1
|
Aquino IG, Cuadra-Zelaya FJM, Bizeli ALV, Palma PVB, Mariano FV, Salo T, Coletta RD, Bastos DC, Graner E. Isolation and phenotypic characterization of cancer stem cells from metastatic oral cancer cells. Oral Dis 2024; 30:4886-4897. [PMID: 38764396 DOI: 10.1111/odi.15003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/13/2023] [Revised: 04/12/2024] [Accepted: 05/03/2024] [Indexed: 05/21/2024]
Abstract
OBJECTIVES To isolate cancer stem cells (CSC) from a metastatic oral squamous cell carcinoma (OSCC) cell line and investigate their in vitro and in vivo phenotypic characteristics. MATERIALS AND METHODS Subpopulations with individual staining intensities for CD44 and CD326 were isolated from the OSCC cell line LN-1A by FACS: CD44Low/CD326- (CSC-M1), CD44Low/CD326High (CSC-E), and CD44High/CD326- (CSC-M2). Proliferation, clonogenic potential, adhesion, migration, epithelial-mesenchymal transition markers, and sensitivity to cisplatin and TVB-3166 were analyzed in vitro. Tumor formation and metastasis were assessed by subcutaneous and orthotopic inoculations into BALB/c mice. RESULTS E-cadherin levels were higher in CSC-E cells while vimentin and Slug more produced by CSC-M2 cells. CSC-M1 and CSC-M2 subpopulations showed higher proliferation, produced more colonies, and have stronger adhesion to the extracellular matrix. All cell lines established tumors; however, CSC-E and CSC-M2 formed larger masses and produced more metastases. CONCLUSION The CSC subpopulations here described show increased cancer capabilities in vitro, tumorigenic and metastatic potential in vivo, and may be exploited in the search for novel therapeutic targets for OSCC.
Collapse
Affiliation(s)
- Iara Gonçalves Aquino
- Departamento de Diagnóstico Oral, Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
| | | | - Ana Laura Valença Bizeli
- Departamento de Diagnóstico Oral, Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
| | | | - Fernanda Viviane Mariano
- Departamento de Diagnóstico Oral, Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
- Departamento de Patologia, Faculdade de Ciências Médicas, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Tuula Salo
- Cancer and Translational Medicine Research Unit, Faculty of Medicine and Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
- Department of Oral and Maxillofacial Diseases, Helsinki University Central Hospital, and Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland
- HUSLAB, Department of Pathology, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
| | - Ricardo Della Coletta
- Departamento de Diagnóstico Oral, Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
- Programa de Pós-Graduação Em Biologia Buco-Dental, Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
| | - Débora Campanella Bastos
- Programa de Pós-Graduação Em Biologia Buco-Dental, Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
- Faculdade de Medicina São Leopoldo Mandic, Campinas, São Paulo, Brazil
| | - Edgard Graner
- Departamento de Diagnóstico Oral, Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
| |
Collapse
|
2
|
Wada H, Otsuka R, Germeraad WTV, Murata T, Kondo T, Seino KI. Tumor cell-induced macrophage senescence plays a pivotal role in tumor initiation followed by stable growth in immunocompetent condition. J Immunother Cancer 2023; 11:e006677. [PMID: 37963635 PMCID: PMC10649871 DOI: 10.1136/jitc-2023-006677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 10/06/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND The cancer stem cell theory proposes that tumor formation in vivo is driven only by specific tumor-initiating cells having stemness; however, clinical trials conducted to test drugs that target the tumor stemness provided unsatisfactory results thus far. Recent studies showed clear involvement of immunity in tumors; however, the requirements of tumor-initiation followed by stable growth in immunocompetent individuals remain largely unknown. METHODS To clarify this, we used two similarly induced glioblastoma lines, 8B and 9G. They were both established by overexpression of an oncogenic H-RasL61 in p53-deficient neural stem cells. In immunocompromised animals in an orthotopic transplantation model using 1000 cells, both show tumor-forming potential. On the other hand, although in immunocompetent animals, 8B shows similar tumor-forming potential but that of 9G's are very poor. This suggests that 8B cells are tumor-initiating cells in immunocompetent animals. Therefore, we hypothesized that the differences in the interaction properties of 8B and 9G with immune cells could be used to identify the factors responsible for its tumor forming potential in immunocompetent animals and performed analysis. RESULTS Different from 9G, 8B cells induced senescence-like state of macrophages around tumors. We investigated the senescence-inducing factor of macrophages by 8B cells and found that it was interleukin 6. Such senescence-like macrophages produced Arginase-1, an immunosuppressive molecule known to contribute to T-cell hyporesponsiveness. The senescence-like macrophages highly expressed CD38, a nicotinamide adenine dinucleotide (NAD) glycohydrolase associated with NAD shortage in senescent cells. The addition of nicotinamide mononucleotide (NMN), an NAD precursor, in vitro inhibited to the induction of macrophage senescence-like phenotype and inhibited Arginase-1 expression resulting in retaining T-cell function. Moreover, exogenous in vivo administration of NMN after tumor inoculation inhibited tumor-initiation followed by stable growth in the immunocompetent mouse tumor model. CONCLUSIONS We identified one of the requirements for tumor-initiating cells in immunocompetent animals. In addition, we have shown that tumor growth can be inhibited by externally administered NMN against macrophage senescence-like state that occurs in the very early stages of tumor-initiating cell development. This therapy targeting the immunosuppressive environment formed by macrophage senescence-like state is expected to be a novel promising cancer therapeutic strategy.
Collapse
Affiliation(s)
- Haruka Wada
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Ryo Otsuka
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Wilfred T V Germeraad
- GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, Limburg, The Netherlands
- Department of Internal Medicine, Division of Hematology, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Tomoki Murata
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Toru Kondo
- Division of Stem Cell Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Ken-Ichiro Seino
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
3
|
Kuo YC, Kou HW, Hsu CP, Lo CH, Hwang TL. Identification and Clinical Significance of Pancreatic Cancer Stem Cells and Their Chemotherapeutic Drug Resistance. Int J Mol Sci 2023; 24:ijms24087331. [PMID: 37108495 PMCID: PMC10138402 DOI: 10.3390/ijms24087331] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/17/2023] [Revised: 04/07/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Pancreatic cancer ranks in the 10th-11th position among cancers affecting men in Taiwan, besides being a rather difficult-to-treat disease. The overall 5-year survival rate of pancreatic cancer is only 5-10%, while that of resectable pancreatic cancer is still approximately 15-20%. Cancer stem cells possess intrinsic detoxifying mechanisms that allow them to survive against conventional therapy by developing multidrug resistance. This study was conducted to investigate how to overcome chemoresistance and its mechanisms in pancreatic cancer stem cells (CSCs) using gemcitabine-resistant pancreatic cancer cell lines. Pancreatic CSCs were identified from human pancreatic cancer lines. To determine whether CSCs possess a chemoresistant phenotype, the sensitivity of unselected tumor cells, sorted CSCs, and tumor spheroid cells to fluorouracil (5-FU), gemcitabine (GEM), and cisplatin was analyzed under stem cell conditions or differentiating conditions. Although the mechanisms underlying multidrug resistance in CSCs are poorly understood, ABC transporters such as ABCG2, ABCB1, and ABCC1 are believed to be responsible. Therefore, we measured the mRNA expression levels of ABCG2, ABCB1, and ABCC1 by real-time RT-PCR. Our results showed that no significant differences were found in the effects of different concentrations of gemcitabine on CSCs CD44+/EpCAM+ of various PDAC cell line cultures (BxPC-3, Capan-1, and PANC-1). There was also no difference between CSCs and non-CSCs. Gemcitabine-resistant cells exhibited distinct morphological changes, including a spindle-shaped morphology, the appearance of pseudopodia, and reduced adhesion characteristics of transformed fibroblasts. These cells were found to be more invasive and migratory, and showed increased vimentin expression and decreased E-cadherin expression. Immunofluorescence and immunoblotting experiments demonstrated increased nuclear localization of total β-catenin. These alterations are hallmarks of epithelial-to-mesenchymal transition (EMT). Resistant cells showed activation of the receptor protein tyrosine kinase c-Met and increased expression of the stem cell marker cluster of differentiation (CD) 24, CD44, and epithelial specific antigen (ESA). We concluded that the expression of the ABCG2 transporter protein was significantly higher in CD44+ and EpCAM+ CSCs of PDAC cell lines. Cancer stem-like cells exhibited chemoresistance. Gemcitabine-resistant pancreatic tumor cells were associated with EMT, a more aggressive and invasive phenotype of numerous solid tumors. Increased phosphorylation of c-Met may also be related to chemoresistance, and EMT and could be used as an attractive adjunctive chemotherapeutic target in pancreatic cancer.
Collapse
Affiliation(s)
- Yu-Chi Kuo
- Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Lin-Kou, Taoyuan 333, Taiwan
| | - Hao-Wei Kou
- Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Lin-Kou, Taoyuan 333, Taiwan
| | - Chih-Po Hsu
- Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Lin-Kou, Taoyuan 333, Taiwan
| | - Chih-Hong Lo
- Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Lin-Kou, Taoyuan 333, Taiwan
| | - Tsann-Long Hwang
- Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Lin-Kou, Taoyuan 333, Taiwan
| |
Collapse
|
4
|
Predicting Prognosis and Platinum Resistance in Ovarian Cancer: Role of Immunohistochemistry Biomarkers. Int J Mol Sci 2023; 24:ijms24031973. [PMID: 36768291 PMCID: PMC9916805 DOI: 10.3390/ijms24031973] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/11/2022] [Revised: 12/17/2022] [Accepted: 12/20/2022] [Indexed: 01/20/2023] Open
Abstract
Ovarian cancer is a lethal reproductive tumour affecting women worldwide. The advancement in presentation and occurrence of chemoresistance are the key factors for poor survival among ovarian cancer women. Surgical debulking was the mainstay of systemic treatment for ovarian cancer, which was followed by a successful start to platinum-based chemotherapy. However, most women develop platinum resistance and relapse within six months of receiving first-line treatment. Thus, there is a great need to identify biomarkers to predict platinum resistance before enrolment into chemotherapy, which would facilitate individualized targeted therapy for these subgroups of patients to ensure better survival and an improved quality of life and overall outcome. Harnessing the immune response through immunotherapy approaches has changed the treatment way for patients with cancer. The immune outline has emerged as a beneficial tool for recognizing predictive and prognostic biomarkers clinically. Studying the tumour microenvironment (TME) of ovarian cancer tissue may provide awareness of actionable targets for enhancing chemotherapy outcomes and quality of life. This review analyses the relevance of immunohistochemistry biomarkers as prognostic biomarkers in predicting chemotherapy resistance and improving the quality of life in ovarian cancer.
Collapse
|
5
|
Down-Regulation of lncRNA MBNL1-AS1 Promotes Tumor Stem Cell-like Characteristics and Prostate Cancer Progression through miR-221-3p/CDKN1B/C-myc Axis. Cancers (Basel) 2022; 14:cancers14235783. [PMID: 36497267 PMCID: PMC9739743 DOI: 10.3390/cancers14235783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/20/2022] [Revised: 11/19/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022] Open
Abstract
The recurrence, progression, and drug resistance of prostate cancer (PC) is closely related to the cancer stem cells (CSCs). Therefore, it is necessary to find the key regulators of prostate cancer stem cells (PCSCs). Here, we analyzed the results of a single-class logistic regression machine learning algorithm (OCLR) to identify the PCSC-associated lncRNA MBNL1-AS1. The effects of MBNL1-AS1 on the stemness of CSCs was assessed using qPCR, western blot and sphere-forming assays. The role of MBNL1-AS1 in mediating the proliferation and invasion of the PC cell lines was examined using Transwell, wounding-healing, CCK-8, EdU and animal assays. Dual-luciferase and ChIRP assays were used to examine the molecular mechanism of MBNL1-AS1 in PCSCs. MBNL1-AS1 was shown to be negatively correlated with stemness index (mRNAsi), and even prognosis, tumor progression, recurrence, and drug resistance in PC patients. The knockdown of MBNL1-AS1 significantly affected the stemness of the PC cells, and subsequently their invasive and proliferative abilities. Molecular mechanism studies suggested that MBNL1-AS1 regulates CDKN1B through competitive binding to miR-221-3p, which led to the inhibition of the Wnt signaling pathway to affect PCSCs. In conclusion, our study identified MBNL1-AS1 as a key regulator of PCSCs and examined its mechanism of action in the malignant progression of PC.
Collapse
|
6
|
Polyethylenimine, an Autophagy-Inducing Platinum-Carbene-Based Drug Carrier with Potent Toxicity towards Glioblastoma Cancer Stem Cells. Cancers (Basel) 2022; 14:cancers14205057. [PMID: 36291841 PMCID: PMC9599868 DOI: 10.3390/cancers14205057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/23/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022] Open
Abstract
The difficulty involved in the treatment of many tumours due to their recurrence and resistance to chemotherapy is tightly linked to the presence of cancer stem cells (CSCs). This CSC sub-population is distinct from the majority of cancer cells of the tumour bulk. Indeed, CSCs have increased mitochondrial mass that has been linked to increased sensitivity to mitochondrial targeting compounds. Thus, a platinum-based polyethylenimine (PEI) polymer-drug conjugate (PDC) was assessed as a potential anti-CSC therapeutic since it has previously displayed mitochondrial accumulation. Our results show that CSCs have increased specific sensitivity to the PEI carrier and to the PDC. The mechanism of cell death seems to be necrotic in nature, with an absence of apoptotic markers. Cell death is accompanied by the induction of a protective autophagy. The interference in the balance of this pathway, which is highly important for CSCs, may be responsible for a partial reversion of the stem-like phenotype observed with prolonged PEI and PDC treatment. Several markers also indicate the cell death mode to be capable of inducing an anti-cancer immune response. This study thus indicates the potential therapeutic perspectives of polycations against CSCs.
