1
|
Ríos-Arrabal S, Puentes-Pardo JD, Moreno-SanJuan S, Szuba Á, Casado J, García-Costela M, Escudero-Feliu J, Verbeni M, Cano C, González-Puga C, Martín-Lagos Maldonado A, Carazo Á, León J. Endothelin-1 as a Mediator of Heme Oxygenase-1-Induced Stemness in Colorectal Cancer: Influence of p53. J Pers Med 2021; 11:jpm11060509. [PMID: 34199777 PMCID: PMC8227293 DOI: 10.3390/jpm11060509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/26/2021] [Accepted: 06/01/2021] [Indexed: 12/24/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is an antioxidant protein implicated in tumor progression, metastasis, and resistance to therapy. Elevated HO-1 expression is associated with stemness in several types of cancer, although this aspect has not yet been studied in colorectal cancer (CRC). Using an in vitro model, we demonstrated that HO-1 overexpression regulates stemness and resistance to 5-FU treatment, regardless of p53. In samples from CRC patients, HO-1 and endothelin converting enzyme-1 (ECE-1) expression correlated significantly, and p53 had no influence on this result. Carbon monoxide (CO) activated the ECE-1/endothelin-1 (ET-1) pathway, which could account for the protumoral effects of HO-1 in p53 wild-type cells, as demonstrated after treatment with bosentan (an antagonist of both ETRA and ETRB endothelin-1 receptors). Surprisingly, in cells with a non-active p53 or a mutated p53 with gain-of-function, ECE-1-produced ET-1 acted as a protective molecule, since treatment with bosentan led to increased efficiency for spheres formation and percentage of cancer stem cells (CSCs) markers. In these cells, HO-1 could activate or inactivate certain unknown routes that could induce these contrary responses after treatment with bosentan in our cell model. However more research is warranted to confirm these results. Patients carrying tumors with a high expression of both HO-1 and ECE-1 and a non-wild-type p53 should be considered for HO-1 based-therapies instead of ET-1 antagonists-based ones.
Collapse
Affiliation(s)
- Sandra Ríos-Arrabal
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain; (S.R.-A.); (J.D.P.-P.); (S.M.-S.); (J.C.); (M.G.-C.); (J.E.-F.); (C.G.-P.); (A.M.-L.M.); (Á.C.)
| | - Jose D. Puentes-Pardo
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain; (S.R.-A.); (J.D.P.-P.); (S.M.-S.); (J.C.); (M.G.-C.); (J.E.-F.); (C.G.-P.); (A.M.-L.M.); (Á.C.)
- Departamento de Farmacología, Facultad de Farmacia, Universidad de Granada, 18071 Granada, Spain
| | - Sara Moreno-SanJuan
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain; (S.R.-A.); (J.D.P.-P.); (S.M.-S.); (J.C.); (M.G.-C.); (J.E.-F.); (C.G.-P.); (A.M.-L.M.); (Á.C.)
- Cytometry and Microscopy Research Service, Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain
| | - Ágata Szuba
- Unidad de Gestión Clínica de Cirugía, Complejo Hospitalario de Jaén, 23007 Jaén, Spain;
| | - Jorge Casado
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain; (S.R.-A.); (J.D.P.-P.); (S.M.-S.); (J.C.); (M.G.-C.); (J.E.-F.); (C.G.-P.); (A.M.-L.M.); (Á.C.)
| | - María García-Costela
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain; (S.R.-A.); (J.D.P.-P.); (S.M.-S.); (J.C.); (M.G.-C.); (J.E.-F.); (C.G.-P.); (A.M.-L.M.); (Á.C.)
| | - Julia Escudero-Feliu
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain; (S.R.-A.); (J.D.P.-P.); (S.M.-S.); (J.C.); (M.G.-C.); (J.E.-F.); (C.G.-P.); (A.M.-L.M.); (Á.C.)
| | - Michela Verbeni
- Departamento de Ciencias de la Computación e Inteligencia Artificial, E.T.S. de Ingenierías Informática y de Telecomunicación, Universidad de Granada, 18014 Granada, Spain; (M.V.); (C.C.)
| | - Carlos Cano
- Departamento de Ciencias de la Computación e Inteligencia Artificial, E.T.S. de Ingenierías Informática y de Telecomunicación, Universidad de Granada, 18014 Granada, Spain; (M.V.); (C.C.)
| | - Cristina González-Puga
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain; (S.R.-A.); (J.D.P.-P.); (S.M.-S.); (J.C.); (M.G.-C.); (J.E.-F.); (C.G.-P.); (A.M.-L.M.); (Á.C.)
- Unidad de Gestión Clínica de Cirugía, Hospital Universitario San Cecilio de Granada, 18016 Granada, Spain
| | - Alicia Martín-Lagos Maldonado
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain; (S.R.-A.); (J.D.P.-P.); (S.M.-S.); (J.C.); (M.G.-C.); (J.E.-F.); (C.G.-P.); (A.M.-L.M.); (Á.C.)
- Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario San Cecilio de Granada, 18016 Granada, Spain
| | - Ángel Carazo
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain; (S.R.-A.); (J.D.P.-P.); (S.M.-S.); (J.C.); (M.G.-C.); (J.E.-F.); (C.G.-P.); (A.M.-L.M.); (Á.C.)
| | - Josefa León
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain; (S.R.-A.); (J.D.P.-P.); (S.M.-S.); (J.C.); (M.G.-C.); (J.E.-F.); (C.G.-P.); (A.M.-L.M.); (Á.C.)
- Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario San Cecilio de Granada, 18016 Granada, Spain
- Correspondence: ; Tel.: +34-958023199
| |
Collapse
|
2
|
Zhou Y, Li X, Ye M. Morusin inhibits the growth of human colorectal cancer HCT116‑derived sphere‑forming cells via the inactivation of Akt pathway. Int J Mol Med 2021; 47:1. [PMID: 33576447 PMCID: PMC7891835 DOI: 10.3892/ijmm.2021.4884] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 11/05/2020] [Indexed: 12/20/2022] Open
Abstract
The existence of colorectal cancer stem-like cells (CSC) is responsible for the failure of current treatments against colorectal cancer. Therefore, novel therapies need be developed to target CSCs. Some natural agents, including morusin have been proposed as possible candidates for this purpose. Morusin has been shown to exert antitumor effects. In the present study, it is demonstrated that morusin exerts antitumor effects on colorectal CSCs (CCSCs). The viability of human CCSCs was enhanced when the CCSCs formed spheroids in a serum-free and non-adhesive floating culture system. HCT116 sphere cells exhibited an increased proliferative capacity and a higher expression of stemness markers [octamer-binding transcription factor 4 (Oct4), Sox2 and Nanog]. Morusin inhibited the development of cancer spheroids and suppressed the growth of sphere cells via the induction of cell cycle arrest. Similarly, morusin decreased the expression levels of the stemness markers, Nanog and Oct4. The data partially revealed the molecular mechanisms involved: β-catenin signaling maintains the growth of CSCs and directly modulates the expression of Nanog and Oct4. Morusin suppressed the activity of β-catenin signaling via the inactivation of Akt; the executive β-catenin/TCF4 complex and the downstream targets, c-Myc, survivin and cyclin D1, were also downregulated. Moreover, the morusin-induced inactivation of Akt also increased the expression of p21Cip1/WAF1 and p27Kip, which can block the cell cycle by interacting with cyclin-dependent kinase (CDK) complexes. On the whole, the present study demonstrates that morusin inhibited the growth of colorectal cancer sphere cells, which were enriched with CCSCs via the inactivation of the Akt pathway.
Collapse
Affiliation(s)
- Yuqi Zhou
- Department of Hematology and Oncology, 904 Hospital of PLA Joint Logistic Support Force, Wuxi, Jiangsu 214000, P.R. China
| | - Xiangyong Li
- Department of Hematology and Oncology, 904 Hospital of PLA Joint Logistic Support Force, Wuxi, Jiangsu 214000, P.R. China
| | - Min Ye
- Department of Traditional Chinese Medicine, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
3
|
Pérez-Moreno P, Indo S, Niechi I, Huerta H, Cabello P, Jara L, Aguayo F, Varas-Godoy M, Burzio VA, Tapia JC. Endothelin-converting enzyme-1c promotes stem cell traits and aggressiveness in colorectal cancer cells. Mol Oncol 2019; 14:347-362. [PMID: 31788944 PMCID: PMC6998658 DOI: 10.1002/1878-0261.12609] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 11/07/2019] [Accepted: 11/29/2019] [Indexed: 12/21/2022] Open
Abstract
Endothelin-1 is a mitogenic peptide that activates several proliferation, survival, and invasiveness pathways. The effects of endothelin-1 rely on its activation by endothelin-converting enzyme-1 (ECE1), which is expressed as four isoforms with different cytoplasmic N termini. Recently, isoform ECE1c has been suggested to have a role in cancer aggressiveness. The N terminus of ECE1c is phosphorylated by protein kinase CK2 (also known as casein kinase 2), and this enhances its stability and promotes invasiveness in colorectal cancer cells. However, it is not known how phosphorylation improves stability and why this is correlated with increased aggressiveness. We hypothesized that CK2 phosphorylation protects ECE1c from N-terminal ubiquitination and, consequently, from proteasomal degradation. Here, we show that lysine 6 is the bona fide residue involved in ubiquitination of ECE1c and its mutation to arginine (ECE1cK6R ) significantly impairs proteasomal degradation, thereby augmenting ECE1c stability, even in the presence of the CK2 inhibitor silmitasertib. Furthermore, colorectal cancer cells overexpressing ECE1cK6R displayed enhanced cancer stem cell (CSC) traits, including increased stemness gene expression, chemoresistance, self-renewal, and colony formation and spheroid formation in vitro, as well as enhanced tumor growth and metastasis in vivo. These findings suggest that CK2-dependent phosphorylation enhances ECE1c stability, promoting an increase in CSC-like traits. Therefore, phospho-ECE1c may be a biomarker of poor prognosis and a potential therapeutic target for colorectal cancer.
Collapse
Affiliation(s)
- Pablo Pérez-Moreno
- Departamento de Oncología Básico Clínica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Sebastián Indo
- Departamento de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Ignacio Niechi
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Hernán Huerta
- Departamento de Oncología Básico Clínica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Pablo Cabello
- Departamento de Oncología Básico Clínica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Lilian Jara
- Programa de Genética Humana, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Francisco Aguayo
- Departamento de Oncología Básico Clínica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Manuel Varas-Godoy
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Verónica A Burzio
- Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile.,Fundación Ciencia & Vida, Andes Biotechnologies SpA, Santiago, Chile
| | - Julio C Tapia
- Departamento de Oncología Básico Clínica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
4
|
Wang X, Ghareeb WM, Zhang Y, Yu Q, Lu X, Huang Y, Huang S, Sun Y, Chi P. Hypermethylated and downregulated MEIS2 are involved in stemness properties and oxaliplatin-based chemotherapy resistance of colorectal cancer. J Cell Physiol 2019; 234:18180-18191. [PMID: 30859572 DOI: 10.1002/jcp.28451] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 01/26/2019] [Accepted: 01/30/2019] [Indexed: 12/16/2022]
Abstract
The resistance against oxaliplatin (L-OHP) based regimens remains a major obstacle for its efficient usage in treating metastatic colorectal cancer (mCRC). In this study, we performed weighted gene coexpression network analysis (WGCNA) to systematically screen the relevant hub genes for L-OHP resistance using the raw microarray data of 30 consecutive mCRC samples from our earlier study (GSE69657). The results were further confirmed through datasets from Gene Expression Omnibus (GEO). From L-OHP resistance module, nine genes in both the coexpression and protein-protein interaction networks were chosen as hub genes. Among these genes, Meis Homeobox 2 (MEIS2) had the highest correlation with L-OHP resistance (r = -0.443) and was deregulated in L-OHP resistant tissues compared with L-OHP sensitive tissues in both our own dataset and GSE104645 testing dataset. The receiver operating characteristic curve validated that MEIS2 had a good ability in predicting L-OHP response in both our own dataset (area under the curve [AUC] = 0.802) and GSE104645 dataset (AUC = 0.746). Then, the down expression of MEIS2 was observed in CRC tissue compared with normal tissue in 12 GEO-sourced datasets and The Cancer Genome Atlas (TCGA) and was correlated with poor event-free survival. Furthermore, analyzing methylation data from TCGA showed that MEIS2 had increased promoter hypermethylation. In addition, MEIS2 expression was significantly decreased in CRC stem cells compared with nonstem cells in two GEO datasets (GSE14773 and GSE24747). Further methylation analysis from GSE104271 demonstrated that CRC stem cells had higher MEIS2 promoter methylation levels in cg00366722 and cg00610348 sites. Gene set enrichment analysis showed that MEIS2 might be involved in the Wnt/β-catenin pathway. In the overall view, MEIS2 had increased promoter hypermethylation and was downregulated in poor L-OHP response mCRC tissues. MEIS2 might be involved in the Wnt/β-catenin pathway to maintain CRC stemness, which leads to L-OHP resistance.
