangchao Zhu
- Medical Experimental Center, Hunan Provincial Tumor Hospital and The Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, P.R. China ; Molecular Genetics Laboratory, Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Changsha, Hunan 410078, P.R. China ; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Shan Liao
- Medical Experimental Center, Hunan Provincial Tumor Hospital and The Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, P.R. China ; Molecular Genetics Laboratory, Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Changsha, Hunan 410078, P.R. China ; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Wei Yi
- Medical Experimental Center, Hunan Provincial Tumor Hospital and The Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, P.R. China ; Molecular Genetics Laboratory, Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Changsha, Hunan 410078, P.R. China ; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Gengqiu Luo
- Department of Pathology, Basic School of Medicine, Central South University, Changsha, Hunan 410008, P.R. China
| | - Junyu He
- Medical Experimental Center, Hunan Provincial Tumor Hospital and The Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, P.R. China ; Molecular Genetics Laboratory, Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Changsha, Hunan 410078, P.R. China ; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Zhen Pei
- Medical Experimental Center, Hunan Provincial Tumor Hospital and The Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, P.R. China ; Molecular Genetics Laboratory, Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Changsha, Hunan 410078, P.R. China ; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Guiyuan Li
- Medical Experimental Center, Hunan Provincial Tumor Hospital and The Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, P.R. China ; Molecular Genetics Laboratory, Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Changsha, Hunan 410078, P.R. China ; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Yanhong Zhou
- Medical Experimental Center, Hunan Provincial Tumor Hospital and The Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, P.R. China ; Molecular Genetics Laboratory, Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Changsha, Hunan 410078, P.R. China ; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
38
|
Liang Y, Feng Q, Hong J, Feng F, Sang Y, Hu W, Xu M, Peng R, Kang T, Bei J, Zeng Y. Tumor growth and metastasis can be inhibited by maintaining genomic stability in cancer cells. Front Med 2015; 9:57-62. [DOI: 10.1007/s11684-015-0389-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Accepted: 12/26/2014] [Indexed: 11/25/2022]
|
39
|
Hu K, Liao D, Wu W, Han AJ, Shi HJ, Wang F, Wang X, Zhong L, Duan T, Wu Y, Cao J, Tang J, Sang Y, Wang L, Lv X, Xu S, Zhang RH, Deng WG, Li SP, Zeng YX, Kang T. Targeting the anaphase-promoting complex/cyclosome (APC/C)- bromodomain containing 7 (BRD7) pathway for human osteosarcoma. Oncotarget 2015; 5:3088-100. [PMID: 24840027 PMCID: PMC4102794 DOI: 10.18632/oncotarget.1816] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Osteosarcoma is the most common primary malignant bone tumor in childhood and adolescence and has a propensity for local invasion and early lung metastasis. However, the current therapies often result in chemoresistance, and a therapeutic target is not available in the clinic for osteosarcoma. Here, we report that BRD7 forms a complex with the anaphase-promoting complex/cyclosome (APC/C) and is degraded by APC/Ccdh1 and APC/Ccdc20 during the cell cycle. Moreover, BRD7 is a tumor suppressor in osteosarcoma, and the BRD7 mutant resistant to degradation by APC/C is more efficient than the wild-type protein at suppressing proliferation, colony formation, and tumor growth of osteosarcoma in vitro and in vivo. The combination of proTAME, an inhibitor of APC/C, with chemotherapeutic drugs efficiently targets osteosarcoma in vitro. Furthermore, there is a strong inverse correlation of protein levels between BRD7 and Cdh1 or Cdc20, and lower BRD7 expression is an indicator for poor prognosis in patients with osteosarcoma. Collectively, our results indicate that targeting the APC/C-BRD7 pathway may be a novel strategy for treating osteosarcoma.