Collapse
|
7
|
Wang LL, Tang X, Zhou G, Liu S, Wang Y, Chen F, Li T, Wen F, Liu S, Mai H. PROM1 and CTGF Expression in Childhood MLL-Rearrangement Acute Lymphoblastic Leukemia. JOURNAL OF ONCOLOGY 2022; 2022:5896022. [PMID: 36276286 PMCID: PMC9586771 DOI: 10.1155/2022/5896022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 03/24/2022] [Revised: 08/28/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022]
Abstract
The prognosis of over 90% of infant acute lymphoblastic leukemia (ALL) remains poor because of harboring the mixed-lineage leukemia gene (MLL) fusion. To give insight into the critical coexpressed genes related to the MLL-rearrangement (MLL-R) gene in childhood acute lymphoblastic leukemia, we integrated different bioinformatic methods. First, the gene expression data of MLL-R ALL and normal samples from GSE13159 and GSE13164 were analyzed using "compare" function in the Oncomine database. The top 150 overexpressed and 150 underexpressed genes were identified by the Oncomine website. Then, we employed the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) to define functional genes for the 300 DEGs. The Cytoscape identified two important networks for overexpressed genes, including 35 functional genes, among which PROM1, FLT3, CTGF, LGALS1, IGFBP7, ZNRF1, and RUNX2 were considered as the key genes because of their high expression in MLL-R ALL compared to the expression in other subclassification of leukemia in the MILE dataset. Further analysis of GSE68720, GSE19475, and Therapeutically Applicable Research to Generate Effective Treatments (TARGET) ALL (phase I) database confirmed the robust expression of 7 key genes in MLL-R compared to MLL-germline (MLL-G) childhood ALL. Kaplan-Meier analysis indicated that childhood ALL patients with high PROM1 and CTGF expression had significantly poor overall survival. These findings suggest that PROM1 and CTGF represent two potential therapeutic targets for childhood MLL-R ALL.
Collapse
Affiliation(s)
- Lu-lu Wang
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen 518038, China
| | - Xue Tang
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen 518038, China
| | - Guichi Zhou
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen 518038, China
| | - Shilin Liu
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen 518038, China
| | - Ying Wang
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen 518038, China
| | - Fen Chen
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen 518038, China
| | - Tonghui Li
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen 518038, China
| | - Feiqiu Wen
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen 518038, China
| | - Sixi Liu
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen 518038, China
| | - Huirong Mai
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen 518038, China
| |
Collapse
|
8
|
Hefni AM, Sayed AM, Hussien MT, Abdalla AZ, Gabr AG. CD133 is an independent predictive and prognostic marker in metastatic breast cancer. Cancer Biomark 2022; 35:207-215. [PMID: 36120770 DOI: 10.3233/cbm-210539] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND CD133 is a transmembrane glycoprotein and is considered the most common cell surface marker to identify cancer stem cells in hematological and solid tumors, including breast cancer. OBJECTIVES To evaluate the impact of immunohistochemical expression of CD133 on response rate and survival in metastatic breast cancer, as well as to correlate it with various demographics and clinicopathological characteristics. METHODS One-hundred metastatic breast cancer patients were prospectively recruited at the Medical Oncology Department at South Egypt Cancer Institute during the period from January 2018 to January 2020. RESULTS There was a statistically significant correlation between CD133 positive patients with various adverse clinicopathological parameters such as high grade (p= 0.013), higher tumor (p= 0.001), and nodal staging (p= 0.024) during a median follow-up time of 17 months. In addition, Cases with CD133 positive expression had a significantly lower survival time than those with negative expression (3-years OS 37.4% versus 85.5%, p= 0.024). Regarding the response rate, CD133 positive patients had a lower response rate than negative patients (50% versus 54%, p= 0.012). CONCLUSIONS Positive CD133 is correlated with poor prognosis in metastatic breast cancer patients.
Collapse
Affiliation(s)
- Ahmed Mubarak Hefni
- Medical Oncology, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Ayat Mohammed Sayed
- Medical Oncology, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Marwa T Hussien
- Oncologic Pathology, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | | | - Adel Gomaa Gabr
- Medical Oncology, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| |
Collapse
|
9
|
Wei Y, Chen Q, Huang S, Liu Y, Li Y, Xing Y, Shi D, Xu W, Liu W, Ji Z, Wu B, Chen X, Jiang J. The Interaction between DNMT1 and High-Mannose CD133 Maintains the Slow-Cycling State and Tumorigenic Potential of Glioma Stem Cell. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202216. [PMID: 35798319 PMCID: PMC9475542 DOI: 10.1002/advs.202202216] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 04/16/2022] [Indexed: 05/24/2023]
Abstract
The quiescent/slow-cycling state preserves the self-renewal capacity of cancer stem cells (CSCs) and leads to the therapy resistance of CSCs. The mechanisms maintaining CSCs quiescence remain largely unknown. Here, it is demonstrated that lower expression of MAN1A1 in glioma stem cell (GSC) resulted in the formation of high-mannose type N-glycan on CD133. Furthermore, the high-mannose type N-glycan of CD133 is necessary for its interaction with DNMT1. Activation of p21 and p27 by the CD133-DNMT1 interaction maintains the slow-cycling state of GSC, and promotes chemotherapy resistance and tumorigenesis of GSCs. Elimination of the CD133-DNMT1 interaction by a cell-penetrating peptide or MAN1A1 overexpression inhibits the tumorigenesis of GSCs and increases the sensitivity of GSCs to temozolomide. Analysis of glioma samples reveals that the levels of high-mannose type N-glycan are correlated with glioma recurrence. Collectively, the high mannose CD133-DNMT1 interaction maintains the slow-cycling state and tumorigenic potential of GSC, providing a potential strategy to eliminate quiescent GSCs.
Collapse
Affiliation(s)
- Yuanyan Wei
- NHC Key Laboratory of Glycoconjuates ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200032P. R. China
| | - Qihang Chen
- NHC Key Laboratory of Glycoconjuates ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200032P. R. China
| | - Sijing Huang
- NHC Key Laboratory of Glycoconjuates ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200032P. R. China
| | - Yingchao Liu
- Department of NeurosurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandong250021P. R. China
| | - Yinan Li
- NHC Key Laboratory of Glycoconjuates ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200032P. R. China
| | - Yang Xing
- NHC Key Laboratory of Glycoconjuates ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200032P. R. China
| | - Danfang Shi
- NHC Key Laboratory of Glycoconjuates ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200032P. R. China
| | - Wenlong Xu
- Division of NeurosurgeryZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Weitao Liu
- NHC Key Laboratory of Glycoconjuates ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200032P. R. China
| | - Zhi Ji
- NHC Key Laboratory of Glycoconjuates ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200032P. R. China
| | - Bingrui Wu
- NHC Key Laboratory of Glycoconjuates ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200032P. R. China
| | - Xiaoning Chen
- NHC Key Laboratory of Glycoconjuates ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200032P. R. China
| | - Jianhai Jiang
- NHC Key Laboratory of Glycoconjuates ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200032P. R. China
| |
Collapse
|
10
|
Glamoclija U, Mahmutovic L, Bilajac E, Soljic V, Vukojevic K, Suljagic M. Metformin and Thymoquinone Synergistically Inhibit Proliferation of Imatinib-Resistant Human Leukemic Cells. Front Pharmacol 2022; 13:867133. [PMID: 35496297 PMCID: PMC9043685 DOI: 10.3389/fphar.2022.867133] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/31/2022] [Accepted: 03/14/2022] [Indexed: 12/12/2022] Open
Abstract
Chemotherapy resistance is one of the major challenges in cancer treatment, including leukemia. A massive array of research is evaluating combinations of drugs directed against different intracellular signaling molecules to overcome cancer resistance, increase therapy effectiveness, and decrease its adverse effects. Combining chemicals with proven safety profiles, such as drugs already used in therapy and active substances isolated from natural sources, could potentially have superior effects compared to monotherapies. In this study, we evaluated the effects of metformin and thymoquinone (TQ) as monotherapy and combinatorial treatments in chronic myeloid leukemia (CML) cell lines sensitive and resistant to imatinib therapy. The effects were also evaluated in primary monocytic acute myeloid leukemia (AML) and chronic lymphocytic leukemia (CLL) cells. Both compounds induced a dose- and time-dependent decrease of viability and proliferation in tested cells. Metformin had similar IC50 values in imatinib-sensitive and imatinib-resistant cell lines. IC50 values of TQ were significantly higher in imatinib-resistant cells, but with a limited resistance index (2.4). Synergistic effects of combinatorial treatments were observed in all tested cell lines, as well as in primary cells. The strongest synergistic effects were observed in the inhibition of imatinib-resistant cell line proliferation. Metformin and TQ inhibited the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling and induced apoptosis in tested cell lines and primary cells. The enhanced effects of combinatorial treatments on the induction of apoptosis were more dominant in imatinib-resistant compared to imatinib-sensitive CML cells. Primary cells were more sensitive to combinatorial treatments compared to cell lines. A combination of 1.25 mM metformin and 0.625 µM TQ increased the levels of cleaved poly (ADP-ribose) polymerase (PARP), decreased the levels of proliferation regulatory proteins, and inhibited protein kinase B (Akt) and NF-κB signaling in primary CLL cells. This study demonstrates that combinatorial treatments of imatinib-resistant malignant clones with metformin and TQ by complementary intracellular multi-targeting represents a promising approach in future studies.
Collapse
Affiliation(s)
- Una Glamoclija
- Department of Biochemistry and Clinical Analysis, University of Sarajevo-Faculty of Pharmacy, Sarajevo, Bosnia and Herzegovina
- Department of Histology and Embryology, School of Medicine, University of Mostar, Mostar, Bosnia and Herzegovina
- Scientific Research Unit, Bosnalijek JSC, Sarajevo, Bosnia and Herzegovina
| | - Lejla Mahmutovic
- Genetics and Bioengineering Department, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Esma Bilajac
- Genetics and Bioengineering Department, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Violeta Soljic
- Department of Histology and Embryology, School of Medicine, University of Mostar, Mostar, Bosnia and Herzegovina
- Faculty of Health Studies, University of Mostar, Mostar, Bosnia and Herzegovina
| | - Katarina Vukojevic
- Department of Histology and Embryology, School of Medicine, University of Mostar, Mostar, Bosnia and Herzegovina
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Split, Croatia
| | - Mirza Suljagic
- 3D BioLabs, FabLab Bosnia and Herzegovina, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
- *Correspondence: Mirza Suljagic,
| |
Collapse
|
11
|
Store-Operated Calcium Entry and Its Implications in Cancer Stem Cells. Cells 2022; 11:cells11081332. [PMID: 35456011 PMCID: PMC9032688 DOI: 10.3390/cells11081332] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/02/2022] [Revised: 04/04/2022] [Accepted: 04/12/2022] [Indexed: 12/25/2022] Open
Abstract
Tumors are composed by a heterogeneous population of cells. Among them, a sub-population of cells, termed cancer stem cells, exhibit stemness features, such as self-renewal capabilities, disposition to differentiate to a more proliferative state, and chemotherapy resistance, processes that are all mediated by Ca2+. Ca2+ homeostasis is vital for several physiological processes, and alterations in the patterns of expressions of the proteins and molecules that modulate it have recently become a cancer hallmark. Store-operated Ca2+ entry is a major mechanism for Ca2+ entry from the extracellular medium in non-excitable cells that leads to increases in the cytosolic Ca2+ concentration required for several processes, including cancer stem cell properties. Here, we focus on the participation of STIM, Orai, and TRPC proteins, the store-operated Ca2+ entry key components, in cancer stem cell biology and tumorigenesis.
Collapse
|
12
|
The Natural Compound Dehydrocrenatidine Attenuates Nicotine-Induced Stemness and Epithelial-Mesenchymal Transition in Hepatocellular Carcinoma by Regulating a7nAChR-Jak2 Signaling Pathways. DISEASE MARKERS 2022; 2022:8316335. [PMID: 35111269 PMCID: PMC8803439 DOI: 10.1155/2022/8316335] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 10/31/2021] [Accepted: 12/16/2021] [Indexed: 12/23/2022]
Abstract
Background Exposure to nicotine has been observed associated with tumor progression, metastasis, and therapy resistance of many cancers. Hepatocellular carcinoma (HCC) is one major cancer related to the liver and the most difficult to treat malignancies worldwide. The underlying mechanism of nicotine in the stimulation of HCC tumorigenesis is still not studied well. Methods Classically, nicotine binds to nicotinic acetylcholine receptors (nAChRs) and induces many downstream cancer-associated signaling pathways. Big data analysis is used to explore the importance of a7nAChR-Jak2 axis in the progression of hepatocellular carcinoma. Bioinformatic analysis was performed to determine gene associated with a7nAChR-Jak2 axis of HCC patients. Biological importance of a7nAChR-Jak2 axis was investigated in vitro (Hun7 and HepG2 cell lines), and athymic nude mouse models bearing HepG2-HCC cells xenografts were established in vivo. Result We found that nicotine exposure stimulated the HCC tumorigenicity by inducing the expression of one of the key nAChRs subunit that is α7nAChR as well as the expression of Janus kinase (JAK)-2. In both the in vitro and in vivo studies, the reduced overexpression of α7nAChR and increased sensitization of HCC towards treatment is observed with dehydrocrenatidine (DHCT), a novel and potent JAK family kinase inhibitor. Interestingly, DHCT treatment results in the reduction of the epithelial-mesenchymal transition process which leads to a significant reduction of clonogenicity, migratory, and invasive ability of HCC cells. Moreover, DHCT treatment also inhibits the cancer stem cell phenotype by inhibiting the tumor-sphere formation and reducing the number of ALDH1+ cells population in nicotine-stimulated HCC cells. Conclusions Taken together, the presented results indicate the positive effect of inhibition of nicotine induced overexpression of α7nAChR and JAK2, unique to HCC. Thus, these findings suggest the nicotine effect on HCC progression via α7nAChR-mediated JAK2 signaling pathways, and DHCT treatment enhances the therapeutic potential of HCC patients via overcoming/reversing the effect of nicotine in HCC patients.