Collapse
Affiliation(s)
- Xiaojie Wang
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Waleed M Ghareeb
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of General and Gastrointestinal Surgery, Suez Canal University, Ismailia, Egypt
| | - Yiyi Zhang
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Qian Yu
- Department of Pathology, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xingrong Lu
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Ying Huang
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Shenghui Huang
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Yanwu Sun
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Pan Chi
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
5
|
Kazemi Sefat NA, Mohammadi MM, Hadjati J, Talebi S, Ajami M, Daneshvar H. Sodium Butyrate as a Histone Deacetylase Inhibitor Affects Toll-Like Receptor 4 Expression in Colorectal Cancer Cell Lines. Immunol Invest 2019; 48:759-769. [PMID: 31117848 DOI: 10.1080/08820139.2019.1595643] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We assessed the effect of sodium butyrate (SB) as a histone deacetylase inhibitor (HDACi) on Toll-like receptor 4 (TLR4) gene expression levels, in low TLR4 expressing (HCT116) and high TLR4 expressing (SW480) colorectal cancer cells. The cytotoxic effect of SB was assessed by culturing SW480 and HCT116 cell lines using a broad spectrum of times and concentrations of SB. The MTT assay was done to check the cytotoxic properties of different SB concentrations. Gene expression levels of TLR4 was then evaluated for non-cytotoxic SB concentrations. Morphological analysis and MTT assay confirmed that SB concentrations equal to or less than 5mM were not cytotoxic for both cell lines. At 5mM concentration of SB in SW480 cell line and 1mM concentration of SB in HCT116 cell line, TLR4 gene expression level significantly increased from 24 to 48 hrs and decreased significantly from 48 to 72 hrs with an "early increased and late decreased pattern". At 1mM concentration of SB in SW480 cell line and 5mM concentration of SB in HCT116 cell line, TLR4 expression had a "gradually increased pattern". This study focuses on the dose-time-effect of SB in the pathogenesis of colorectal cancer. SB alters the expression level of TLR4 in colorectal cancer cells. This effect may depend on the cell type, treatment duration and SB concentration. The alterations in TLR4 expression may be due to the direct effect of SB on TLR4 and/or the expression changes of in other genes which may indirectly affect the TLR4 expression. Abbreviations: TLR4: Toll-like receptor 4; HDACi: histone deacetylase inhibitor; SB: sodium Butyrate; CRC: colorectal cancer; SCFA: short-chain fatty acid; hrs: hours.
Collapse
Affiliation(s)
- Nazanin Atieh Kazemi Sefat
- Department of Medical Immunology, Faculty of Medicine, Kerman University of Medical Sciences (KMU) , Kerman , Iran.,Department of Medical Immunology, Faculty of Medical Sciences, Tarbiat Modares University (TMU) , Tehran , Iran
| | - Mohammad Mahdi Mohammadi
- Department of Medical Immunology, Faculty of Medicine, Kerman University of Medical Sciences (KMU) , Kerman , Iran.,Kerman Physiology Research Center (KPRC), Kerman University of Medical sciences (KMU) , Kerman , Iran
| | - Jamshid Hadjati
- Department of Medical Immunology, Faculty of Medicine, Tehran University of Medical Sciences (TUMS) , Tehran , Iran
| | - Saeed Talebi
- Department of Medical Genetics and Molecular biology, Iran University of Medical Sciences (IUMS) , Tehran , Iran
| | - Maryam Ajami
- Department of Medical Immunology, Faculty of Medical Sciences, Tarbiat Modares University (TMU) , Tehran , Iran
| | - Hamid Daneshvar
- Department of Medical Immunology, Faculty of Medicine, Kerman University of Medical Sciences (KMU) , Kerman , Iran.,Kerman Physiology Research Center (KPRC), Kerman University of Medical sciences (KMU) , Kerman , Iran
| |
Collapse
|
6
|
Bellamkonda K, Satapathy SR, Douglas D, Chandrashekar N, Selvanesan BC, Liu M, Savari S, Jonsson G, Sjölander A. Montelukast, a CysLT1 receptor antagonist, reduces colon cancer stemness and tumor burden in a mouse xenograft model of human colon cancer. Cancer Lett 2018; 437:13-24. [PMID: 30144515 DOI: 10.1016/j.canlet.2018.08.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 08/13/2018] [Accepted: 08/16/2018] [Indexed: 12/16/2022]
Abstract
Inflammation is implicated in the etiology of sporadic colon cancer (CC), which is one of the leading causes of cancer-related deaths worldwide. Here, we report that inhibition of the inflammatory receptor CysLT1 through its antagonist, montelukast, is beneficial in minimizing stemness in CC and thereby minimizing tumor growth in a mouse xenograft model of human colon cancer. Upon treatment with montelukast, colonospheres derived from HT-29 and SW-480 human colon cancer cells exhibited a significant phenotypic change coupled with the downregulation of mRNA and protein expression of cancer stem cell (CSC) markers ALDH1 and DCLK1. Moreover, montelukast reduced the size of HT-29 cell-derived tumors in mice. The reduction in tumor size was associated with decreased levels of ALDH1A1, DCLK1, BCL2 mRNA and macrophage infiltration into the tumor tissue. Interestingly, this treatment elevated levels of the tumor suppressor 15-PGDH while reducing COX-2 expression. Our data highlight the association of CysLT1R with CSCs and demonstrate that inhibition of CysLT1R could prove beneficial in minimizing CSC-induced tumor growth. This work advances the notion that targeting CSCs is a promising approach to improve outcomes in those afflicted with colon cancer.