Collapse
Affiliation(s)
- Kaishun Hu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Lun SWM, Cheung ST, Lo KW. Cancer stem-like cells in Epstein-Barr virus-associated nasopharyngeal carcinoma. CHINESE JOURNAL OF CANCER 2014; 33:529-38. [PMID: 25223912 PMCID: PMC4244315 DOI: 10.5732/cjc.014.10081] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Although the Epstein-Barr virus (EBV) has spread to all populations in the world, EBV-associated nasopharyngeal carcinoma (NPC) is prevalent only in South China and Southeast Asia. The role of EBV in the malignant transformation of nasopharyngeal epithelium is the main focus of current researches. Radiotherapy and chemoradiotherapy have been successful in treating early stage NPC, but the recurrence rates remain high. Unfortunately, local relapse and metastasis are commonly unresponsive to conventional treatments. These recurrent and metastatic lesions are believed to arise from residual or surviving cells that have the properties of cancer stem cells. These cancer stem-like cells (CSCs) have the ability to self-renew, differentiate, and sustain propagation. They are also chemo-resistant and can form spheres in anchorage-independent environments. This review summarizes recent researches on the CSCs in EBV-associated NPC, including the findings regarding cell surface markers, stem cell-related transcription factors, and various signaling pathways. In particular, the review focuses on the roles of EBV latent genes [latent membrane protein 1 (LMP1) and latent membrane protein 2A (LMP2A)], cellular microRNAs, and adenosine triphosphate (ATP)-binding cassette chemodrug transporters in contributing to the properties of CSCs, including the epithelial-mesenchymal transition, stem-like transition, and chemo-resistance. Novel therapeutics that enhance the efficacy of radiotherapy and chemoradiotherapy and inhibitors that suppress the properties of CSCs are also discussed.
Collapse
Affiliation(s)
- Samantha Wei-Man Lun
- Department of Anatomical and Cellular Pathology, State Key Laboratory in Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR.
| | | | | |
Collapse
|
41
|
Liao WT, Ye YP, Deng YJ, Bian XW, Ding YQ. Metastatic cancer stem cells: from the concept to therapeutics. AMERICAN JOURNAL OF STEM CELLS 2014; 3:46-62. [PMID: 25232505 PMCID: PMC4163604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 08/07/2014] [Indexed: 06/03/2023]
Abstract
Metastatic cancer stem cells (MCSCs) refer to a subpopulation of cancer cells with both stem cell properties and invasion capabilities that contribute to cancer metastasis. MCSCs have capability of self-renewal, potentials of multiple differentiation and development and/or reconstruction of cancer tissues. As compared with stationary cancer stem cells, MCSCs are capable of invasion to normal tissues such as vasculatures, resistance to chemo- and/or radio-therapies, escape from immune surveillance, survival in circulation and formation of metastasis. MCSCs are derived from invasive cancer stem cells (iCSCs) due to the plasticity of cancer stem cells, which is one of the characteristics of cancer cell heterogeneity. Both stages of iCSCs and MSCSs are the potential therapeutic targets for cancer metastasis in the future strategies of personalized cancer therapy.
Collapse
Affiliation(s)
- Wen-Ting Liao
- Department of Pathology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhou 510515, China
| | - Ya-Ping Ye
- Department of Pathology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhou 510515, China
| | - Yong-Jian Deng
- Department of Pathology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhou 510515, China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical UniversityChongqing 400038, China
| | - Yan-Qing Ding
- Department of Pathology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhou 510515, China
| |
Collapse
|
42
|
Zhang L, Xia Y, Li L, Wang Y, Liu Y, Li C, Yu T. Cancer stem cell-like cells exist in mucoepidermoid carcinoma cell line MC3. Oncol Res 2014; 20:589-600. [PMID: 24139417 DOI: 10.3727/096504013x13775486749452] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Strong evidence for the presence of cancer stem cells (CSCs) in tumors exists. CSCs play an important role in the development, invasion, and drug resistance of carcinoma. Poorly differentiated mucoepidermoid carcinoma (MEC) is a lethal malignancy of human salivary gland tumors. However, whether there are CSCs in MEC and their phenotypes remains unclear. We isolated side population (SP) and sphere-forming cells from the MEC cell line MC3 and identified their characteristics. The results showed that sphere-forming assays could enrich stem cell-like cells, with this group of cells exhibiting high cloning efficiency, possessing strong tumorigenic ability, and highly expressing Oct4 based on PCR and immunocytochemistry assays. They also highly expressed CD44 and lowly expressed CD24 according to PCR, immunocytochemistry assays, and fluorescence-activated cell sorting analysis. Higher cloning efficiency was observed in the SP cells, but PCR revealed that the SP and non-SP cells did not statistically differ in their expression of ABCG2, Oct4, CD44, and CD24. In spite of these, the findings were not conclusive on whether SP cells are stem cell-like cells. In conclusion, CSC-like cells do exist in the MC3 cell line, and sphere-forming assays could enrich them, sphere-forming and SP cells are not the same kind of cell subpopulations, and the characteristics of SP cells need to be further investigated.