Collapse
|
13
|
Ganbat D, Jugder BE, Ganbat L, Tomoeda M, Dungubat E, Takahashi Y, Mori I, Shiomi T, Tomita Y. The Efficacy of Vitamin K, A Member Of Naphthoquinones in the Treatment of Cancer: A Systematic Review and Meta-Analysis. Curr Cancer Drug Targets 2021; 21:495-513. [PMID: 33475062 DOI: 10.2174/1568009621999210120182834] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/05/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Redox dysregulation originating from metabolic alterations in cancer cells contributes to their proliferation, invasion, and resistance to therapy. Conversely, these features represent a specific vulnerability of malignant cells that can be selectively targeted by redox chemotherapeutics. Amongst them, Vitamin K (VitK) carries the potential against cancer stem cells, in addition to the rest of tumor mass. OBJECTIVES To assess the possible benefits and safety of VitK for cancer treatment using a systematic review and meta-analysis with a mixed-methods approach. METHODS We performed a systematic search on several electronic databases for studies comparing VitK treatment with and without combination to the control groups. For quantitative studies, fully or partially reported clinical outcomes such as recurrence rates, survival, overall response and adverse reactions were assessed. For qualitative studies, a narrative synthesis was accomplished. RESULTS Our analysis suggested that the clinical outcome of efficacy, the pooled hazard ratio for progression-free survival, and the pooled relative risk for overall survival, and overall response were significantly higher in the VitK therapy group compared to the placebo group (p<0.05). We did not observe any significant difference in the occurrence of adverse events between groups. Among qualitative studies, VitK treatment targeting myelodysplastic syndrome and advanced solid tumors resulted in 24.1% and 10% of clinical response, respectively. CONCLUSION VitK not only exerts antitumor effects against a wide range of tumor types, but it also has excellent synergism with other therapeutic agents.
Collapse
Affiliation(s)
- Dariimaa Ganbat
- Department of Pathology, School of Medicine, International University of Health and Welfare, Narita, Japan
| | - Bat-Erdene Jugder
- Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, Boston Children's Hospital, United States
| | - Lkhamaa Ganbat
- Department of Administration, MCS Property, Ulan-Bator, Mongolia
| | - Miki Tomoeda
- Department of Rehabilitation, Kobe International University, Kobe, Japan
| | - Erdenetsogt Dungubat
- Department of Pathology, School of Medicine, International University of Health and Welfare, Narita, Japan
| | - Yoshihisa Takahashi
- Department of Pathology, School of Medicine, International University of Health and Welfare, Narita, Japan
| | - Ichiro Mori
- Department of Pathology, School of Medicine, International University of Health and Welfare, Narita, Japan
| | - Takayuki Shiomi
- Department of Pathology, School of Medicine, International University of Health and Welfare, Narita, Japan
| | - Yasuhiko Tomita
- Department of Pathology, School of Medicine, International University of Health and Welfare, Narita, Japan
| |
Collapse
|
14
|
Ngo MHT, Peng SW, Kuo YC, Lin CY, Wu MH, Chuang CH, Kao CX, Jeng HY, Lin GW, Ling TY, Chang TS, Huang YH. A Yes-Associated Protein (YAP) and Insulin-Like Growth Factor 1 Receptor (IGF-1R) Signaling Loop Is Involved in Sorafenib Resistance in Hepatocellular Carcinoma. Cancers (Basel) 2021; 13:3812. [PMID: 34359714 PMCID: PMC8345119 DOI: 10.3390/cancers13153812] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/31/2021] [Revised: 07/12/2021] [Accepted: 07/22/2021] [Indexed: 12/24/2022] Open
Abstract
The role of a YAP-IGF-1R signaling loop in HCC resistance to sorafenib remains unknown. METHOD Sorafenib-resistant cells were generated by treating naïve cells (HepG2215 and Hep3B) with sorafenib. Different cancer cell lines from databases were analyzed through the ONCOMINE web server. BIOSTORM-LIHC patient tissues (46 nonresponders and 21 responders to sorafenib) were used to compare YAP mRNA levels. The HepG2215_R-derived xenograft in SCID mice was used as an in vivo model. HCC tissues from a patient with sorafenib failure were used to examine differences in YAP and IGF-R signaling. RESULTS Positive associations exist among the levels of YAP, IGF-1R, and EMT markers in HCC tissues and the levels of these proteins increased with sorafenib failure, with a trend of tumor-margin distribution in vivo. Blocking YAP downregulated IGF-1R signaling-related proteins, while IGF-1/2 treatment enhanced the nuclear translocation of YAP in HCC cells through PI3K-mTOR regulation. The combination of YAP-specific inhibitor verteporfin (VP) and sorafenib effectively decreased cell viability in a synergistic manner, evidenced by the combination index (CI). CONCLUSION A YAP-IGF-1R signaling loop may play a role in HCC sorafenib resistance and could provide novel potential targets for combination therapy with sorafenib to overcome drug resistance in HCC.
Collapse
Affiliation(s)
- Mai-Huong T. Ngo
- International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (M.-H.T.N.); (C.-X.K.)
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (S.-W.P.); (G.-W.L.)
| | - Sue-Wei Peng
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (S.-W.P.); (G.-W.L.)
- TMU Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-C.K.); (H.-Y.J.)
| | - Yung-Che Kuo
- TMU Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-C.K.); (H.-Y.J.)
| | - Chun-Yen Lin
- Institute of Information Science, Academia Sinica, Taipei 11529, Taiwan; (C.-Y.L.); (C.-H.C.)
| | - Ming-Heng Wu
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Chia-Hsien Chuang
- Institute of Information Science, Academia Sinica, Taipei 11529, Taiwan; (C.-Y.L.); (C.-H.C.)
| | - Cheng-Xiang Kao
- International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (M.-H.T.N.); (C.-X.K.)
| | - Han-Yin Jeng
- TMU Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-C.K.); (H.-Y.J.)
| | - Gee-Way Lin
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (S.-W.P.); (G.-W.L.)
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Thai-Yen Ling
- Department and Graduate Institute of Pharmacology, National Taiwan University, Taipei 100, Taiwan;
| | - Te-Sheng Chang
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33382, Taiwan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
| | - Yen-Hua Huang
- International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (M.-H.T.N.); (C.-X.K.)
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (S.-W.P.); (G.-W.L.)
- TMU Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-C.K.); (H.-Y.J.)
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Comprehensive Cancer Center of Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
15
|
Induced Mitochondrial Alteration and DNA Damage via IFNGR-JAK2-STAT1-PARP1 Pathway Facilitates Viral Hepatitis Associated Hepatocellular Carcinoma Aggressiveness and Stemness. Cancers (Basel) 2021; 13:cancers13112755. [PMID: 34199353 PMCID: PMC8199505 DOI: 10.3390/cancers13112755] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/18/2021] [Revised: 05/17/2021] [Accepted: 05/28/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Hepatitis virus is a major risk factor for liver cancer. We analyzed possible synergism between momelotinib and sorafenib in hepatitis virus-associated liver cancer. The combined effect of momelotinib and sorafenib both at in vitro and in vivo synergistically sup-presses the proliferation of vHCC cells and effectively reduces the tumor burden. Our results showed that momelotinib effectively suppressed the expression of the IFNGR-JAK-STAT-PARP1 pathway, which results in the downregulation of cancer stem cell genes and enhances the antitumor efficacy of sorafenib by initiating the expression of apoptosis-related genes and inhibiting the DNA repair gene in vHCC cells, thus maximizing its therapeutic potential for patients with HCC. Abstract Background: Hepatitis virus is a major risk factor for liver cancer. The mitochondrial dysfunction IFN gamma-related pathways are activated after virus infection. Jak family-related protein is involved in the downstream of IFN gamma-related pathways. However, the effect of the IFNGR-JAK-STAT pathway acting as functional regulators of their related protein expression on virus infection and hepatocellular carcinoma (HCC) remains unclear. Interestingly, the role of the DNA repair gene (PARP1) in therapy resistant cancers also has not been studied and explored well. In this study, we hypothesized that momelotinib could suppress the progression of HCC by targeting Jak family related and PARP1 DNA repair protein. Based on this observation, we link the relevant targets of the JAK family and the potential applications of targeted therapy inhibitors. Methods: We analyzed possible synergism between momelotinib and sorafenib in hepatitis virus-associated liver cancer. Immunostaining, colony formation assay, cell invasion, migration, and tumorsphere-formation assay were used for drug cytotoxicity, cell viability, and possible molecular mechanism. Result: We first demonstrated that the expression of Jak1 and 2 is significantly upregulated in vHCC than in nvHCC/normal liver tissues. In addition, the gene expression of IFN gamma-related pathways is activated after virus infection. Additionally, we found that momelotinib significantly inhibited the growth of HCC cells and reduces the expression of Jak2, which showed the importance of momelotinib in targeting Jak2 and reducing tumorigenesis in HCC. Meanwhile, momelotinib effectively inhibited the IFNGR-JAK-STAT pathway and reduced the migratory/invasive ability of vHCC cells through down-regulating EMT biomarkers (E-cadherin and vimentin), transcription factor (Slug), and significantly inhibits the DNA damage repair enzyme PARP1. It also induced cell apoptosis of vHCC cells. Furthermore, the combined effect of momelotinib and sorafenib both at in vitro and in vivo synergistically suppresses the proliferation of vHCC cells and effectively reduces the tumor burden. Conclusions: Our results showed that momelotinib effectively suppressed the expression of the IFNGR-JAK-STAT-PARP1 pathway, which results in the downregulation of cancer stem cell genes and enhances the antitumor efficacy of sorafenib by initiating the expression of apoptosis-related genes and inhibiting the DNA repair gene in vHCC cells, thus maximizing its therapeutic potential for patients with HCC.
Collapse
|
16
|
Wang Z, Snyder M, Kenison JE, Yang K, Lara B, Lydell E, Bennani K, Novikov O, Federico A, Monti S, Sherr DH. How the AHR Became Important in Cancer: The Role of Chronically Active AHR in Cancer Aggression. Int J Mol Sci 2020; 22:ijms22010387. [PMID: 33396563 PMCID: PMC7795223 DOI: 10.3390/ijms22010387] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/08/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 12/13/2022] Open
Abstract
For decades, the aryl hydrocarbon receptor (AHR) was studied for its role in environmental chemical toxicity i.e., as a quirk of nature and a mediator of unintended consequences of human pollution. During that period, it was not certain that the AHR had a “normal” physiological function. However, the ongoing accumulation of data from an ever-expanding variety of studies on cancer, cancer immunity, autoimmunity, organ development, and other areas bears witness to a staggering array of AHR-controlled normal and pathological activities. The objective of this review is to discuss how the AHR has gone from a likely contributor to genotoxic environmental carcinogen-induced cancer to a master regulator of malignant cell progression and cancer aggression. Particular focus is placed on the association between AHR activity and poor cancer outcomes, feedback loops that control chronic AHR activity in cancer, and the role of chronically active AHR in driving cancer cell invasion, migration, cancer stem cell characteristics, and survival.
Collapse
Affiliation(s)
- Zhongyan Wang
- Department of Environmental Health, Boston University School of Public Health, Boston, MA 02118, USA; (Z.W.); (K.Y.); (E.L.)
| | - Megan Snyder
- Graduate Program in Genetics and Genomics, Division of Graduate Medical Sciences, Boston University School of Medicine, Boston, MA 02118, USA;
| | - Jessica E. Kenison
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA;
| | - Kangkang Yang
- Department of Environmental Health, Boston University School of Public Health, Boston, MA 02118, USA; (Z.W.); (K.Y.); (E.L.)
| | - Brian Lara
- Department of Environmental Health, Boston University, Boston, MA 02118, USA; (B.L.); (K.B.)
| | - Emily Lydell
- Department of Environmental Health, Boston University School of Public Health, Boston, MA 02118, USA; (Z.W.); (K.Y.); (E.L.)
| | - Kawtar Bennani
- Department of Environmental Health, Boston University, Boston, MA 02118, USA; (B.L.); (K.B.)
| | | | - Anthony Federico
- Division of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA; (A.F.); (S.M.)
| | - Stefano Monti
- Division of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA; (A.F.); (S.M.)
| | - David H. Sherr
- Department of Environmental Health, Boston University School of Public Health, Boston, MA 02118, USA; (Z.W.); (K.Y.); (E.L.)