Collapse
Affiliation(s)
- Kishan Bellamkonda
- Division of Cell and Experimental Pathology, Department of Translational Medicine, Lund University, Clinical Research Center, Skåne University Hospital, Malmo¨, Sweden
| | - Shakti Ranjan Satapathy
- Division of Cell and Experimental Pathology, Department of Translational Medicine, Lund University, Clinical Research Center, Skåne University Hospital, Malmo¨, Sweden
| | - Desiree Douglas
- Division of Cell and Experimental Pathology, Department of Translational Medicine, Lund University, Clinical Research Center, Skåne University Hospital, Malmo¨, Sweden
| | - Naveenkumar Chandrashekar
- Division of Cell and Experimental Pathology, Department of Translational Medicine, Lund University, Clinical Research Center, Skåne University Hospital, Malmo¨, Sweden
| | - Benson Chellakkan Selvanesan
- Division of Cell and Experimental Pathology, Department of Translational Medicine, Lund University, Clinical Research Center, Skåne University Hospital, Malmo¨, Sweden
| | - Minghui Liu
- Division of Cell and Experimental Pathology, Department of Translational Medicine, Lund University, Clinical Research Center, Skåne University Hospital, Malmo¨, Sweden
| | - Sayeh Savari
- Division of Cell and Experimental Pathology, Department of Translational Medicine, Lund University, Clinical Research Center, Skåne University Hospital, Malmo¨, Sweden
| | - Gunilla Jonsson
- Division of Cell and Experimental Pathology, Department of Translational Medicine, Lund University, Clinical Research Center, Skåne University Hospital, Malmo¨, Sweden
| | - Anita Sjölander
- Division of Cell and Experimental Pathology, Department of Translational Medicine, Lund University, Clinical Research Center, Skåne University Hospital, Malmo¨, Sweden.
| |
Collapse
|
7
|
Szaryńska M, Olejniczak A, Kobiela J, Spychalski P, Kmieć Z. Therapeutic strategies against cancer stem cells in human colorectal cancer. Oncol Lett 2017; 14:7653-7668. [PMID: 29250169 PMCID: PMC5727596 DOI: 10.3892/ol.2017.7261] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 09/01/2017] [Indexed: 12/16/2022] Open
Abstract
Colorectal cancer (CRC) is the third most frequent malignancy and represents the fourth most common cause of cancer-associated mortalities in the world. Despite many advances in the treatment of CRC, the 5-year survival rate of patients with CRC remains unsatisfactory due to tumor recurrence and metastases. Recently, cancer stem cells (CSCs), have been suggested to be responsible for the initiation and relapse of the disease, and have been identified in CRC. Due to their basic biological features, which include self-renewal and pluripotency, CSCs may be novel therapeutic targets for CRC and other cancer types. Conventional therapeutics only act on proliferating and mature cancer cells, while quiescent CSCs survive and often become resistant to chemotherapy. In this review, markers of CRC-CSCs are evaluated and the recently introduced experimental therapies that specifically target these cells by inducing CSC proliferation, differentiation and sensitization to apoptotic signals via molecules including Dickkopf-1, bone morphogenetic protein 4, Kindlin-1, tankyrases, and p21-activated kinase 1, are discussed. In addition, novel strategies aimed at inhibiting some crucial processes engaged in cancer progression regulated by the Wnt, transforming growth factor β and Notch signaling pathways (pyrvinium pamoate, silibinin, PRI-724, P17, and P144 peptides) are also evaluated. Although the metabolic alterations in cancer were first described decades ago, it is only recently that the concept of targeting key regulatory molecules of cell metabolism, such as sirtuin 1 (miR-34a) and AMPK (metformin), has emerged. In conclusion, the discovery of CSCs has resulted in the definition of novel therapeutic targets and the development of novel experimental therapies for CRC. However, further investigations are required in order to apply these novel drugs in human CRC.
Collapse
Affiliation(s)
- Magdalena Szaryńska
- Department of Histology, Medical University of Gdańsk, 80-210 Gdańsk; Gdańsk, Poland
| | - Agata Olejniczak
- Department of Histology, Medical University of Gdańsk, 80-210 Gdańsk; Gdańsk, Poland
| | - Jarosław Kobiela
- Department of General, Endocrine and Transplant Surgery, Invasive Medicine Center, Medical University of Gdańsk, 80-214 Gdańsk, Poland
| | - Piotr Spychalski
- Department of General, Endocrine and Transplant Surgery, Invasive Medicine Center, Medical University of Gdańsk, 80-214 Gdańsk, Poland
| | - Zbigniew Kmieć
- Department of Histology, Medical University of Gdańsk, 80-210 Gdańsk; Gdańsk, Poland
| |
Collapse
|
8
|
Blocking endothelin-1-receptor/β-catenin circuit sensitizes to chemotherapy in colorectal cancer. Cell Death Differ 2017; 24:1811-1820. [PMID: 28708138 PMCID: PMC5596423 DOI: 10.1038/cdd.2017.121] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/09/2017] [Accepted: 06/23/2017] [Indexed: 12/16/2022] Open
Abstract
The limited clinical response to conventional chemotherapeutics observed in colorectal cancer (CRC) may be related to the connections between the hyperactivated β-catenin signaling and other pathways in CRC stem-like cells (CRC-SC). Here, we show the mechanistic link between the endothelin-1 (ET-1)/ET-1 receptor (ET-1R) signaling and β-catenin pathway through the specific interaction with the signal transducer β-arrestin1 (β-arr1), which initiates signaling cascades as part of the signaling complex. Using a panel of patient-derived CRC-SC, we show that these cells secrete ET-1 and express ETAR and β-arr1, and that the activation of ETAR/β-arr1 axis promotes the cross-talk with β-catenin signaling to sustain stemness, epithelial-to-mesenchymal transition (EMT) phenotype and response to chemotherapy. Upon ETAR activation, β-arr1 acts as a transcription co-activator that binds β-catenin, thereby promoting nuclear complex with β-catenin/TFC4 and p300 and histone acetylation, inducing chromatin reorganization on target genes, such as ET-1. The enhanced transcription of ET-1 increases the self-sustained ET-1/β-catenin network. All these findings provide a strong rationale for targeting ET-1R to hamper downstream β-catenin/ET-1 autocrine circuit. Interestingly, treatment with macitentan, a dual ETAR and ETBR antagonist, able to interfere with tumor and microenvironment, disrupts the ET-1R/β-arr1-β-catenin interaction impairing pathways involved in cell survival, EMT, invasion, and enhancing sensitivity to oxaliplatin (OX) and 5-fluorouracil (5-FU). In CRC-SC xenografts, the combination of macitentan and OX or 5-FU enhances the therapeutic effects of cytotoxic drugs. Together, these results provide mechanistic insight into how ET-1R coopts β-catenin signaling and offer a novel therapeutic strategy to manage CRC based on the combination of macitentan and chemotherapy that might benefit patients whose tumors show high ETAR and β-catenin expression.