Collapse
Affiliation(s)
- Louqiang Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
43
|
Hu WR, Lian YF, Peng LX, Lei JJ, Deng CC, Xu M, Feng QS, Chen LZ, Bei JX, Zeng YX. Monoacylglycerol lipase promotes metastases in nasopharyngeal carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:3704-3713. [PMID: 25120746 PMCID: PMC4128981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 05/21/2014] [Indexed: 06/03/2023]
Abstract
Monoacylglycerol lipase (MAGL) is a serine hydrolase that hydrolyzes monoacylglycerides into free fatty acids and glycerol. It has recently been found to be involved in cancer progression through the free fatty acid or endocannabinoid network after studies on its function in the endocannabinoid system. Here, we determined a role for MAGL in nasopharyngeal carcinoma (NPC), which is known for its high metastatic potential. Among the different NPC cells we tested, MAGL was highly expressed in high metastatic NPC cells, whereas low metastatic potential NPC cells exhibited lower expression of MAGL. Overexpression of MAGL in low metastatic NPC cells enhanced their motile behavior and metastatic capacity in vivo. Conversely, knockdown of MAGL reduced the motility of highly metastatic cells, reducing their metastatic capacity in vivo. Growth rate was not influenced by MAGL in either high or low metastatic cells. MAGL expression was associated with the epithelial-mesenchymal transition (EMT) proteins, such as E-cadherin, vimentin and Snail. It was also related to the sidepopulation (SP) of NPC cells. Our findings establish that MAGL promotes metastases in NPC through EMT, and it may serve as a target for the prevention of NPC metastases.
Collapse
Affiliation(s)
- Wen-Rong Hu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer CenterGuangzhou, China
- Department of Experimental Research, Sun Yat-Sen University Cancer CenterGuangzhou, China
| | - Yi-Fan Lian
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer CenterGuangzhou, China
- Department of Experimental Research, Sun Yat-Sen University Cancer CenterGuangzhou, China
| | - Li-Xia Peng
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer CenterGuangzhou, China
- Department of Experimental Research, Sun Yat-Sen University Cancer CenterGuangzhou, China
| | - Jin-Ju Lei
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer CenterGuangzhou, China
- Department of Experimental Research, Sun Yat-Sen University Cancer CenterGuangzhou, China
| | - Cheng-Cheng Deng
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer CenterGuangzhou, China
- Department of Experimental Research, Sun Yat-Sen University Cancer CenterGuangzhou, China
| | - Miao Xu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer CenterGuangzhou, China
- Department of Experimental Research, Sun Yat-Sen University Cancer CenterGuangzhou, China
| | - Qi-Sheng Feng
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer CenterGuangzhou, China
- Department of Experimental Research, Sun Yat-Sen University Cancer CenterGuangzhou, China
| | - Li-Zhen Chen
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer CenterGuangzhou, China
- Department of Experimental Research, Sun Yat-Sen University Cancer CenterGuangzhou, China
| | - Jin-Xin Bei
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer CenterGuangzhou, China
- Department of Experimental Research, Sun Yat-Sen University Cancer CenterGuangzhou, China
| | - Yi-Xin Zeng
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer CenterGuangzhou, China
- Department of Experimental Research, Sun Yat-Sen University Cancer CenterGuangzhou, China
| |
Collapse
|
44
|
González-Herrero I, Romero-Camarero I, Cañueto J, Cardeñoso-Álvarez E, Fernández-López E, Pérez-Losada J, Sánchez-García I, Román-Curto C. CD133+ cell content correlates with tumour growth in melanomas from skin with chronic sun-induced damage. Br J Dermatol 2014; 169:830-7. [PMID: 23662851 DOI: 10.1111/bjd.12428] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2013] [Indexed: 01/05/2023]
Abstract
BACKGROUND Melanoma is responsible for almost 80% of the deaths attributed to skin cancer. Stem cells, defined by CD133 expression, have been implicated in melanoma tumour growth, but their specific role is still uncertain. OBJECTIVES We hypothesized that the phenotypic heterogeneity of human cutaneous melanomas is related to their content of CD133+ cells. METHODS We compared the percentages of CD133+ cells in 29 tumours from four classic types of melanoma: lentigo maligna melanoma (LMM), superficial spreading melanoma, nodular melanoma and acral lentiginous melanoma (ALM). Also, we compared the percentages of CD133+ cells in melanomas with different degrees of exposure to ultraviolet radiation: 16 melanomas from skin with