- Correspondence: ; Tel.: +1-617-358-1707
| |
Collapse
|
17
|
Rossi F, Noren H, Jove R, Beljanski V, Grinnemo KH. Differences and similarities between cancer and somatic stem cells: therapeutic implications. Stem Cell Res Ther 2020; 11:489. [PMID: 33208173 PMCID: PMC7672862 DOI: 10.1186/s13287-020-02018-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/09/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023] Open
Abstract
Over the last decades, the cancer survival rate has increased due to personalized therapies, the discovery of targeted therapeutics and novel biological agents, and the application of palliative treatments. Despite these advances, tumor resistance to chemotherapy and radiation and rapid progression to metastatic disease are still seen in many patients. Evidence has shown that cancer stem cells (CSCs), a sub-population of cells that share many common characteristics with somatic stem cells (SSCs), contribute to this therapeutic failure. The most critical properties of CSCs are their self-renewal ability and their capacity for differentiation into heterogeneous populations of cancer cells. Although CSCs only constitute a low percentage of the total tumor mass, these cells can regrow the tumor mass on their own. Initially identified in leukemia, CSCs have subsequently been found in cancers of the breast, the colon, the pancreas, and the brain. Common genetic and phenotypic features found in both SSCs and CSCs, including upregulated signaling pathways such as Notch, Wnt, Hedgehog, and TGF-β. These pathways play fundamental roles in the development as well as in the control of cell survival and cell fate and are relevant to therapeutic targeting of CSCs. The differences in the expression of membrane proteins and exosome-delivered microRNAs between SSCs and CSCs are also important to specifically target the stem cells of the cancer. Further research efforts should be directed toward elucidation of the fundamental differences between SSCs and CSCs to improve existing therapies and generate new clinically relevant cancer treatments.
Collapse
Affiliation(s)
- Fiorella Rossi
- NSU Cell Therapy Institute, Nova Southeastern University, 3301 College Ave, 3200 South University Drive, Fort Lauderdale, FL, 33328, USA
| | - Hunter Noren
- NSU Cell Therapy Institute, Nova Southeastern University, 3301 College Ave, 3200 South University Drive, Fort Lauderdale, FL, 33328, USA
| | - Richard Jove
- NSU Cell Therapy Institute, Nova Southeastern University, 3301 College Ave, 3200 South University Drive, Fort Lauderdale, FL, 33328, USA
| | - Vladimir Beljanski
- NSU Cell Therapy Institute, Nova Southeastern University, 3301 College Ave, 3200 South University Drive, Fort Lauderdale, FL, 33328, USA.
| | - Karl-Henrik Grinnemo
- NSU Cell Therapy Institute, Nova Southeastern University, 3301 College Ave, 3200 South University Drive, Fort Lauderdale, FL, 33328, USA. .,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden. .,Department of Surgical Sciences, Division of Cardiothoracic Surgery and Anaesthesiology, Uppsala University, Akademiska University Hospital, Akademiska sjukhuset, ingång 50, 4 tr, 751 85, Uppsala, Sweden.
| |
Collapse
|
18
|
Loss of the Ste20-like kinase induces a basal/stem-like phenotype in HER2-positive breast cancers. Oncogene 2020; 39:4592-4602. [PMID: 32393835 DOI: 10.1038/s41388-020-1315-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/29/2019] [Revised: 04/22/2020] [Accepted: 04/24/2020] [Indexed: 01/20/2023]
Abstract
HER2 is overexpressed in 20-30% of all breast cancers and is associated with an invasive disease and poor clinical outcome. The Ste20-like kinase (SLK) is activated downstream of HER2/Neu and is required for efficient epithelial-to-mesenchymal transition, cell cycle progression, and migration in the mammary epithelium. Here we show that loss of SLK in a murine model of HER2/Neu-positive breast cancers significantly accelerates tumor onset and decreases overall survival. Transcriptional profiling of SLK knockout HER2/Neu-derived tumor cells revealed a strong induction in the triple-negative breast cancer marker, Sox10, accompanied by an increase in mammary stem/progenitor activity. Similarly, we demonstrate that SLK and Sox10 expression are inversely correlated in patient samples, with the loss of SLK and acquisition of Sox10 marking the triple-negative subtype. Furthermore, pharmacological inhibition of AKT reduces SLK-null tumor growth in vivo and is rescued by ectopic Sox10 expression, suggesting that Sox10 is a critical regulator of tumor growth downstream of SLK/AKT. These findings highlight a role for SLK in negatively regulating HER2-induced mammary tumorigenesis and provide mechanistic insight into the regulation of Sox10 expression in breast cancer.
Collapse
|
19
|
Mahmoodi S, Nezafat N, Negahdaripour M, Ghasemi Y. A New Approach for Cancer Immunotherapy Based on the Cancer Stem Cell Antigens Properties. Curr Mol Med 2020; 19:2-11. [PMID: 30714514 DOI: 10.2174/1566524019666190204114721] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/18/2019] [Revised: 01/24/2019] [Accepted: 02/11/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Cancer stem cells (CSCs) are a rare population of tumor cells, which play an important role in tumor initiation, progression, and maintenance. The concept that cancer cells arise from stem cells was presented about 150 years ago. Recently, this hypothesis was renewed considering the heterogeneity of tumor cells. CSCs are resistant to chemo- and radio-therapy. Therefore, targeting CSCs could be a novel and effective strategy to struggle with tumor cells. OBJECTIVE In this mini-review, we highlight that different immunotherapeutic approaches can be used to target cancer cells and eradicate different tumor cells. The most important targets are specific markers recognized on the CSC surface as CSC antigens such as CD44, CD133, Aldehyde Dehydrogenase (ALDH), and SOX family members. This article emphasizes recent advances in CSCs in cancer therapy. RESULTS Our results present that CSC antigens play an important role in tumor initiation, especially in the cells that are resistant to chemo- and radiotherapy agents. Therefore, they are ideal targets for cancer immunotherapy, for instance, in developing different types of cancer vaccines or antibodies against tumor cells. CONCLUSION The current studies related to cancer immunotherapy through targeting the CSC antigens based on their properties are briefly summarized. Altogether, CSC antigens can be efficiently targeted to treat cancer patients.
Collapse
Affiliation(s)
- Shirin Mahmoodi
- Department of Medical Biotechnology, Fasa University of Medical Sciences, Fasa, Iran
| | - Navid Nezafat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Manica Negahdaripour
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Younes Ghasemi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
20
|
Hu Z, Liu H, Zhang X, Hong B, Wu Z, Li Q, Zhou C. Promoter hypermethylation of CD133/PROM1 is an independent poor prognosis factor for head and neck squamous cell carcinoma. Medicine (Baltimore) 2020; 99:e19491. [PMID: 32176088 PMCID: PMC7440166 DOI: 10.1097/md.0000000000019491] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 12/23/2022] Open
Abstract
PROM1 has played a pivotal role in the identification and isolation of tumor stem cells. This study aimed to assess the association between PROM1 promoter methylation and head and neck squamous cell carcinoma (HNSCC), and its diagnostic and prognostic value.Bioinformatic analysis was performed using data from the Cancer Genome Atlas-HNSC and Gene Expression Omnibus datasets.The results showed that PROM1 promoter was hypermethylated in HNSCCs compared with normal head and neck tissues (P = 4.58E-37). The area under the receiver-operating characteristic curve based on methylated PROM1 data was 0.799. In addition, PROM1 hypermethylation independently predicted poor overall survival (hazard ratio [HR]: 1.459, 95% confidence interval [CI]: 1.071-1.987, P = .016) and recurrence-free survival (HR: 1.729, 95% CI: 1.088-2.749, P = .021) in HNSCC patients. Moreover, PROM1 methylation was weakly negatively correlated with its mRNA expression (Pearson r = -0.148, P < .001).In summary, our study reveals that methylated PROM1 might serve as a valuable diagnostic biomarker and predictor of poor survival for HNSCC patients. PROM1 hypermethylation might partially contribute to its downregulation in HNSCC.
Collapse
Affiliation(s)
- Zele Hu
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Zhenhai Longsai Hospital
| | - Huigao Liu
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Zhenhai Longsai Hospital
| | - Xinrong Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Zhenhai Longsai Hospital
| | - Bin Hong
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Zhenhai Longsai Hospital
| | - Zhenhua Wu
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Center, Lihuili Eastern Hospital
| | - Qun Li
- Department of Otorhinolaryngology Head and Neck Surgery
- Laboratory of Otorhinolaryngology Head and Neck Surgery
- Diagnosis and Treatment Center of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Chongchang Zhou
- Department of Otorhinolaryngology Head and Neck Surgery
- Laboratory of Otorhinolaryngology Head and Neck Surgery
- Diagnosis and Treatment Center of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
21
|
Alvarado-Ortiz E, Sarabia-Sánchez MÁ, García-Carrancá A. Molecular Mechanisms Underlying the Functions of Cellular Markers Associated with the Phenotype of Cancer Stem Cells. Curr Stem Cell Res Ther 2019; 14:405-420. [PMID: 30147013 DOI: 10.2174/1574888x13666180821154752] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/11/2018] [Revised: 07/18/2018] [Accepted: 08/13/2018] [Indexed: 12/19/2022]
Abstract
Cancer Stem Cells (CSC) generally constitute a minor cellular population within tumors that exhibits some capacities of normal Stem Cells (SC). The existence of CSC, able to self-renew and differentiate, influences central aspects of tumor biology, in part because they can continue tumor growth, give rise to metastasis, and acquire drug and radioresistance, which open new avenues for therapeutics. It is well known that SC constantly interacts with their niche, which includes mesenchymal cells, extracellular ligands, and the Extra Cellular Matrix (ECM). These interactions regularly lead to homeostasis and maintenance of SC characteristics. However, the exact participation of each of these components for CSC maintenance is not clear, as they appear to be context- or cell-specific. In the recent past, surface cellular markers have been fundamental molecular tools for identifying CSC and distinguishing them from other tumor cells. Importantly, some of these cellular markers have been shown to possess functional roles that affect central aspects of CSC. Likewise, some of these markers can participate in regulating the interaction of CSC with their niche, particularly the ECM. We focused this review on the molecular mechanisms of surface cellular markers commonly employed to identify CSC, highlighting the signaling pathways and mechanisms involved in CSC-ECM interactions, through each of the cellular markers commonly used in the study of CSC, such as CD44, CD133, CD49f, CD24, CXCR4, and LGR5. Their presence does not necessarily implicate them in CSC biology.
Collapse
Affiliation(s)
- Eduardo Alvarado-Ortiz
- Programa de Maestría y Doctorado en Ciencias Biológicas, Facultad de Ciencias, Universidad Nacional Autónoma de México, México City, México.,Laboratory of Virus and Cancer, Unidad de Investigacion Biomedica en Cáncer, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico & Subdireccion de Investigacion Basica, Instituto Nacional de Cancerologia, Secretaria de Salud, Ciudad de Mexico, Mexico
| | - Miguel Á Sarabia-Sánchez
- Laboratory of Virus and Cancer, Unidad de Investigacion Biomedica en Cáncer, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico & Subdireccion de Investigacion Basica, Instituto Nacional de Cancerologia, Secretaria de Salud, Ciudad de Mexico, Mexico.,Programa de Maestría y Doctorado en Ciencias Bioquímicas, Facultad de Química, Universidad Nacional Autónoma de México, , México City, México
| | - Alejandro García-Carrancá
- Laboratory of Virus and Cancer, Unidad de Investigacion Biomedica en Cáncer, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico & Subdireccion de Investigacion Basica, Instituto Nacional de Cancerologia, Secretaria de Salud, Ciudad de Mexico, Mexico
| |
Collapse
|
22
|
Saw PE, Song EW. Phage display screening of therapeutic peptide for cancer targeting and therapy. Protein Cell 2019; 10:787-807. [PMID: 31140150 PMCID: PMC6834755 DOI: 10.1007/s13238-019-0639-7] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/30/2018] [Accepted: 04/21/2019] [Indexed: 12/14/2022] Open
Abstract
Recently, phage display technology has been announced as the recipient of Nobel Prize in Chemistry 2018. Phage display technique allows high affinity target-binding peptides to be selected from a complex mixture pool of billions of displayed peptides on phage in a combinatorial library and could be further enriched through the biopanning process; proving to be a powerful technique in the screening of peptide with high affinity and selectivity. In this review, we will first discuss the modifications in phage display techniques used to isolate various cancer-specific ligands by in situ, in vitro, in vivo, and ex vivo screening methods. We will then discuss prominent examples of solid tumor targeting-peptides; namely peptide targeting tumor vasculature, tumor microenvironment (TME) and over-expressed receptors on cancer cells identified through phage display screening. We will also discuss the current challenges and future outlook for targeting peptide-based therapeutics in the clinics.
Collapse
Affiliation(s)
- Phei Er Saw
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Er-Wei Song
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
| |
Collapse
|
23
|
Antrodia cinnamomea Enhances Chemo-Sensitivity of 5-FU and Suppresses Colon Tumorigenesis and Cancer Stemness via Up-Regulation of Tumor Suppressor miR-142-3p. Biomolecules 2019; 9:biom9080306. [PMID: 31349708 PMCID: PMC6723279 DOI: 10.3390/biom9080306] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/05/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 12/20/2022] Open
Abstract
5-Fluorouracil (5-FU) regimen remains the backbone of the first-line agent to treat colon cancer, but often these patients develop resistance. Cancer stem cells (CSC's) are considered as one of the key contributors in the development of drug resistance and tumor recurrence. We aimed to provide preclinical evidence for Antrodia cinnamomea (AC), as a potential in suppressing colon cancer CSC's to overcome 5-FU drug-resistant. In-vitro assays including cell viability, colony formation, AC + 5-FU drug combination index and tumor sphere generation were applied to determine the inhibitory effect of AC. Mouse xenograft models also incorporated to evaluate in vivo effect of AC. AC treatment significantly inhibited the proliferation, colony formation and tumor sphere generation. AC also inhibited the expression of oncogenic markers (NF-κB, and C-myc), EMT/metastasis markers (vimentin and MMP3) and stemness associated markers (β-catenin, SOX-2 and Nanog). Sequential treatment of AC and 5-FU synergized and reduces colon cancer viability both in vivo and in vitro. Mechanistically, AC mediated anti-tumor effect was associated with an increased level of tumor suppressor microRNAs especially, miR142-3p. AC can be a potent synergistic adjuvant, down-regulates cancer stemness genes and enhances the antitumor ability of 5-FU by stimulating apoptosis-associated genes, suppressing inflammation and metastasis genes through miR142-3p in colon cancer.