Collapse
|
9
|
Gharib AF, Shalaby SM, Raafat N, Fawzy WMS, Abdel Hakim NH. Assessment of neutralizing interleukin-4 effect on CD133 gene expression in colon cancer cell line. Cytokine 2017; 97:66-72. [PMID: 28578295 DOI: 10.1016/j.cyto.2017.05.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 05/20/2017] [Accepted: 05/24/2017] [Indexed: 12/14/2022]
Abstract
Colorectal cancer may be maintained by cancer stem-like cells (CSCs) that express the cell surface marker CD133. CSCs (CD133+cells) exhibits greater resistance to the chemotherapy and this resistance may be mediated in part by an autocrine response to IL4. The aim of the study was to assess the effect of anti-IL4 antibody alone or in combination with chemotherapy on the CD133 expression andthe tumor growth. We used Caco cell line in our experiments and the samples were as the following; untreated colorectal cell line, cells treated by chemotherapy, cells treated by anti-IL4 antibody in 3doses (2.5, 5, 10μg/ml), cells treatedby combination of chemotherapy and anti-IL4 antibody in 3 doses. Results of our in vitro studies demonstrated that anti-IL4 inhibited growth of Caco cell line in a dose-dependent manner revealing a 32.11% inhibition at the highest concentration (10µg/ml). There was further significant inhibition by combination of anti IL4 and chemotherapy in a dose response manner when compared to group treated by chemotherapy only. These effects were associated with decreased expression of CD133 in tumor cells also. Lastly, anti-IL4 antibody stimulated apoptosis. Our study suggested that neutralizing of IL4 by anti IL4 antibody affect the CD133+ cells may be by increasing their apoptosis. The effects of anti IL4 antibody either, alone or in combination with chemotherapy, inhibited the tumor growth and decreased the viable tumor cells. Furthermore, neutralizing of IL4 increased the efficacy of chemotherapy treatment.
Collapse
Affiliation(s)
- Amal F Gharib
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Sally M Shalaby
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Nermin Raafat
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Walaa M S Fawzy
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Nabila H Abdel Hakim
- Clinical Oncology and Nuclear Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
10
|
Puglisi MA, Cenciarelli C, Tesori V, Cappellari M, Martini M, Di Francesco AM, Giorda E, Carsetti R, Ricci-Vitiani L, Gasbarrini A. High nitric oxide production, secondary to inducible nitric oxide synthase expression, is essential for regulation of the tumour-initiating properties of colon cancer stem cells. J Pathol 2015; 236:479-490. [PMID: 25875314 DOI: 10.1002/path.4545] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 03/19/2015] [Accepted: 04/11/2015] [Indexed: 01/06/2023]
Abstract
Chronic inflammation is a leading cause of neoplastic transformation in many human cancers and especially in colon cancer (CC), in part due to tumour promotion by nitric oxide (NO) generated at inflammatory sites. It has also been suggested that high NO synthesis, secondary to inducible NO synthase (iNOS) expression, is a distinctive feature of cancer stem cells (CSCs), a small subset of tumour cells with self-renewal capacity. In this study we explored the contribution of NO to the development of colon CSC features and evaluated potential strategies to treat CC by modulating NO production. Our data show an integral role for endogenous NO and iNOS activity in the biology of colon CSCs. Indeed, colon CSCs with high endogenous NO production (NO(high)) displayed higher tumourigenic abilities than NO(low) fractions. The blockade of endogenous NO availability, using either a specific iNOS inhibitor or a genetic knock-down of iNOS, resulted in a significant reduction of colon CSC tumourigenic capacities in vitro and in vivo. Interestingly, analysis of genes altered by iNOS-directed shRNA showed that the knockdown of iNOS expression was associated with a significant down-regulation of signalling pathways involved in stemness and tumour progression in colon CSCs. These findings confirm that endogenous NO plays an important role in defining the stemness properties of colon CSCs through cross-regulation of several cellular signalling pathways. This discovery could shed light on the mechanisms by which NO induces the growth and invasiveness of CC, providing new insights into the link between inflammation and colon tumourigenesis.