chronic sun-induced damage and 13 melanomas from skin without such damage. RESULTS We found a statistically significant increase of CD133+ cells in three different contexts: in melanomas arising on skin with signs of chronic sun-induced damage vs. nonexposed skin, in melanomas in situ vs. invasive melanomas, and in LMM vs. ALM. The proportions of CD133+ cells did not differ among samples of normal skin with different degrees of sun exposure. A distinct subpopulation of CD133+CXCR4+ cancer stem cells (CSCs) was identified and shown to be related to the invasive phenotype of the tumours. CONCLUSIONS Here, we provide evidence showing, for the first time, that an increase in the CD133+ cell content is associated both with melanomas arising on skin with signs of chronic sun-induced damage and in melanomas in situ with better prognosis. Moreover, our study further confirms the existence of a subpopulation of CD133+CXCR4+ CSCs in cutaneous melanomas with invasive phenotype and poor prognosis.
Collapse
Affiliation(s)
- I González-Herrero
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, CSIC/Universidad de Salamanca, Campus M. de Unamuno s/n, 37007, Salamanca, Spain; Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Zhang H, Chang WJ, Li XY, Zhang N, Kong JJ, Wang YF. Liver cancer stem cells are selectively enriched by low-dose cisplatin. ACTA ACUST UNITED AC 2014; 47:478-82. [PMID: 24770568 PMCID: PMC4086174 DOI: 10.1590/1414-431x20143415] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Accepted: 02/20/2014] [Indexed: 12/14/2022]
Abstract
Accumulating evidence has indicated the importance of cancer stem cells in
carcinogenesis. The goal of the present study was to determine the effect of low-dose
cisplatin on enriched liver cancer stem cells (LCSCs). Human hepatoblastoma HepG2
cells were treated with concentrations of cisplatin ranging from 1 to 5 μg/mL. Cell
survival and proliferation were evaluated using a tetrazolium dye (MTT) assay. LCSCs
were identified using specific markers, namely aldehyde dehydrogenase-1 (ALDH1) and
CD133. The percentage of ALDH1+ or CD133+ cells was examined by flow cytometric
analysis. The expression of ALDH1 and/or CD133 in HepG2 cells was determined by
immunocytochemical analysis. Low-dose cisplatin treatment significantly decreased
cell survival in HepG2 cells after 24 or 72 h. However, the percentage of LCSCs in
the surviving cells was greatly increased. The percentage of ALDH1+ or CD133+ cells
was increased in a time- and dose-dependent manner after treatment with 1-4 μg/mL
cisplatin, whereas 5 μg/mL cisplatin exposure slightly reduced the number of positive
cells. These findings indicate that low-dose cisplatin treatment may efficiently
enrich the LCSC population in HepG2 cells.
Collapse
Affiliation(s)
- H Zhang
- Department of Internal Medicine, First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - W J Chang
- Department of Internal Medicine, First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - X Y Li
- Department of Internal Medicine, First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - N Zhang
- Department of Internal Medicine, First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - J J Kong
- Department of Internal Medicine, First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Y F Wang
- Department of Internal Medicine, First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
46
|
Rastogi V, Yadav P, Bhattacharya SS, Mishra AK, Verma N, Verma A, Pandit JK. Carbon nanotubes: an emerging drug carrier for targeting cancer cells. JOURNAL OF DRUG DELIVERY 2014; 2014:670815. [PMID: 24872894 PMCID: PMC4020363 DOI: 10.1155/2014/670815] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 01/26/2014] [Accepted: 03/12/2014] [Indexed: 12/18/2022]
Abstract
During recent years carbon nanotubes (CNTs) have been attracted by many researchers as a drug delivery carrier. CNTs are the third allotropic form of carbon-fullerenes which were rolled into cylindrical tubes. To be integrated into the biological systems, CNTs can be chemically modified or functionalised with therapeutically active molecules by forming stable covalent bonds or supramolecular assemblies based on noncovalent interactions. Owing to their high carrying capacity, biocompatibility, and specificity to cells, various cancer cells have been explored with CNTs for evaluation of pharmacokinetic parameters, cell viability, cytotoxicty, and drug delivery in tumor cells. This review attempts to highlight all aspects of CNTs which render them as an effective anticancer drug carrier and imaging agent. Also the potential application of CNT in targeting metastatic cancer cells by entrapping biomolecules and anticancer drugs has been covered in this review.