Collapse
|
24
|
Shi L, Wu Z, Miao J, Du S, Ai S, Xu E, Feng M, Song J, Guan W. Adenosine interaction with adenosine receptor A2a promotes gastric cancer metastasis by enhancing PI3K-AKT-mTOR signaling. Mol Biol Cell 2019; 30:2527-2534. [PMID: 31339445 PMCID: PMC6743355 DOI: 10.1091/mbc.e19-03-0136] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/04/2023] Open
Abstract
The accumulation of adenosine in the tumor microenvironment is associated with tumor progression in many cancers. However, whether adenosine is involved in gastric cancer (GC) metastasis and progression, and the underlying molecular mechanism, is largely unclear. In this study, we find that GC tissues and cell lines had higher A2aR levels than nontumor gastric tissues and cell lines. A2aR expression correlated positively with TNMstage, and associated with poor outcomes. Adenosine enhanced the expression of the stemness and epithelial-mesenchymal transition-associated genes by binding to A2aR. A2aR expression on GC cells promoted metastasis in vivo. The PI3K-AKT-mTOR signaling pathway was involved in adenosine-stimulated GC cell migration and invasion. Our results indicate that adenosine promotes GC cell invasion and metastasis by interacting with A2aR to enhance PI3K-AKT-mTOR pathway signaling.
Collapse
Affiliation(s)
- Linsen Shi
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006, People's Republic of China.,The Affiliated Nanjing Drum Tower Clinical College of Nanjing Medical University, Nanjing 210002, People's Republic of China
| | - Zhaoying Wu
- Xuzhou Medical University, Xuzhou 221006, People's Republic of China
| | - Ji Miao
- Department of Gastrointestinal Surgery, The Affiliated Drum Tower Hospital of Nanjing University, Nanjing 210000, People's Republic of China
| | - Shangce Du
- The Affiliated Nanjing Drum Tower Clinical College of Nanjing Medical University, Nanjing 210002, People's Republic of China
| | - Shichao Ai
- Nanjing University, Nanjing 21000, People's Republic of China
| | - En Xu
- Nanjing University, Nanjing 21000, People's Republic of China
| | - Min Feng
- Department of Gastrointestinal Surgery, The Affiliated Drum Tower Hospital of Nanjing University, Nanjing 210000, People's Republic of China
| | - Jun Song
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006, People's Republic of China.,Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou 221006, People's Republic of China
| | - Wenxian Guan
- The Affiliated Nanjing Drum Tower Clinical College of Nanjing Medical University, Nanjing 210002, People's Republic of China.,Department of Gastrointestinal Surgery, The Affiliated Drum Tower Hospital of Nanjing University, Nanjing 210000, People's Republic of China
| |
Collapse
|
25
|
Ganjibakhsh M, Mehraein F, Koruji M, Aflatoonian R, Farzaneh P. Three-dimensional decellularized amnion membrane scaffold as a novel tool for cancer research; cell behavior, drug resistance and cancer stem cell content. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 100:330-340. [DOI: 10.1016/j.msec.2019.02.090] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 09/23/2018] [Revised: 02/21/2019] [Accepted: 02/22/2019] [Indexed: 12/15/2022]
|
26
|
Hu AX, Adams JJ, Vora P, Qazi M, Singh SK, Moffat J, Sidhu SS. EPH Profiling of BTIC Populations in Glioblastoma Multiforme Using CyTOF. Methods Mol Biol 2019; 1869:155-168. [PMID: 30324522 DOI: 10.1007/978-1-4939-8805-1_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 06/08/2023]
Abstract
The ability to elucidate the phenotype of brain tumor initiating cell (BTIC) in the context of bulk tumor in glioblastoma multiforme (GBM) provides significant therapeutic benefits for therapeutic evaluation. For the identification of such an elusive and rare subpopulation of cells, a single cell analysis technology with deep profiling capabilities known as Mass Cytometry (CyTOF) can prove to be highly useful. CyTOF circumvents the spectral overlap limitations of traditional flow cytometry by replacing fluorophores with metal isotope tags, allowing the accurate detection of significantly more parameters at the same time. In this chapter, we demonstrate that synthetic antibodies can be conjugated with metal isotope tags for CyTOF analysis, resulting in the development of a highly tailored, custom multi-parameter panel. This toolset was used to stain patient-derived GBM cells, which was analyzed via CyTOF. Analysis software viSNE and SPADE were applied to study the co-expression patterns of the Eph Receptor (EphR) family and several putative BTIC markers in GBM, resulting in the identification of a distinct group of cells consistent with a BTIC subpopulation. This approach can be readily adapted to the detection of cancer stem-like cells in other cancer types.
Collapse
Affiliation(s)
- Amy X Hu
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.
| | - Jarrett J Adams
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Parvez Vora
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Maleeha Qazi
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Sheila K Singh
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
- Department of Surgery, McMaster University, Hamilton, ON, Canada
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
| | - Jason Moffat
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
- Canadian Institute for Advanced Research, Toronto, ON, Canada
| | - Sachdev S Sidhu
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
27
|
Nunes T, Hamdan D, Leboeuf C, El Bouchtaoui M, Gapihan G, Nguyen TT, Meles S, Angeli E, Ratajczak P, Lu H, Di Benedetto M, Bousquet G, Janin A. Targeting Cancer Stem Cells to Overcome Chemoresistance. Int J Mol Sci 2018; 19:E4036. [PMID: 30551640 PMCID: PMC6321478 DOI: 10.3390/ijms19124036] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/20/2018] [Revised: 12/08/2018] [Accepted: 12/10/2018] [Indexed: 12/18/2022] Open
Abstract
Cancers are heterogeneous at the cell level, and the mechanisms leading to cancer heterogeneity could be clonal evolution or cancer stem cells. Cancer stem cells are resistant to most anti-cancer treatments and could be preferential targets to reverse this resistance, either targeting stemness pathways or cancer stem cell surface markers. Gold nanoparticles have emerged as innovative tools, particularly for photo-thermal therapy since they can be excited by laser to induce hyperthermia. Gold nanoparticles can be functionalized with antibodies to specifically target cancer stem cells. Preclinical studies using photo-thermal therapy have demonstrated the feasibility of targeting chemo-resistant cancer cells to reverse clinical chemoresistance. Here, we review the data linking cancer stem cells and chemoresistance and discuss the way to target them to reverse resistance. We particularly focus on the use of functionalized gold nanoparticles in the treatment of chemo-resistant metastatic cancers.
Collapse
Affiliation(s)
- Toni Nunes
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
- Laboratoire de Pathologie, Université Paris Diderot, Sorbonne Paris Cité, UMR_S1165, F-75010 Paris, France.
| | - Diaddin Hamdan
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
- Hôpital de La Porte Verte, F-78004 Versailles, France.
| | - Christophe Leboeuf
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
- Laboratoire de Pathologie, Université Paris Diderot, Sorbonne Paris Cité, UMR_S1165, F-75010 Paris, France.
| | - Morad El Bouchtaoui
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
- Laboratoire de Pathologie, Université Paris Diderot, Sorbonne Paris Cité, UMR_S1165, F-75010 Paris, France.
| | - Guillaume Gapihan
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
- Laboratoire de Pathologie, Université Paris Diderot, Sorbonne Paris Cité, UMR_S1165, F-75010 Paris, France.
| | - Thi Thuy Nguyen
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
| | - Solveig Meles
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
| | - Eurydice Angeli
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
| | - Philippe Ratajczak
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
- Laboratoire de Pathologie, Université Paris Diderot, Sorbonne Paris Cité, UMR_S1165, F-75010 Paris, France.
| | - He Lu
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
- Laboratoire de Pathologie, Université Paris Diderot, Sorbonne Paris Cité, UMR_S1165, F-75010 Paris, France.
| | - Mélanie Di Benedetto
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
- Laboratoire de Pathologie, Université Paris Diderot, Sorbonne Paris Cité, UMR_S1165, F-75010 Paris, France.
- Université Paris 13, F-93430 Villetaneuse, France.
| | - Guilhem Bousquet
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
- Laboratoire de Pathologie, Université Paris Diderot, Sorbonne Paris Cité, UMR_S1165, F-75010 Paris, France.
- Université Paris 13, F-93430 Villetaneuse, France.
- Service d'Oncologie Médicale, AP-HP-Hôpital Avicenne, F-93008 Bobigny, France.
| | - Anne Janin
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1165, F-75010 Paris, France.
- Laboratoire de Pathologie, Université Paris Diderot, Sorbonne Paris Cité, UMR_S1165, F-75010 Paris, France.
- Service de Pathologie, AP-HP-Hôpital Saint-Louis, F-75010 Paris, France.
| |
Collapse
|
28
|
Arai MA, Ochi F, Makita Y, Chiba T, Higashi K, Suganami A, Tamura Y, Toida T, Iwama A, Sadhu SK, Ahmed F, Ishibashi M. GLI1 Inhibitors Identified by Target Protein Oriented Natural Products Isolation (TPO-NAPI) with Hedgehog Inhibition. ACS Chem Biol 2018; 13:2551-2559. [PMID: 30160475 DOI: 10.1021/acschembio.8b00492] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/15/2023]
Abstract
This report describes the development of a target-protein-oriented natural-products-isolation (TPO-NAPI) method for Hedgehog inhibitors and the direct GLI1 inhibitor, 5'- O-methyl-3-hydroxyflemingin A (3), which inhibited hedgehog (Hh) signal transduction and diminished characteristics of cancer stem cells. Eight natural products (including three newly described products) that directly bind to GLI1 were rapidly obtained via the TPO-NAPI method developed using GLI1 protein-immobilized beads. 5'- O-Methyl-3-hydroxyflemingin A (3) inhibited Hh signaling (IC50 7.3 μM), leading to decreasing production of the Hh target proteins BCL2, PTCH1, and BMI1. 5'- O-Methyl-3-hydroxyflemingin A (3) was cytotoxic to Hh-related cancer cells. CD experiments revealed that 5'- O-methyl-3-hydroxyflemingin A (3) directly bound GLI1 ( Kd = 7.7 μM). Moreover, 5'- O-methyl-3-hydroxyflemingin A (3) diminished cancer stem cell characters of Huh7 such as sphere formation and production of the cancer stem cell marker EpCAM. These results suggest that Hh inhibitors can efficiently suppress the activity of cancer stem cells.
Collapse
Affiliation(s)
- Midori A. Arai
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Fumie Ochi
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Yoshinori Makita
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Tetsuhiro Chiba
- Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Kyohei Higashi
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Akiko Suganami
- Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Yutaka Tamura
- Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Toshihiko Toida
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Atsushi Iwama
- Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
- The Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Samir K. Sadhu
- Pharmacy Discipline, Khulna University, Khulna-9208, Bangladesh
| | - Firoj Ahmed
- Department of Pharmaceutical Chemistry, University of Dhaka, Dhaka-1000, Bangladesh
| | - Masami Ishibashi
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| |
Collapse
|
29
|
Affiliation(s)
- Amir Barzegar Behrooz
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Science, Universiti Putra Malaysia, Serdang, Malaysia
| | - Amir Syahir
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Science, Universiti Putra Malaysia, Serdang, Malaysia
| | - Syahida Ahmad
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Science, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
30
|
Liu R, Wei H, Gao P, Yu H, Wang K, Fu Z, Ju B, Zhao M, Dong S, Li Z, He Y, Huang Y, Yao Z. CD47 promotes ovarian cancer progression by inhibiting macrophage phagocytosis. Oncotarget 2018; 8:39021-39032. [PMID: 28380460 PMCID: PMC5503592 DOI: 10.18632/oncotarget.16547] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/20/2017] [Accepted: 02/22/2017] [Indexed: 12/24/2022] Open
Abstract
Targeting CD47 efficiently enhances macrophage phagocytosis in both physiological and pathological conditions. Anti-CD47 antibodies have been shown to inhibit the progression of several types of cancer. However, the mechanism of anti-CD47 monoclonal antibody (mAb) treatment remains controversial. In this study, we confirmed that CD47 protein is highly expressed in ovarian cancer, and is correlated with poor clinical characteristics and prognosis. CD47 knockdown in the ovarian cancer cell line, SK-OV-3, promoted phagocytosis by macrophages in vitro and inhibited tumor growth in vivo. These data combined suggest that CD47 inhibition is a potential strategy for cancer treatment. Using an anti-CD47 mAb, we found that CD47 inhibition in both SK-OV-3 cells and primary cancer cells was able to recapitulate our knockdown results and led to an increase in the number of infiltrating macrophages. In addition, the CD133+ tumor initiating cells expressed a high level of CD47, and anti-CD47 mAb treatment was able to trigger the phagocytosis of this cell population. In conclusion, our results indicate that CD47 inhibits macrophage phagocytosis of ovarian cancer cells, and down-regulation of CD47 or inhibiting CD47 by mAb was able to reverse the negative effect. Thus, CD47 antibody therapy may be a promising strategy to treat ovarian cancer.