Collapse
Affiliation(s)
| | - Carlo Cenciarelli
- Institute of Translational Pharmacology, National Research Council of Italy, Rome, Italy
| | - Valentina Tesori
- Department of Internal Medicine and Gastroenterology, Gemelli Hospital, Rome, Italy
| | - Marianna Cappellari
- Department of Haematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Maurizio Martini
- Department of Anatomical Pathology, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Ezio Giorda
- Cytofluorimetry Laboratory, Bambino Gesù Paediatric Hospital, Rome, Italy
| | - Rita Carsetti
- Cytofluorimetry Laboratory, Bambino Gesù Paediatric Hospital, Rome, Italy
| | - Lucia Ricci-Vitiani
- Department of Haematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Antonio Gasbarrini
- Department of Internal Medicine and Gastroenterology, Gemelli Hospital, Rome, Italy
| |
Collapse
|
11
|
Fukuda N, Tsuchikawa T, Fukunaga A, Kawase H, Homma N, Nakamura T, Shichinohe T, Hirano S. Validation of histological diagnostic methods for detecting endothelin B receptor expression. Oncol Rep 2014; 31:1561-6. [PMID: 24549269 DOI: 10.3892/or.2014.3031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 12/09/2013] [Indexed: 11/05/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has an extremely poor prognosis. Recently, it was reported that the endothelin B receptor (ETBR) of tumor endothelial cells prevents antitumor immunity. However, the immuno-histochemistry (IHC) conditions required to detect ETBR expression remain unclear. The aim of the present study was to confirm the appropriate conditions for IHC for ETBR using ETBR cDNA and transfectant cells and to assess ETBR expression in PDAC patients. An ETBR-expressing cell was established as an objective positive control and the detectability of ETBR expression was evaluated using several types of anti-ETBR antibodies. ETBR mRNA expression was then studied. Finally, ETBR expression was examined in human PDAC tissue using IHC. As a result, four different anti-ETBR antibodies recognized the cell surface ETBR appropriately. A non-specific reaction was shown in the detection of ETBR in normal human tissues. ETBR mRNA expression was weakly detected only in the adrenal gland. No biologically significant correlation was observed in the ETBR-IHC of human PDAC sections. In conclusion, it is necessary to perform IHC using an appropriate control to assess the tissue expression of ETBR.
Collapse
Affiliation(s)
- Naoya Fukuda
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Takahiro Tsuchikawa
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Akira Fukunaga
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hiroshi Kawase
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Naotake Homma
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Toru Nakamura
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Toshiaki Shichinohe
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Satoshi Hirano
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
12
|
Spinella F, Caprara V, Cianfrocca R, Rosanò L, Di Castro V, Garrafa E, Natali PG, Bagnato A. The interplay between hypoxia, endothelial and melanoma cells regulates vascularization and cell motility through endothelin-1 and vascular endothelial growth factor. Carcinogenesis 2014; 35:840-8. [PMID: 24473118 DOI: 10.1093/carcin/bgu018] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Reciprocal growth factor exchanges between endothelial and malignant cells within the hypoxic microenvironment determine tumor progression. However, the nature of these exchanges has not yet been fully explored. We studied the mutual regulation between endothelial cells (EC), melanoma cells and hypoxia that dictate tumor aggressiveness and angiogenic activity. Here, we investigated the presence of bidirectional autocrine/paracrine endothelin (ET)-1/ET receptor (ETBR) signaling in melanoma cells, blood and lymphatic EC. In all these cells, hypoxia enhanced ET-1 expression, which in turn induced vascular endothelial growth factor (VEGF)-A and VEGF-C secretion, through the hypoxia-inducible growth factor (HIF)-1α and HIF-2α. Autocrine/paracrine exchanges of ET-1, VEGF-A and VEGF-C promoted tumor aggressiveness and morphological changes in blood and lymphatic EC. Furthermore, conditioned media from EC enhanced melanoma cell migration and vessel-like channel formation. This regulation was inhibited by ETBR blockade, by using the selective ETBR antagonist, or ETBR small interfering RNA (siRNA), and by VEGFR-2/-3 antibodies, indicating that ET-1, VEGF-A/VEGF-C, produced by melanoma cells or EC mediated inter-regulation between these cells. Interestingly, HIF-1α/HIF-2α siRNA, impaired this reciprocal regulation, demonstrating the key role of these transcriptional factors in signaling exchanges. In melanoma xenografts, the ETBR antagonist reduced tumor growth and the number of blood and lymphatic vessels. These results reveal an interplay between melanoma cells and EC mediated by ET-1 and VEGF-A/-C and coordinated by the hypoxic microenvironment through HIF-1α/2α transcriptional programs. Thus, targeting ETBR may improve melanoma treatment for tumor and EC, by inhibiting autocrine/paracrine signaling that sustains melanoma progression.
Collapse
Affiliation(s)
- Francesca Spinella
- Experimental Oncology Department, Regina Elena National Cancer Institute, 00144 Rome
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Tumor initiating cells and chemoresistance: which is the best strategy to target colon cancer stem cells? BIOMED RESEARCH INTERNATIONAL 2014; 2014:859871. [PMID: 24527460 PMCID: PMC3914574 DOI: 10.1155/2014/859871] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 10/24/2013] [Indexed: 12/12/2022]
Abstract
There is an emerging body of evidence that chemoresistance and minimal residual disease result from selective resistance of a cell subpopulation from the original tumor that is molecularly and phenotypically distinct. These cells are called “cancer stem cells” (CSCs). In this review, we analyze the potential targeting strategies for eradicating CSCs specifically in order to develop more effective therapeutic strategies for metastatic colon cancer. These include induction of terminal epithelial differentiation of CSCs or targeting some genes expressed only in CSCs and involved in self-renewal and chemoresistance. Ideal targets could be cell regulators that simultaneously control the stemness and the resistance of CSCs. Another important aspect of cancer biology, which can also be harnessed to create novel broad-spectrum anticancer agents, is the Warburg effect, also known as aerobic glycolysis. Actually, little is yet known with regard to the metabolism of CSCs population, leaving an exciting unstudied avenue in the dawn of the emerging field of metabolomics.
Collapse
|
14
|
Rosanò L, Spinella F, Bagnato A. Endothelin 1 in cancer: biological implications and therapeutic opportunities. Nat Rev Cancer 2013; 13:637-51. [PMID: 23884378 DOI: 10.1038/nrc3546] [Citation(s) in RCA: 233] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Activation of autocrine and paracrine signalling by endothelin 1 (ET1) binding to its receptors elicits pleiotropic effects on tumour cells and on the host microenvironment. This activation modulates cell proliferation, apoptosis, migration, epithelial-to-mesenchymal transition, chemoresistance and neovascularization, thus providing a strong rationale for targeting ET1 receptors in cancer. In this Review, we discuss the advances in our understanding of the diverse biological roles of ET1 in cancer and describe the latest preclinical and clinical progress that has been made using small-molecule antagonists of ET1 receptors that inhibit ET1-driven signalling.