Collapse
Affiliation(s)
- Vaibhav Rastogi
- School of Pharmaceutical Sciences, IFTM University, Moradabad, Uttar Pradesh 244001, India
| | - Pragya Yadav
- School of Pharmaceutical Sciences, IFTM University, Moradabad, Uttar Pradesh 244001, India
| | | | - Arun Kumar Mishra
- School of Pharmaceutical Sciences, IFTM University, Moradabad, Uttar Pradesh 244001, India
| | - Navneet Verma
- School of Pharmaceutical Sciences, IFTM University, Moradabad, Uttar Pradesh 244001, India
| | - Anurag Verma
- School of Pharmaceutical Sciences, IFTM University, Moradabad, Uttar Pradesh 244001, India
| | - Jayanta Kumar Pandit
- Department of Pharmaceutics, Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| |
Collapse
|
47
|
Bao YN, Cao X, Luo DH, Sun R, Peng LX, Wang L, Yan YP, Zheng LS, Xie P, Cao Y, Liang YY, Zheng FJ, Huang BJ, Xiang YQ, Lv X, Chen QY, Chen MY, Huang PY, Guo L, Mai HQ, Guo X, Zeng YX, Qian CN. Urokinase-type plasminogen activator receptor signaling is critical in nasopharyngeal carcinoma cell growth and metastasis. Cell Cycle 2014; 13:1958-69. [PMID: 24763226 DOI: 10.4161/cc.28921] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is one of the most common malignancies in southern China and Southeast Asia, with the highest metastasis rate among head and neck cancers. The mechanisms underlying NPC progression remain poorly understood. Genome-wide expression profiling on 18 NPC vs. 18 noncancerous nasopharyngeal tissues together with GeneGo pathway analysis and expression verification in NPC cells and tissues revealed a potential role of urokinase-type plasminogen activator receptor (uPAR) in NPC progression, which has not been investigated in NPC. We then observed that uPAR expression is increased in poorly differentiated, highly metastatic NPC cells compared with lowly metastatic cells or differentiated NPC cells. In vitro studies demonstrated that uPAR regulates NPC cell growth, colony formation, migration, and invasion and promotes the epithelial-mesenchymal transition (EMT). Additional tumor xenograft and spontaneous metastasis experiments revealed that uPAR promotes NPC cell growth and metastasis in vivo. The JAK-STAT pathway is involved in uPAR-regulated signaling in NPC cells as determined by immunoblotting. Moreover, uPAR-mediated growth and motility is partially abolished upon treatment with the Jak1/Jak2 inhibitor INCB018424. We suppressed uPA expression in uPAR-overexpressing NPC cells and found that uPAR-mediated cellular growth and motility is not exclusively dependent on uPA. In summary, uPAR is a significant regulator of NPC progression and could serve as a promising therapeutic target.