Collapse
Affiliation(s)
- Ran Liu
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, China
| | - Huiting Wei
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300071, China
| | - Peng Gao
- University of the District of Columbia, Washington D.C., 20008, United States
| | - Hu Yu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300040, China
| | - Ke Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300040, China
| | - Zheng Fu
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300071, China
| | - Baohui Ju
- Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Meng Zhao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300040, China
| | - Shangwen Dong
- Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Zhijun Li
- Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yifeng He
- Department of Gynecology and Obstetrics, Renji Hospital, Shanghai, 200127, China
| | - Yuting Huang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300040, China
| | - Zhi Yao
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300071, China
| |
Collapse
|
31
|
Song L, Ye W, Cui Y, Lu J, Zhang Y, Ding N, Hu W, Pei H, Yue Z, Zhou G. Ecto-5'-nucleotidase (CD73) is a biomarker for clear cell renal carcinoma stem-like cells. Oncotarget 2018; 8:31977-31992. [PMID: 28404888 PMCID: PMC5458263 DOI: 10.18632/oncotarget.16667] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/29/2016] [Accepted: 03/17/2017] [Indexed: 12/16/2022] Open
Abstract
Identification of a specific biomarker for cancer stem cells (CSCs) is of potential applications in the development of effective therapeutic strategies for renal cell carcinoma (RCC). In this study, both the RCC cell line 786-O and surgically removed clear cell RCC (ccRCC) tissues were implemented to grew as spheroids in serum-free medium supplemented with mitogens. This subpopulation possessed key characteristics defining CSCs. We also identified that surgically removed ccRCC tissues were heterogenic and there was a subpopulation of cells that was highly stained with rhodamine-123. Based on membrane-proteomic analyses, CD73 was identified as a candidate biomarker. We further found that CD73high cells were highly tumorigenic. As few as 100 CD73high cells were capable of forming xenograft tumors in non obese diabetic/severe combined immunodeficiency disease mice, whereas 1 × 105 CD73low cells did not initiate tumor formation. During successive culture, the CD73high population regenerated both CD73high and CD73low cells, whereas the CD73low population remained low expression level of CD73. Furthermore, the CD73high cells were more resistant to radiation and DNA-damaging agents than the CD73low cells, and expressed a panel of 'stemness' genes at a higher level than the CD73low cells. These findings suggest that a high level of CD73 expression is a bona fide biomarker of ccRCC stem-like cells. Future research will aim at the elucidation of the underlying mechanisms of CD73 in RCC development and the distinct aspects of ccRCC stem-like cells from other tumor types.
Collapse
Affiliation(s)
- Lei Song
- Medical College, Northwest Minzu University, Lanzhou 730030, China.,Department of Space Radiobiology, Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modem Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Wenling Ye
- Medical College, Henan University, Kaifeng 475001, China.,Department of Space Radiobiology, Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modem Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Yong Cui
- Department of Urology Surgery, Shuyang Hospital of Traditional Chinese Medicine, Suqian 223600, China.,Department of Space Radiobiology, Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modem Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Jianzhong Lu
- Institute of Urology, Department of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Yanan Zhang
- Department of Space Radiobiology, Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modem Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Nan Ding
- Department of Space Radiobiology, Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modem Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Wentao Hu
- School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| | - Hailong Pei
- School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| | - Zhongjin Yue
- Institute of Urology, Department of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Guangming Zhou
- School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| |
Collapse
|
32
|
Resistance of glioma cells to nutrient-deprived microenvironment can be enhanced by CD133-mediated autophagy. Oncotarget 2018; 7:76238-76249. [PMID: 27780926 PMCID: PMC5342810 DOI: 10.18632/oncotarget.12803] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/28/2016] [Accepted: 09/24/2016] [Indexed: 01/04/2023] Open
Abstract
CD133 is a pentaspan transmembrane protein that can serve as a biomarker for cancer stem cells, although its biochemical mechanism remains unclear. Here we report that CD133 expression enhances glioma cell tolerance of a nutrient-deprived microenvironment. Under starvation conditions, CD133-positive cells exhibited higher survival and decreased levels of apoptosis. These changes were dependent on activation of autophagy-associated gene signaling and were impaired by the autophagic inhibitor chloroquine. Furthermore, rapamycin up-regulated the level of autophagy and inversely reduced CD133 expression. Immunofluorescence confirmed that starvation promoted release of CD133 from the plasma membrane to the cytoplasm, with CD133 also partially co-localizing with LC3 upon starvation. Additionally, CD133 partially co-localized with Beclin1, Atg5, and lysosomes, indicating that CD133 directly participates in the autophagosome membrane fusion process and ultimately undergoes lysosomal degradation. Collectively, our results demonstrate that CD133 contributes to cell survival by regulating autophagy, and that targeting CD133-linked signaling and autophagy may be useful in improving anti-cancer treatments.
Collapse
|
33
|
Deng J, Wang L, Chen H, Hao J, Ni J, Chang L, Duan W, Graham P, Li Y. Targeting epithelial-mesenchymal transition and cancer stem cells for chemoresistant ovarian cancer. Oncotarget 2018; 7:55771-55788. [PMID: 27304054 PMCID: PMC5342453 DOI: 10.18632/oncotarget.9908] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/11/2016] [Accepted: 05/30/2016] [Indexed: 12/29/2022] Open
Abstract
Chemoresistance is the main challenge for the recurrent ovarian cancer therapy and responsible for treatment failure and unfavorable clinical outcome. Understanding mechanisms of chemoresistance in ovarian cancer would help to predict disease progression, develop new therapies and personalize systemic therapy. In the last decade, accumulating evidence demonstrates that epithelial-mesenchymal transition and cancer stem cells play important roles in ovarian cancer chemoresistance and metastasis. Treatment of epithelial-mesenchymal transition and cancer stem cells holds promise for improving current ovarian cancer therapies and prolonging the survival of recurrent ovarian cancer patients in the future. In this review, we focus on the role of epithelial-mesenchymal transition and cancer stem cells in ovarian cancer chemoresistance and explore the therapeutic implications for developing epithelial-mesenchymal transition and cancer stem cells associated therapies for future ovarian cancer treatment.
Collapse
Affiliation(s)
- Junli Deng
- Cancer Care Centre, St George Hospital, Kogarah, NSW, Australia.,St George and Sutherland Clinical School, University of New South Wales (UNSW), Kensington, NSW, Australia.,Department of Gynecological Oncology, Henan Cancer Hospital, Zhengzhou, Henan, China.,Zhengzhou University, Zhengzhou, Henan, China
| | - Li Wang
- Department of Gynecological Oncology, Henan Cancer Hospital, Zhengzhou, Henan, China.,Zhengzhou University, Zhengzhou, Henan, China
| | - Hongmin Chen
- Department of Gynecological Oncology, Henan Cancer Hospital, Zhengzhou, Henan, China.,Zhengzhou University, Zhengzhou, Henan, China
| | - Jingli Hao
- Cancer Care Centre, St George Hospital, Kogarah, NSW, Australia.,St George and Sutherland Clinical School, University of New South Wales (UNSW), Kensington, NSW, Australia
| | - Jie Ni
- Cancer Care Centre, St George Hospital, Kogarah, NSW, Australia.,St George and Sutherland Clinical School, University of New South Wales (UNSW), Kensington, NSW, Australia
| | - Lei Chang
- Cancer Care Centre, St George Hospital, Kogarah, NSW, Australia.,St George and Sutherland Clinical School, University of New South Wales (UNSW), Kensington, NSW, Australia
| | - Wei Duan
- School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Peter Graham
- Cancer Care Centre, St George Hospital, Kogarah, NSW, Australia.,St George and Sutherland Clinical School, University of New South Wales (UNSW), Kensington, NSW, Australia
| | - Yong Li
- Cancer Care Centre, St George Hospital, Kogarah, NSW, Australia.,St George and Sutherland Clinical School, University of New South Wales (UNSW), Kensington, NSW, Australia
| |
Collapse
|
34
|
Al-Lamki RS, Wang J, Yang J, Burrows N, Maxwell PH, Eisen T, Warren AY, Vanharanta S, Pacey S, Vandenabeele P, Pober JS, Bradley JR. Tumor necrosis factor receptor 2-signaling in CD133-expressing cells in renal clear cell carcinoma. Oncotarget 2018; 7:24111-24. [PMID: 26992212 PMCID: PMC5029688 DOI: 10.18632/oncotarget.8125] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/30/2015] [Accepted: 03/02/2016] [Indexed: 01/29/2023] Open
Abstract
Compared to normal kidney, renal clear cell carcinomas (ccRCC) contain increased numbers of interstitial, non-hematopoietic CD133+cells that express stem cell markers and exhibit low rates of proliferation. These cells fail to form tumors upon transplantation but support tumor formation by differentiated malignant cells. We hypothesized that killing of ccRCC CD133+ (RCCCD133+) cells by cytotoxic agents might be enhanced by inducing them to divide. Since tumor necrosis factor-alpha (TNF), signalling through TNFR2, induces proliferation of malignant renal tubular epithelial cells, we investigated whether TNFR2 might similarly affect RCCCD133+cells. We compared treating organ cultures of ccRCC vs adjacent nontumour kidney (NK) and RCCCD133+vs NK CD133+ (NKCD133+) cell cultures with wild-type TNF (wtTNF) or TNF muteins selective for TNFR1 (R1TNF) or TNFR2 (R2TNF). In organ cultures, R2TNF increased expression of TNFR2 and promoted cell cycle entry of both RCCCD133+ and NKCD133+ but effects were greater in RCCCD133+. In contrast, R1TNF increased TNFR1 expression and promoted cell death. Importantly, cyclophosphamide triggered much more cell death in RCCCD133+ and NKCD133+cells pre-treated with R2TNF as compared to untreated controls. We conclude that selective engagement of TNFR2 by TNF can drives RCCCD133+ proliferation and thereby increase sensitivity to cell cycle-dependent cytotoxicity.
Collapse
Affiliation(s)
- Rafia S Al-Lamki
- Department of Medicine, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Jun Wang
- Department of Medicine, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Jun Yang
- Department of Medicine, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Natalie Burrows
- School of Clinical Medicine, Cambridge Institute of Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Patrick H Maxwell
- School of Clinical Medicine, Cambridge Institute of Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Timothy Eisen
- Department of Oncology, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Anne Y Warren
- Department of Pathology, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Sakari Vanharanta
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge CB2 0XZ, UK
| | - Simon Pacey
- Department of Oncology, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Peter Vandenabeele
- VIB Inflammation Research Center, Ghent University, UGhent-VIB Research Building FSVM, 9052 Ghent, Belgium
| | - Jordan S Pober
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520-8089, USA
| | - John R Bradley
- Department of Medicine, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
35
|
Chen HP, Lee YK, Huang SY, Shi PC, Hsu PC, Chang CF. Phthalate exposure promotes chemotherapeutic drug resistance in colon cancer cells. Oncotarget 2017; 9:13167-13180. [PMID: 29568348 PMCID: PMC5862569 DOI: 10.18632/oncotarget.23481] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/07/2016] [Accepted: 09/08/2017] [Indexed: 12/30/2022] Open
Abstract
Phthalates are widely used as plasticizers. Humans can be exposed to phthalates through ingestion, inhalation, or treatments that release di(2-ethylhexyl) phthalate (DEHP) and its metabolite, mono(2-ehylhexyl) phthalate (MEHP), into the body from polyvinyl chloride-based medical devices. Phthalate exposure may induce reproductive toxicity, liver damage, and carcinogenesis in humans. This study found that colon cancer cells exposed to DEHP or MEHP exhibited increased cell viability and increased levels of P-glycoprotein, CD133, Bcl-2, Akt, ERK, GSK3β, and β-catenin when treated with oxaliplatin or irinotecan, as compared to control. The P-glycoprotein inhibitor, tariquidar, which blocks drug efflux, reduced the viability of DEHP- or MEHP-treated, anti-cancer drug-challenged cells. DEHP or MEHP treatment also induced colon cancer cell migration and epithelial-mesenchymal transformation. Elevated stemness-related protein levels (β-catenin, Oct4, Sox2, and Nanog) and increased cell sphere sizes indicated that DEHP- or MEHP-treated cells were capable of self-renewal. We also found that serum DEHP concentrations were positively correlated with cancer recurrence. These results suggest phthalate exposure enhances colon cancer cell metastasis and chemotherapeutic drug resistance by increasing cancer cell stemness, and that P-glycoprotein inhibitors might improve outcomes for advanced or drug-resistant colon cancer patients.