Collapse
Affiliation(s)
- Laura Rosanò
- Laboratory of Molecular Pathology A, Regina Elena National Cancer Institute, Via Elio Chianesi 53, Rome 00144, Italy
| | | | | |
Collapse
|
15
|
Han Y, Cai H, Ma L, Ding Y, Tan X, Liu Y, Su T, Yu Y, Chang W, Zhang H, Fu C, Cao G. Nuclear orphan receptor NR4A2 confers chemoresistance and predicts unfavorable prognosis of colorectal carcinoma patients who received postoperative chemotherapy. Eur J Cancer 2013; 49:3420-30. [PMID: 23809767 DOI: 10.1016/j.ejca.2013.06.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 05/25/2013] [Accepted: 06/05/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND NR4A2, an orphan nuclear receptor essential in neuron generation, has been recently linked to inflammatory and metabolic pathways of colorectal carcinoma (CRC). However, the effects of NR4A2 on chemo-resistance and postoperative prognosis of CRC remain unknown. METHODS NR4A2 was transfected into CRC cells to investigate its effects on chemo-resistance to 5-fluorouracil and oxaliplatin and chemotherapeutics-induced apoptosis. We also investigated prostaglandin E2 (PGE2)-induced NR4A2 expression and its effect on chemo-resistance. Tissue microarrays including 51 adenoma, 14 familial adenomatous polyposis with CRC, 17 stage IV CRC with adjacent mucosa and 682 stage I-III CRC specimens were examined immunohistochemically for NR4A2 expression. Median follow-up time for stage I-III CRC patients was 53 months. RESULTS Ectopic expression of NR4A2 increased the chemo-resistance, and attenuated the chemotherapeutics-induced apoptosis. Transient treatment of PGE2 significantly up-regulated NR4A2 expression via protein kinase A pathway and increased the chemo-resistance. NR4A2 expression in epithelials consecutively increased from adenoma, adjacent mucosa to CRC (P(trend)<0.001). In multivariate Cox regression analyses, high NR4A2 expression in cancer nuclei (immunoreactive score ≥ 4) significantly predicted a shorter disease-specific survival (DSS) of CRC patients (hazard ratio [HR]=1.88, P=0.024). High NR4A2 expression specifically predicted a shorter DSS of colon cancer patients (dichotomisation, HR=2.55, log-rank test P=0.011), especially for those who received postoperative 5-fluorouracil/leucovorin plus oxaliplatin (FOLFOX) chemotherapy (3-score range, HR=1.86, log-rank test P=0.020). CONCLUSION High expression of NR4A2 in CRC cells confers chemo-resistance, attenuates chemotherapeutics-induced apoptosis, and predicts unfavorable prognosis of colon cancer patients, especially for those who received postoperative chemotherapy. NR4A2 may be prognostic and predictive for colon cancer.
Collapse
Affiliation(s)
- Yifang Han
- Department of Epidemiology, Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Puglisi MA, Tesori V, Lattanzi W, Gasbarrini GB, Gasbarrini A. Colon cancer stem cells: Controversies and perspectives. World J Gastroenterol 2013; 19:2997-3006. [PMID: 23716979 PMCID: PMC3662939 DOI: 10.3748/wjg.v19.i20.2997] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Revised: 03/25/2013] [Accepted: 04/04/2013] [Indexed: 02/06/2023] Open
Abstract
Tumors have long been viewed as a population in which all cells have the equal propensity to form new tumors, the so called conventional stochastic model. The cutting-edge theory on tumor origin and progression, tends to consider cancer as a stem cell disease. Stem cells are actively involved in the onset and maintenance of colon cancer. This review is intended to examine the state of the art on colon cancer stem cells (CSCs), with regard to the recent achievements of basic research and to the corresponding translational consequences. Specific prominence is given to the hypothesized origin of CSCs and to the methods for their identification. The growing understanding of CSC biology is driving the optimization of novel anti-cancer targeted drugs.
Collapse
|
17
|
Hayashi S, Tanaka J, Okada S, Isobe T, Yamamoto G, Yasuhara R, Irie T, Akiyama C, Kohno Y, Tachikawa T, Mishima K. Lin28a is a putative factor in regulating cancer stem cell-like properties in side population cells of oral squamous cell carcinoma. Exp Cell Res 2013; 319:1220-8. [PMID: 23500413 DOI: 10.1016/j.yexcr.2013.03.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 02/24/2013] [Accepted: 03/02/2013] [Indexed: 01/06/2023]
Abstract
Cancer stem cells (CSCs) are among the target cells of cancer therapy because they are uniquely involved in both cancer progression and sensitivity to chemotherapeutic agents. We identified side population (SP) cells, which are known to be an enriched population of CSC, in five oral squamous cell carcinoma (OSCC) cells (SCC9, SCC25, TOSCC7, TOSCC17, and TOSCC23). The percentages of SP cells ranged from 0% to 3.3%, with TOSCC23 cells showing the highest percentages of SP cells (3.3% of the total cell population). The SP cells isolated from TOSCC23 cells also showed greater cell proliferation and invasion compared to non-SP (MP) cells. Therefore, our initial findings suggested that SP cells were enriched for CSC-like cells. Furthermore, DNA microarray analysis revealed that the expression of cell proliferation-related and anti-apoptotic genes was greater in SP cells compared to MP cells. We focused on Lin28a, which showed the highest expression (approximately 22-fold) among the upregulated genes. The overexpression of Lin28a in TOSCC23 cells increased their proliferation, colony formation, and invasion. These findings suggest that Lin28a is an appropriate CSC target molecule for OSCC treatment.