Collapse
Affiliation(s)
- Ying-Na Bao
- State Key Laboratory of Oncology in South China; Sun Yat-sen University Cancer Center; Guangzhou, China; Department of Radiotherapy; Affiliated Hospital of Inner Mongolia Medical University; Hohhot City, Inner Mongolia Autonomous Region, China
| | - Xue Cao
- State Key Laboratory of Oncology in South China; Sun Yat-sen University Cancer Center; Guangzhou, China
| | - Dong-Hua Luo
- Department of Nasopharyngeal Carcinoma; Sun Yat-sen University Cancer Center; Guangzhou, China
| | - Rui Sun
- Department of Nasopharyngeal Carcinoma; Sun Yat-sen University Cancer Center; Guangzhou, China
| | - Li-Xia Peng
- State Key Laboratory of Oncology in South China; Sun Yat-sen University Cancer Center; Guangzhou, China
| | - Lin Wang
- Department of Nasopharyngeal Carcinoma; Sun Yat-sen University Cancer Center; Guangzhou, China
| | | | - Li-Sheng Zheng
- State Key Laboratory of Oncology in South China; Sun Yat-sen University Cancer Center; Guangzhou, China
| | - Ping Xie
- State Key Laboratory of Oncology in South China; Sun Yat-sen University Cancer Center; Guangzhou, China
| | - Yun Cao
- State Key Laboratory of Oncology in South China; Sun Yat-sen University Cancer Center; Guangzhou, China
| | - Ying-Ying Liang
- State Key Laboratory of Oncology in South China; Sun Yat-sen University Cancer Center; Guangzhou, China
| | - Fang-Jing Zheng
- State Key Laboratory of Oncology in South China; Sun Yat-sen University Cancer Center; Guangzhou, China
| | - Bi-Jun Huang
- State Key Laboratory of Oncology in South China; Sun Yat-sen University Cancer Center; Guangzhou, China
| | - Yan-Qun Xiang
- Department of Nasopharyngeal Carcinoma; Sun Yat-sen University Cancer Center; Guangzhou, China
| | - Xing Lv
- Department of Nasopharyngeal Carcinoma; Sun Yat-sen University Cancer Center; Guangzhou, China
| | - Qiu-Yan Chen
- Department of Nasopharyngeal Carcinoma; Sun Yat-sen University Cancer Center; Guangzhou, China
| | - Ming-Yuan Chen
- Department of Nasopharyngeal Carcinoma; Sun Yat-sen University Cancer Center; Guangzhou, China
| | - Pei-Yu Huang
- Department of Nasopharyngeal Carcinoma; Sun Yat-sen University Cancer Center; Guangzhou, China
| | - Ling Guo
- Department of Nasopharyngeal Carcinoma; Sun Yat-sen University Cancer Center; Guangzhou, China
| | - Hai-Qiang Mai
- Department of Nasopharyngeal Carcinoma; Sun Yat-sen University Cancer Center; Guangzhou, China
| | - Xiang Guo
- Department of Nasopharyngeal Carcinoma; Sun Yat-sen University Cancer Center; Guangzhou, China
| | - Yi-Xin Zeng
- State Key Laboratory of Oncology in South China; Sun Yat-sen University Cancer Center; Guangzhou, China
| | - Chao-Nan Qian
- State Key Laboratory of Oncology in South China; Sun Yat-sen University Cancer Center; Guangzhou, China; Department of Nasopharyngeal Carcinoma; Sun Yat-sen University Cancer Center; Guangzhou, China
| |
Collapse
|
48
|
Meta-analysis of the global gene expression profile of triple-negative breast cancer identifies genes for the prognostication and treatment of aggressive breast cancer. Oncogenesis 2014; 3:e100. [PMID: 24752235 PMCID: PMC4007196 DOI: 10.1038/oncsis.2014.14] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 03/10/2014] [Indexed: 02/07/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype lacking expression of estrogen and progesterone receptors (ER/PR) and HER2, thus limiting therapy options. We hypothesized that meta-analysis of TNBC gene expression profiles would illuminate mechanisms underlying the aggressive nature of this disease and identify therapeutic targets. Meta-analysis in the Oncomine database identified 206 genes that were recurrently deregulated in TNBC compared with non-TNBC and in tumors that metastasized or led to death within 5 years. This ‘aggressiveness gene list' was enriched for two core functions/metagenes: chromosomal instability (CIN) and ER signaling metagenes. We calculated an ‘aggressiveness score' as the ratio of the CIN metagene to the ER metagene, which identified aggressive tumors in breast cancer data sets regardless of subtype or other clinico-pathological indicators. A score calculated from six genes from the CIN metagene and two genes from the ER metagene recapitulated the aggressiveness score. By multivariate survival analysis, we show that our aggressiveness scores (from 206 genes or the 8 representative genes) outperformed several published prognostic signatures. Small interfering RNA screen revealed that the CIN metagene holds therapeutic targets against TNBC. Particularly, the inhibition of TTK significantly reduced the survival of TNBC cells and synergized with docetaxel in vitro. Importantly, mitosis-independent expression of TTK protein was associated with aggressive subgroups, poor survival and further stratified outcome within grade 3, lymph node-positive, HER2-positive and TNBC patients. In conclusion, we identified the core components of CIN and ER metagenes that identify aggressive breast tumors and have therapeutic potential in TNBC and aggressive breast tumors. Prognostication from these metagenes at the mRNA level was limited to ER-positive tumors. However, we provide evidence that mitosis-independent expression of TTK protein was prognostic in TNBC and other aggressive breast cancer subgroups, suggesting that protection of CIN/aneuploidy drives aggressiveness and treatment resistance.