Collapse
Affiliation(s)
- Hsin-Pao Chen
- Department of Surgery, E-DA Hospital, I-Shou University, Kaohsiung 824, Taiwan.,Department of Safety, Health and Environmental Engineering, National Kaohsiung First University of Science and Technology, Kaohsiung 811, Taiwan
| | - Yung-Kuo Lee
- Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.,Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Shih Yin Huang
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Pei-Chun Shi
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Ping-Chi Hsu
- Department of Safety, Health and Environmental Engineering, National Kaohsiung First University of Science and Technology, Kaohsiung 811, Taiwan
| | - Chuan-Fa Chang
- Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.,Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.,Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| |
Collapse
|
36
|
Su YK, Shih PH, Lee WH, Bamodu OA, Wu ATH, Huang CC, Tzeng YM, Hsiao M, Yeh CT, Lin CM. Antrodia cinnamomea sensitizes radio-/chemo-therapy of cancer stem-like cells by modulating microRNA expression. JOURNAL OF ETHNOPHARMACOLOGY 2017; 207:47-56. [PMID: 28602756 DOI: 10.1016/j.jep.2017.06.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 09/28/2016] [Revised: 06/02/2017] [Accepted: 06/03/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The discovery of many tissue-specific cancer stem cells (CSCs) continues to attract scientific attention. These CSCs are considered to be associated with chemo- and radio-resistance, and consequently, failure of conventional anticancer therapies. The recent demonstration of several microRNAs as enhancers of tumorigenicity via modulation of epithelial-mesenchymal transition and cancer stemness, makes them putative novel therapeutic target in oncology. Antrodia cinnamomea is a Chinese traditional medicine with several biological functions including anti-inflammation, antioxidant, and cancer prevention. However, the anti-CSC capability of A. Cinnamomea is not clear yet. AIM OF THE STUDY To investigate the inhibitory effect of A. cinnamomea mycelium and extract on CSCs derived from various human cancer cell lines using our in-house therapeutics and human genome-wide miRNA screening panels. MATERIALS AND METHODS A broad range of human cancer cell lines, including the acute monocytic leukemia (THP-1), glioblastoma multiforme (GBM 8401), lung carcinoma (A549), breast adenocarcinoma (MDA-MB-231), hepatoblastoma (HepG2), colorectal adenocarcinoma (SW620), and foreskin fibroblast (HS68), were exposed to A. cinnamomea in this study. CD133+ CSCs generated from the cell lines were characterized and isolated by flow cytometry, effect of chemo- and radiotherapy was assessed using the MTT assay, while the RT-PCR and human genome wide qRT-PCR determined the differential gene expression patterns. A comparative analysis of the anticancer effect of A. cinnamomea and Cisplatin, Taxol, or irradiation was also performed. RESULTS Our results indicated that A. cinnamomea mycelium and its ethyl acetate extracts showed anti-proliferation effects against all types of CSCs, especially the lung, breast, and head and neck squamous cell carcinoma CSCs. Furthermore, CSCs treatment with A. cinnamomea combined with irradiation or chemotherapeutics demonstrated significant anti-cancer effect. We also established an association between the CSC-inhibitory effect of A. cinnamomea and significant downregulation of several microRNAs and cancer stemness expression levels in brain and breast CSCs. More importantly, higher CD133 expression is associated with poor prognosis in glioblastoma and breast cancer patients. CONCLUSION Herein, we demonstrate the putative role of A. cinnamomea as an effective ethnopharmacologic therapeutic agent for cancer treatment.
Collapse
Affiliation(s)
- Yu-Kai Su
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Neurosurgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ping-Hsiao Shih
- Department of Pediatrics, Taipei Medical University-Shuang Ho Hospital, Taipei, Taiwan; Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Wei-Hwa Lee
- Department of Pathology, Taipei Medical University-Shuang Ho Hospital, Taipei, Taiwan
| | - Oluwaseun Adebayo Bamodu
- Cancer Center, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Department of Medical Research & Education, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
| | - Alexander T H Wu
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chun-Chih Huang
- Department of Life Science, National Taitung University, Taitung, Taiwan
| | - Yew-Min Tzeng
- Department of Life Science, National Taitung University, Taitung, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Chi-Tai Yeh
- Cancer Center, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Department of Medical Research & Education, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan.
| | - Chien-Min Lin
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Neurosurgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
37
|
Colorectal Cancer: From the Genetic Model to Posttranscriptional Regulation by Noncoding RNAs. BIOMED RESEARCH INTERNATIONAL 2017; 2017:7354260. [PMID: 28573140 PMCID: PMC5442347 DOI: 10.1155/2017/7354260] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 12/05/2016] [Accepted: 02/16/2017] [Indexed: 12/11/2022]
Abstract
Colorectal cancer is the third most common form of cancer in developed countries and, despite the improvements achieved in its treatment options, remains as one of the main causes of cancer-related death. In this review, we first focus on colorectal carcinogenesis and on the genetic and epigenetic alterations involved. In addition, noncoding RNAs have been shown to be important regulators of gene expression. We present a general overview of what is known about these molecules and their role and dysregulation in cancer, with a special focus on the biogenesis, characteristics, and function of microRNAs. These molecules are important regulators of carcinogenesis, progression, invasion, angiogenesis, and metastases in cancer, including colorectal cancer. For this reason, miRNAs can be used as potential biomarkers for diagnosis, prognosis, and efficacy of chemotherapeutic treatments, or even as therapeutic agents, or as targets by themselves. Thus, this review highlights the importance of miRNAs in the development, progression, diagnosis, and therapy of colorectal cancer and summarizes current therapeutic approaches for the treatment of colorectal cancer.
Collapse
|
38
|
Bielecka ZF, Maliszewska-Olejniczak K, Safir IJ, Szczylik C, Czarnecka AM. Three-dimensional cell culture model utilization in cancer stem cell research. Biol Rev Camb Philos Soc 2016; 92:1505-1520. [DOI: 10.1111/brv.12293] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/18/2015] [Revised: 06/20/2016] [Accepted: 06/27/2016] [Indexed: 12/19/2022]
Affiliation(s)
- Zofia F. Bielecka
- Department of Oncology with Laboratory of Molecular Oncology; Military Institute of Medicine; Szaserów 128 04-141 Warsaw Poland
- Postgraduate School of Molecular Medicine; Medical University of Warsaw; Zwirki i Wigury 61 02-109 Warsaw Poland
| | - Kamila Maliszewska-Olejniczak
- Department of Oncology with Laboratory of Molecular Oncology; Military Institute of Medicine; Szaserów 128 04-141 Warsaw Poland
- Laboratory of DNA Sequencing and Oligonucleotides Synthesis, Institute of Biochemistry and Biophysics; Polish Academy of Sciences; Pawinskiego 5a 02-106 Warsaw Poland
| | - Ilan J. Safir
- Department of Urology; Emory University School of Medicine; Atlanta GA 30322 U.S.A
| | - Cezary Szczylik
- Department of Oncology with Laboratory of Molecular Oncology; Military Institute of Medicine; Szaserów 128 04-141 Warsaw Poland
| | - Anna M. Czarnecka
- Department of Oncology with Laboratory of Molecular Oncology; Military Institute of Medicine; Szaserów 128 04-141 Warsaw Poland
| |
Collapse
|
39
|
Liu Y, Ren S, Xie L, Cui C, Xing Y, Liu C, Cao B, Yang F, Li Y, Chen X, Wei Y, Lu H, Jiang J. Mutation of N-linked glycosylation at Asn548 in CD133 decreases its ability to promote hepatoma cell growth. Oncotarget 2016; 6:20650-60. [PMID: 26029999 PMCID: PMC4653032 DOI: 10.18632/oncotarget.4115] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/11/2015] [Accepted: 04/20/2015] [Indexed: 01/10/2023] Open
Abstract
The membrane glycoprotein CD133 is a popular marker for cancer stem cells and contributes to cancer initiation and invasion in a number of tumor types. CD133 promotes tumorigenesis partly through an interaction between its phosphorylated Y828 residue and the PI3K regulatory subunit p85, and the interaction with β-catenin. Although CD133 glycosylation is supposed to be associated with its function, the contribution of N-glycosylation to its functions remains unclear. Here we analyzed the exact site(s) of N-glycosylation in CD133 by mass spectrometry and found that all eight potential N-glycosylation sites of CD133 could be indeed occupied by N-glycans. Loss of individual N-glycosylation sites had no effect on the level of expression or membrane localization of CD133. However, mutation at glycosylation site Asn548 significantly decreased the ability of CD133 to promote hepatoma cell growth. Furthermore, mutation of Asn548 reduced the interaction between CD133 and β-catenin and inhibited the activation of β-catenin signaling by CD133 overexpression. Our results identified the characteristics and function of CD133 glycosylation sites. These data could potentially shed light on molecular regulation of CD133 by glycosylation and enhance our understanding of the utility of glycosylated CD133 as a target for cancer therapies.
Collapse
Affiliation(s)
- Ying Liu
- Key Laboratory of Glycoconjuates Research, Ministry of Public Health and Gene Research Center, Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| | - Shifang Ren
- Key Laboratory of Glycoconjuates Research, Ministry of Public Health and Gene Research Center, Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| | - Liqi Xie
- Institutes of Biomedical Sciences of Fudan University, Shanghai, People's Republic of China
| | - Chunhong Cui
- Key Laboratory of Glycoconjuates Research, Ministry of Public Health and Gene Research Center, Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| | - Yang Xing
- Key Laboratory of Glycoconjuates Research, Ministry of Public Health and Gene Research Center, Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| | - Chanjuan Liu
- Key Laboratory of Glycoconjuates Research, Ministry of Public Health and Gene Research Center, Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| | - Benjin Cao
- Key Laboratory of Glycoconjuates Research, Ministry of Public Health and Gene Research Center, Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| | - Fan Yang
- Key Laboratory of Glycoconjuates Research, Ministry of Public Health and Gene Research Center, Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| | - Yinan Li
- Key Laboratory of Glycoconjuates Research, Ministry of Public Health and Gene Research Center, Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| | - Xiaoning Chen
- Key Laboratory of Glycoconjuates Research, Ministry of Public Health and Gene Research Center, Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| | - Yuanyan Wei
- Key Laboratory of Glycoconjuates Research, Ministry of Public Health and Gene Research Center, Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| | - Haojie Lu
- Institutes of Biomedical Sciences of Fudan University, Shanghai, People's Republic of China
| | - Jianhai Jiang
- Key Laboratory of Glycoconjuates Research, Ministry of Public Health and Gene Research Center, Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
40
|
Lim W, Kim HE, Kim Y, Na R, Li X, Jeon S, Choi H, Kim O. Association between cancer stem cell-like properties and epithelial-to-mesenchymal transition in primary and secondary cancer cells. Int J Oncol 2016; 49:991-1000. [PMID: 27315437 DOI: 10.3892/ijo.2016.3582] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/15/2016] [Accepted: 06/06/2016] [Indexed: 11/06/2022] Open
Abstract
One of the theories on cancer stem cells (CSCs) states that these cells initiate most tumors and give rise to more-or-less differentiated tumor cells. Genetic signatures of CSCs are thought to predict tumor recurrence and metastases, thus, supporting the notion that CSCs may be metastatic precursors and induce epithelial-to-mesenchymal transition (EMT). In this study, we tried to examine the association between CSCs and EMT (using specific markers) in the mucoepidermoid carcinoma cell line YD15 and its derivative cell line YD15M (lymph node metastasis). Relative protein expression levels were analyzed by western blotting, flow cytometry, and immunofluorescence assays. In addition, cell cycle assay and aldehyde dehydrogenase (ALDH) activity assay were carried out. Under growth conditions, YD15M cells formed irregular spherical colonies consistent with a stem cell phenotype. YD15M cells demonstrated the low expression of E-cadherin and β-catenin but high expression of vimentin than that in YD15 cells. In the metastatic cells (YD15M), the coexpression of vimentin and CD133 was detected. Weak proliferation based on cell cycle analysis and decreased PCNA expression was also observed. In addition, expression levels of ALDHA1, OCT4, and NANOG (CSC-like properties) were significantly increased in YD15M cells. Taken together, these findings should help to elucidate the interplay between EMT and CSC-like properties during metastasis and may provide useful information for the development of a novel classification system and therapeutic strategies against head and neck cancer.
Collapse
Affiliation(s)
- Wonbong Lim
- Department of Premedical Science, College of Medicine, Chosun University, Dong-Gu, Gwangju, Republic of Korea
| | - Hye-Eun Kim
- Department of Oral Pathology, Dental Science Research Institute and Medical Research Center for Biomineralization Disorders, School of Dentistry, Chonnam National University, Bug-Gu, Gwangju, Republic of Korea
| | - Young Kim
- Department of Oral Pathology, Dental Science Research Institute and Medical Research Center for Biomineralization Disorders, School of Dentistry, Chonnam National University, Bug-Gu, Gwangju, Republic of Korea
| | - Risu Na
- Department of Oral Pathology, Dental Science Research Institute and Medical Research Center for Biomineralization Disorders, School of Dentistry, Chonnam National University, Bug-Gu, Gwangju, Republic of Korea
| | - Xiaojie Li
- Stomatology College of Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Sangmi Jeon
- Department of Oral Pathology, Dental Science Research Institute and Medical Research Center for Biomineralization Disorders, School of Dentistry, Chonnam National University, Bug-Gu, Gwangju, Republic of Korea
| | - Hongran Choi
- Department of Oral Pathology, Dental Science Research Institute and Medical Research Center for Biomineralization Disorders, School of Dentistry, Chonnam National University, Bug-Gu, Gwangju, Republic of Korea
| | - Okjoon Kim
- Department of Oral Pathology, Dental Science Research Institute and Medical Research Center for Biomineralization Disorders, School of Dentistry, Chonnam National University, Bug-Gu, Gwangju, Republic of Korea
| |
Collapse
|
41
|
Abstract
The discovery and subsequent isolation of tumor-initiating cells (TICs), a small population of highly tumorigenic and drug-resistant cancer cells also called cancer stem cells (CSCs), have revolutionized our understanding of cancer. TICs are isolated using various methodologies, including selection of surface marker expression, ALDH activity, suspension culture, and chemotherapy/drug resistance. These methods have several drawbacks, including their variability, lack of robustness and scalability, and low specificity. Alternative methods of purification take advantage of biophysical properties of TICs including their adhesion and stiffness. This review will provide a brief overview of TIC biology as well as review the most important methods of TIC isolation with a focus on biophysical methods of TIC purification.