Collapse
Affiliation(s)
- S Hayashi
- Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Han YF, Cao GW. Role of nuclear receptor NR4A2 in gastrointestinal inflammation and cancers. World J Gastroenterol 2012; 18:6865-73. [PMID: 23322982 PMCID: PMC3531668 DOI: 10.3748/wjg.v18.i47.6865] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 08/27/2012] [Accepted: 09/12/2012] [Indexed: 02/06/2023] Open
Abstract
NR4A2 is a transcription factor belonging to the steroid orphan nuclear receptor superfamily. It was originally considered to be essential in the generation and maintenance of dopaminergic neurons, and associated with neurological disorders such as Parkinson’s disease. Recently, NR4A2 has been found to play a critical role in some inflammatory diseases and cancer. NR4A2 can be efficiently trans-activated by some proinflammatory cytokines, such as tumor necrosis factor-α, interleukin-1β, and vascular endothelial growth factor (VEGF). The nuclear factor-κB signaling pathway serves as a principal regulator of inducible NR4A expression in immune cells. NR4A2 can trans-activate Foxp3, a hallmark specifically expressed in regulatory T (Treg) cells, and plays a critical role in the differentiation, maintenance, and function of Treg cells. NR4A2 in T lymphocytes is pivotal for Treg cell induction and suppression of aberrant induction of Th1 under physiological and pathological conditions. High density of Foxp3+ Treg cells is significantly associated with gastrointestinal inflammation, tumor immune escape, and disease progression. NR4A2 is produced at high levels in CD133+ colorectal carcinoma (CRC) cells and significantly upregulated by cyclooxygenase-2-derived prostaglandin E2 in a cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA)-dependent manner in CRC cells. The cAMP/PKA signaling pathway is the common pathway of NR4A2-related inflammation and cancer. NR4A2 trans-activates osteopontin, a direct target of the Wnt/β-catenin pathway associated with CRC invasion, metastasis, and poor prognosis. Knockdown of endogenous NR4A2 expression attenuates VEGF-induced endothelial cell proliferation, migration and in vivo angiogenesis. Taken together, NR4A2 emerges as an important nuclear factor linking gastrointestinal inflammation and cancer, especially CRC, and should serve as a candidate therapeutic target for inflammation-related gastrointestinal cancer.
Collapse
|
19
|
Abstract
The monoclonal antibody against the AC133 epitope of CD133 has been widely used as a cell surface marker of cancer stem cells in several different cancer types. Here, we describe the isolation and characterisation of two RNA aptamers, including the smallest described 15 nucleotide RNA aptamer, which specifically recognise the AC133 epitope and the CD133 protein with high sensitivity. As well, both these aptamers show superior tumour penetration and retention when compared to the AC133 antibody in a 3-D tumour sphere model. These novel CD133 aptamers will aid future development of cancer stem cell targeted therapeutics and molecular imaging.
Collapse
|
20
|
CD133 affects the invasive ability of HCT116 cells by regulating TIMP-2. THE AMERICAN JOURNAL OF PATHOLOGY 2012. [PMID: 23195431 DOI: 10.1016/j.ajpath.2012.10.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
CD133 is widely expressed in colorectal cancer (CRC) tissues and cell lines. This protein has been used as a marker of CRC cancer stem cells, although the function and mechanism of CD133 in CRC invasion and metastasis remain unclear. In our study, we examined the role of CD133 in CRC invasion in vitro and investigated the mechanism involved in CD133-related invasion. CD133(high) and CD133(low) HCT116 cells were isolated, and the proliferation and invasive ability of these two subpopulations were tested. CD133(high) HCT116 cells exhibited greater proliferation and invasion compared with CD133(low) HCT116 cells. CD133 knockdown (using CD133 small-interfering [si]RNA) inhibited the invasive activity of CD133si-HCT116 cells. For the first time, we found that the expression of tissue inhibitor of matrix metalloproteinases-2 (TIMP-2) was down-regulated in CD133si-HCT116 cells. In addition, for the TIMP-2si-HCT116 cells (transfected with TIMP-2 siRNA), in vitro invasion was significantly decreased, whereas the expression of CD133 remained unchanged. Finally, the metalloproteinase 2 and MEK/ERK signaling pathways were examined, and no significant change was observed after the knockdown of CD133 or TIMP-2 in HCT116 cells. In conclusion, we demonstrated that CD133 plays an important role in HCT116 cell invasion, and for the first time, we found that CD133 knockdown significantly down-regulated TIMP-2 expression, which suggests that CD133 affects the invasive ability of HCT116 cells by regulating TIMP-2.
Collapse
|
21
|
Coco C, Zannoni GF, Caredda E, Sioletic S, Boninsegna A, Migaldi M, Rizzo G, Bonetti LR, Genovese G, Stigliano E, Cittadini A, Sgambato A. Increased expression of CD133 and reduced dystroglycan expression are strong predictors of poor outcome in colon cancer patients. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2012; 31:71. [PMID: 22964035 PMCID: PMC3541988 DOI: 10.1186/1756-9966-31-71] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 08/28/2012] [Indexed: 02/08/2023]
Abstract
Background Expression levels of CD133, a cancer stem cell marker, and of the α-subunit of the dystroglycan (α-DG) complex, have been previously reported to be altered in colorectal cancers. Methods Expression levels of CD133 and α-DG were assessed by immunohistochemistry in a series of colon cancers and their prognostic significance was evaluated. Results Scattered cells positive for CD133 were rarely detected at the bases of the crypts in normal colonic mucosa while in cancer cells the median percentage of positive cells was 5% (range 0–80). A significant correlation was observed with pT parameter and tumor stage but not with tumor grade and N status. Recurrence and death from disease were significantly more frequent in CD133-high expressing tumors and Kaplan-Meier curves showed a significant separation between high vs low expressor groups for both disease-free (p = 0.002) and overall (p = 0.008) survival. Expression of α-DG was reduced in a significant fraction of tumors but low α-DG staining did not correlate with any of the classical clinical-pathological parameters. Recurrence and death from the disease were significantly more frequent in α-DG-low expressing tumors and Kaplan-Meier curves showed a significant separation between high vs low expressor tumors for both disease-free (p = 0.02) and overall (p = 0.02) survival. Increased expression of CD133, but not loss of α-DG, confirmed to be an independent prognostic parameters at a multivariate analysis associated with an increased risk of recurrence (RR = 2.4; p = 0.002) and death (RR = 2.3; p = 0.003). Conclusions Loss of α-DG and increased CD133 expression are frequent events in human colon cancer and evaluation of CD133 expression could help to identify high-risk colon cancer patients.
Collapse
Affiliation(s)
- Claudio Coco
- Dipartimento di Scienze Chirurgiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|