Collapse
|
49
|
Dynamics between cancer cell subpopulations reveals a model coordinating with both hierarchical and stochastic concepts. PLoS One 2014; 9:e84654. [PMID: 24416258 PMCID: PMC3886990 DOI: 10.1371/journal.pone.0084654] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Accepted: 11/18/2013] [Indexed: 02/04/2023] Open
Abstract
Tumors are often heterogeneous in which tumor cells of different phenotypes have distinct properties. For scientific and clinical interests, it is of fundamental importance to understand their properties and the dynamic variations among different phenotypes, specifically under radio- and/or chemo-therapy. Currently there are two controversial models describing tumor heterogeneity, the cancer stem cell (CSC) model and the stochastic model. To clarify the controversy, we measured probabilities of different division types and transitions of cells via in situ immunofluorescence. Based on the experiment data, we constructed a model that combines the CSC with the stochastic concepts, showing the existence of both distinctive CSC subpopulations and the stochastic transitions from NSCCs to CSCs. The results showed that the dynamic variations between CSCs and non-stem cancer cells (NSCCs) can be simulated with the model. Further studies also showed that the model can be used to describe the dynamics of the two subpopulations after radiation treatment. More importantly, analysis demonstrated that the experimental detectable equilibrium CSC proportion can be achieved only when the stochastic transitions from NSCCs to CSCs occur, indicating that tumor heterogeneity may exist in a model coordinating with both the CSC and the stochastic concepts. The mathematic model based on experimental parameters may contribute to a better understanding of the tumor heterogeneity, and provide references on the dynamics of CSC subpopulation during radiotherapy.
Collapse
|
50
|
Tian XH, Hou WJ, Fang Y, Fan J, Tong H, Bai SL, Chen Q, Xu H, Li Y. XAV939, a tankyrase 1 inhibitior, promotes cell apoptosis in neuroblastoma cell lines by inhibiting Wnt/β-catenin signaling pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2013; 32:100. [PMID: 24308762 PMCID: PMC3866601 DOI: 10.1186/1756-9966-32-100] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Accepted: 11/04/2013] [Indexed: 12/17/2022]
Abstract
Background Neuroblastoma (NB) is the most common extracranial solid tumor in childhood. The present treatment including surgery, chemotherapy and radiation, which have only 40% long-term cure rates, and usually cause tumor recurrence. Thus, looking for new effective and less toxic therapies has important significance. XAV939 is a small molecule inhibitor of tankyrase 1(TNKS1). The objective of this study is to investigate the effect of XAV939 on the proliferation and apoptosis of NB cell lines, and the related mechanism. Methods In the present study, we used both XAV939 treatment and RNAi method to demonstrate that TNKS1 inhibition may be a potential mechanism to cure NB. MTT method was used for determining the cell viability and the appropriate concerntration for follow-up assays. The colony formation assay, Annexin V staining and cell cycle analysis were used for detecting colony forming ability, cell apoptosis and the percentage of different cell cycle. The Western blot was used for detecting the expression of key proteins of Wnt/ beta-catenin (Wnt/β-catenin) signaling pathway. Results The results showed that TNKS1 inhibition decreased the viability of SH-SY5Y, SK-N-SH and IMR-32 cells, induced apoptosis in SH-SY5Y as well as SK-N-SH cells, and led to the accumulation of NB cells in the S and G2/M phase of the cell cycle. Moreover, we demonstrated TNKS1 inhibition may in part blocked Wnt/β-catenin signaling and reduced the expression of anti-apoptosis protein. Finally, we also demonstrated that TNKS1 inhibition decreased colony formation in vitro. Conclusions These findings suggested that TNKS1 may be a potential molecule target for the treatment of NB.
Collapse
Affiliation(s)
| | | | | | | | | | - Shu-Ling Bai
- Department of Tissue Engineering, College of Basic Medical Sciences, China Medical University, Shenyang 110001, PR China.
| | | | | | | |
Collapse
|