Collapse
Affiliation(s)
- Efraín A. Cermeño
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA
| | - Andrés J. García
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA
| |
Collapse
|
42
|
Affiliation(s)
- Rumena Petkova
- Scientific Technological Service (STS), Ltd., Sofia, Bulgaria
| | - Stoyan Chakarov
- Faculty of Biology, Department of Biochemistry, Sofia University “St. Kliment Ohridsky”, Sofia, Bulgaria
| |
Collapse
|
43
|
Yan B, Dong L, Neuzil J. Mitochondria: An intriguing target for killing tumour-initiating cells. Mitochondrion 2016; 26:86-93. [DOI: 10.1016/j.mito.2015.12.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/23/2015] [Revised: 11/25/2015] [Accepted: 12/14/2015] [Indexed: 12/12/2022]
|
44
|
Borah A, Raveendran S, Rochani A, Maekawa T, Kumar DS. Targeting self-renewal pathways in cancer stem cells: clinical implications for cancer therapy. Oncogenesis 2015; 4:e177. [PMID: 26619402 PMCID: PMC4670961 DOI: 10.1038/oncsis.2015.35] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/16/2015] [Revised: 09/10/2015] [Accepted: 09/22/2015] [Indexed: 12/21/2022] Open
Abstract
Extensive cancer research in the past few decades has identified the existence of a rare subpopulation of stem cells in the grove of cancer cells. These cells are known as the cancer stem cells marked by the presence of surface biomarkers, multi-drug resistance pumps and deregulated self-renewal pathways (SRPs). They have a crucial role in provoking cancer cells leading to tumorigenesis and its progressive metastasis. Cancer stem cells (CSCs) are much alike to normal stem cells in their self-renewal mechanisms. However, deregulations in the SRPs are seen in CSCs, making them resistant to conventional chemotherapeutic agents resulting in the tumor recurrence. Current treatment strategies in cancer fail to detect and differentiate the CSCs from their non-tumorigenic progenies owing to absence of specific biomarkers. Now, it has become imperative to understand complex functional biology of CSCs, especially the signaling pathways to design improved treatment strategies to target them. It is hopeful that the SRPs in CSCs offer a promising target to alter their survival strategies and impede their tumorigenic potential. However, there are many perils associated with the direct targeting method by conventional therapeutic agents such as off targets, poor bioavailability and poor cellular distribution. Recent evidences have shown an increased use of small molecule antagonists directly to target these SRPs may lead to severe side-effects. An alternative to solve these issues could be an appropriate nanoformulation. Nanoformulations of these molecules could provide an added advantage for the selective targeting of the pathways especially Hedgehog, Wnt, Notch and B-cell-specific moloney murine leukemia virus integration site 1 in the CSCs while sparing the normal stem cells. Hence, to achieve this goal a complete understanding of the molecular pathways corroborate with the use of holistic nanosystem (nanomaterial inhibition molecule) could possibly be an encouraging direction for future cancer therapy.
Collapse
Affiliation(s)
- A Borah
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama, Japan
| | - S Raveendran
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama, Japan
| | - A Rochani
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama, Japan
| | - T Maekawa
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama, Japan
| | - D S Kumar
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama, Japan
| |
Collapse
|
45
|
Novel Implications of DNA Damage Response in Drug Resistance of Malignant Cancers Obtained from the Functional Interaction between p53 Family and RUNX2. Biomolecules 2015; 5:2854-76. [PMID: 26512706 PMCID: PMC4693260 DOI: 10.3390/biom5042854] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/02/2015] [Revised: 09/17/2015] [Accepted: 10/16/2015] [Indexed: 12/31/2022] Open
Abstract
During the lifespan of cells, their genomic DNA is continuously exposed to the endogenous and exogenous DNA insults. Thus, the appropriate cellular response to DNA damage plays a pivotal role in maintaining genomic integrity and also acts as a molecular barrier towards DNA legion-mediated carcinogenesis. The tumor suppressor p53 participates in an integral part of proper regulation of DNA damage response (DDR). p53 is frequently mutated in a variety of human cancers. Since mutant p53 displays a dominant-negative behavior against wild-type p53, cancers expressing mutant p53 sometimes acquire drug-resistant phenotype, suggesting that mutant p53 prohibits the p53-dependent cell death pathway following DNA damage, and thereby contributing to the acquisition and/or maintenance of drug resistance of malignant cancers. Intriguingly, we have recently found that silencing of pro-oncogenic RUNX2 enhances drug sensitivity of aggressive cancer cells regardless of p53 status. Meanwhile, cancer stem cells (CSCs) have stem cell properties such as drug resistance. Therefore, the precise understanding of the biology of CSCs is quite important to overcome their drug resistance. In this review, we focus on molecular mechanisms behind DDR as well as the serious drug resistance of malignant cancers and discuss some attractive approaches to improving the outcomes of patients bearing drug-resistant cancers.
Collapse
|
46
|
Song IS, Jeong YJ, Jeong SH, Heo HJ, Kim HK, Bae KB, Park YH, Kim SU, Kim JM, Kim N, Ko KS, Rhee BD, Han J. FOXM1-Induced PRX3 Regulates Stemness and Survival of Colon Cancer Cells via Maintenance of Mitochondrial Function. Gastroenterology 2015; 149:1006-16.e9. [PMID: 26091938 DOI: 10.1053/j.gastro.2015.06.007] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 11/16/2014] [Revised: 06/01/2015] [Accepted: 06/03/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS Reagents designed to target cancer stem cells (CSCs) could reduce tumor growth, recurrence, and metastasis. We investigated the mitochondrial features of CSCs. METHODS Colon adenocarcinoma fragments were obtained from 8 patients during surgery at Busan Paik Hospital in Korea. We used immunohistochemistry and quantitative polymerase chain reaction to compare expression of mitochondrial peroxiredoxin 3 (PRX3) in CD133(+)CD44(+) Lgr5(+)cells (CSCs) vs CD133(-)CD44(-)Lgr5(-) colon tumor cells (non-CSCs). Cell survival and expression of mitochondrial-related genes were analyzed in the presence of 5-fluorouracil and/or antimycin A. We used small-interfering and short-hairpin RNAs and an overexpression vector to study PRX3, which functions in the mitochondria. CD133(+) cells with PRX3 knockdown or overexpressing PRX3 were grown as xenograft tumors in immunocompromised mice. Metastasis was studied after injection of tumor cells in spleens of mice. We used chromatin immunoprecipitation and reporter assays to characterize transcriptional regulation of PRX3 by forkhead box protein 1. RESULTS CSCs had a higher mitochondrial membrane potential and increased levels of adenosine triphosphate, Ca(2+), reactive oxygen species, and oxygen consumption than non-CSCs. Levels of PRX3 were increased in colon CSCs compared with non-CSCs. PRX3 knockdown reduced the viability of CSCs, but non non-CSCs, by inducing mitochondrial dysfunction. PRX3 knockdown reduced growth of CSCs as xenograft tumors or metastases in mice. The expression of FOXM1 activated transcription of PRX3 and expression of CD133 in colon CSCs. CONCLUSIONS Human colon CSCs have increased mitochondrial function compared with colon tumor cells without stem cell properties. Colon CSCs overexpress the mitochondrial gene PRX3, which is required for maintenance of mitochondrial function and tumorigenesis, and is regulated by forkhead box protein 1, which also regulates expression of CD133 in these cells. These proteins might be therapeutic targets for colorectal cancer.
Collapse
Affiliation(s)
- In-Sung Song
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - Yu Jeong Jeong
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - Seung Hun Jeong
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - Hye Jin Heo
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - Hyoung Kyu Kim
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - Ki Beom Bae
- Department of Surgery, College of Medicine, Inje University, Busan Paik Hospital, Busan, Korea
| | - Young-Ho Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Sun Uk Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Jin-Man Kim
- Department of Pathology, School of Medicine, Chungnam National University, Daejeon, Korea
| | - Nari Kim
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - Kyung Soo Ko
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - Byoung Doo Rhee
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea
| | - Jin Han
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea.
| |
Collapse
|
47
|
Kaushal GS, Rognoni E, Lichtenberger BM, Driskell RR, Kretzschmar K, Hoste E, Watt FM. Fate of Prominin-1 Expressing Dermal Papilla Cells during Homeostasis, Wound Healing and Wnt Activation. J Invest Dermatol 2015; 135:2926-2934. [PMID: 26288357 PMCID: PMC4650270 DOI: 10.1038/jid.2015.319] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/23/2015] [Revised: 07/05/2015] [Accepted: 07/22/2015] [Indexed: 12/23/2022]
Abstract
Prominin-1/CD133 (Prom1) is expressed by fibroblasts in the dermal papilla (DP) of the hair follicle (HF). By examining endogenous Prom1 expression and expression of LacZ in the skin of Prom1CreERLacZ (Prom1C-L) mice, in which a CreERT2-IRES-nuclear LacZ cassette is knocked into the first ATG codon of Prom1, we confirmed that Prom1 is expressed in the DP of all developing HFs and also by postnatal anagen follicles. To analyze the fate of Prom1+ DP cells, we crossed Prom1C-L mice with Rosa26-CAG flox/stop/flox tdTomato reporter mice and applied 4-hydroxytamoxifen (4OHT) to back skin at postnatal day (P) 1 and P2. We detected tdTomato+ cells in ~50% of DPs. The proportion of labeled cells per DP increased between P5 and P63, while the total number of cells per DP declined. Following full thickness wounding, there was no migration of tdTomato-labeled cells out of the DP. When β-catenin was activated in Prom1+ DP cells there was an increase in the size of anagen and telogen DP, but the proportion of tdTomato-labeled cells did not increase. We conclude that Prom1+ DP cells do not contribute to dermal repair but are nevertheless capable of regulating DP size via β-catenin-mediated intercellular communication.
Collapse
Affiliation(s)
- Grace S Kaushal
- King's College London, Centre for Stem Cells and Regenerative Medicine, Guy's Hospital Campus, London, UK
| | - Emanuel Rognoni
- King's College London, Centre for Stem Cells and Regenerative Medicine, Guy's Hospital Campus, London, UK
| | - Beate M Lichtenberger
- King's College London, Centre for Stem Cells and Regenerative Medicine, Guy's Hospital Campus, London, UK
| | - Ryan R Driskell
- King's College London, Centre for Stem Cells and Regenerative Medicine, Guy's Hospital Campus, London, UK
| | - Kai Kretzschmar
- King's College London, Centre for Stem Cells and Regenerative Medicine, Guy's Hospital Campus, London, UK
| | - Esther Hoste
- King's College London, Centre for Stem Cells and Regenerative Medicine, Guy's Hospital Campus, London, UK
| | - Fiona M Watt
- King's College London, Centre for Stem Cells and Regenerative Medicine, Guy's Hospital Campus, London, UK.
| |
Collapse
|
48
|
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain tumor and is notorious for its poor prognosis. The highly invasive nature of GBM and its inherent resistance to therapy lead to very high rates of recurrence. Recently, a small cohort of tumor cells, called cancer stem cells (CSCs), has been recognized as a subset of tumor cells with self-renewal ability and multilineage capacity. These properties, along with the remarkable tumorigenicity of CSCs, are thought to account for the high rates of tumor recurrence after treatment. Recent research has been geared toward understanding the unique biological characteristics of CSCs to enable development of targeted therapy. Strategies include inhibition of CSC-specific pathways and receptors; agents that increase sensitivity of CSCs to chemotherapy and radiotherapy; CSC differentiation agents; and CSC-specific immunotherapy, virotherapy, and gene therapy. These approaches could inform the development of newer therapeutics for GBM.
Collapse
|
49
|
Saricanbaz I, Karahacioglu E, Ekinci O, Bora H, Kilic D, Akmansu M. Prognostic significance of expression of CD133 and Ki-67 in gastric cancer. Asian Pac J Cancer Prev 2015; 15:8215-9. [PMID: 25339008 DOI: 10.7314/apjcp.2014.15.19.8215] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/10/2022] Open
Abstract
CD133 is one of the most important stem cell markers in solid cancers and Ki-67 is a marker that reflects cell proliferation. The relationships between the expression of CD133 and Ki-67 and prognosis in gastric carcinoma are unknown and need exploring. We examined 50 gastric cancer patients retrospectively in the Radiation Oncology Department of the Faculty of Medicine, Gazi University. CD133 and Ki-67 expression was examined using immunohistochemical staining. The survival rate in patients with CD133 positive expression was significantly worse than that in the patients with negative expression (p=0.04). Expression of CD133 had a positive correlation with that of Ki-67 (r=0.350; p=0.014). Multivariate analysis revealed that the expression of CD133 was an independent prognostic factor in gastric cancer (p=0.02). Conclusion, expression of CD133 may be a useful prognostic marker in gastric cancer.
Collapse
Affiliation(s)
- Irem Saricanbaz
- Department of Radiation Oncology, Konya Training and Research Hospital, Konya, Turkey E-mail :
| | | | | | | | | | | |
Collapse
|
50
|
Neradil J, Veselska R. Nestin as a marker of cancer stem cells. Cancer Sci 2015; 106:803-11. [PMID: 25940879 PMCID: PMC4520630 DOI: 10.1111/cas.12691] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/19/2015] [Revised: 04/14/2015] [Accepted: 04/27/2015] [Indexed: 12/14/2022] Open
Abstract
The crucial role of cancer stem cells (CSCs) in the pathology of malignant diseases has been extensively studied during the last decade. Nestin, a class VI intermediate filament protein, was originally detected in neural stem cells during development. Its expression has also been reported in different tissues under various pathological conditions. Specifically, nestin has been shown to be expressed in transformed cells of various human malignancies, and a correlation between its expression and the clinical course of some diseases has been proved. Furthermore, the coexpression of nestin with other stem cell markers was described as a CSC phenotype that was subsequently verified using tumorigenicity assays. The primary aim of this review is to summarize the recent findings regarding nestin expression in CSCs, its possible role in CSC phenotypes, particularly with respect to capacity for self-renewal, and its utility as a putative marker of CSCs.
Collapse
Affiliation(s)
- Jakub Neradil
- Laboratory of Tumor Biology, Department of Experimental Biology, School of Science, Masaryk University, Brno, Czech Republic.,Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Renata Veselska
- Laboratory of Tumor Biology, Department of Experimental Biology, School of Science, Masaryk University, Brno, Czech Republic.,Department of Pediatric Oncology, University Hospital Brno and School of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|