101
|
Jagadeeshan S, Prasad M, Ortiz-Cuaran S, Gregoire V, Saintigny P, Elkabets M. Adaptive Responses to Monotherapy in Head and Neck Cancer: Interventions for Rationale-Based Therapeutic Combinations. Trends Cancer 2019; 5:365-390. [PMID: 31208698 DOI: 10.1016/j.trecan.2019.04.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/22/2019] [Accepted: 04/23/2019] [Indexed: 12/16/2022]
Abstract
Most Phase II and III clinical trials in head and neck cancer (HNC) combine two or more treatment modalities, which are based, in part, on knowledge of the molecular mechanisms of innate and acquired resistance to monotherapy. In this review, we describe the range of tumor-cell autonomously derived (intrinsic) and tumor-microenvironment-derived (extrinsic) acquired-resistance mechanisms to various FDA-approved monotherapies for HNC. Specifically, we describe how tumor cells and the tumor microenvironment (TME) respond to radiation, chemotherapy, targeted therapy (cetuximab), and immunotherapies [programmed cell death 1 (PD-1) inhibitors] and adapt to the selective pressure of these monotherapies. Due to the diversity of adaptive responses to monotherapy, monitoring the response to treatment in patients is critical to understand the path that leads to resistance and to guide the optimal therapeutic drug combinations in the clinical setting. We envisage that applying such a rationale-based therapeutic strategy will improve treatment efficacy in HNC patients.
Collapse
Affiliation(s)
- Sankar Jagadeeshan
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Manu Prasad
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Sandra Ortiz-Cuaran
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon 69008, France
| | - Vincent Gregoire
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon 69008, France; Department of Radiation Therapy, Centre Léon Bérard, Lyon 69008, France
| | - Pierre Saintigny
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon 69008, France; Department of Medical Oncology, Centre Léon Bérard, Lyon 69008, France
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.
| |
Collapse
|
102
|
Toole BP. The CD147-HYALURONAN Axis in Cancer. Anat Rec (Hoboken) 2019; 303:1573-1583. [PMID: 31090215 DOI: 10.1002/ar.24147] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 11/08/2018] [Accepted: 11/24/2018] [Indexed: 12/15/2022]
Abstract
CD147 (basigin; EMMPRIN), hyaluronan, and hyaluronan receptors (e.g., CD44) are intimately involved in several phenomena that underlie malignancy. A major avenue whereby they influence tumor progression is most likely their role in the characteristics of cancer stem cells (CSCs), subpopulations of tumor cells that exhibit chemoresistance, invasiveness, and potent tumorigenicity. Both CD147 and hyaluronan have been strongly implicated in chemoresistance and invasiveness, and may be drivers of CSC characteristics, since current evidence indicates that both are involved in epithelial-mesenchymal transition, a crucial process in the acquisition of CSC properties. Hyaluronan is a prominent constituent of the tumor microenvironment whose interactions with cell surface receptors influence several signaling pathways that lead to chemoresistance and invasiveness. CD147 is an integral plasma membrane glycoprotein of the Ig superfamily and cofactor in assembly and activity of monocarboxylate transporters (MCTs). CD147 stimulates hyaluronan synthesis and interaction of hyaluronan with its receptors, in particular CD44 and LYVE-1, which in turn result in activation of multiprotein complexes containing members of the membrane-type matrix metalloproteinase, receptor tyrosine kinase, ABC drug transporter, or MCT families within lipid raft domains. Multivalent hyaluronan-receptor interactions are essential for formation or stabilization of these lipid raft complexes and for downstream signaling pathways or transporter activities. We conclude that stimulation of hyaluronan-receptor interactions by CD147 and the consequent activities of these complexes may be critical to the properties of CSCs and their role in malignancy. Anat Rec, 2019. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Bryan P Toole
- Department of Regenerative Medicine & Cell Biology and Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
103
|
Hyaluronan-CD44 interaction promotes HPV 16 E6 oncogene-mediated oropharyngeal cell carcinoma survival and chemoresistance. Matrix Biol 2019; 78-79:180-200. [DOI: 10.1016/j.matbio.2018.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 07/30/2018] [Accepted: 07/31/2018] [Indexed: 12/12/2022]
|
104
|
BRM transcriptionally regulates miR-302a-3p to target SOCS5/STAT3 signaling axis to potentiate pancreatic cancer metastasis. Cancer Lett 2019; 449:215-225. [DOI: 10.1016/j.canlet.2019.02.031] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/03/2019] [Accepted: 02/14/2019] [Indexed: 12/11/2022]
|
105
|
Wu Y, Hu L, Qin Z, Wang X. MicroRNA‑302a upregulation mediates chemo‑resistance in prostate cancer cells. Mol Med Rep 2019; 19:4433-4440. [PMID: 30942454 DOI: 10.3892/mmr.2019.10098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 12/28/2018] [Indexed: 11/05/2022] Open
Abstract
MicroRNAs (miRNAs) are post‑transcriptional regulators that mediate the initiation and progression of human cancer. Growing evidence suggests that deregulation of miRNA expression levels underlies chemo‑resistance. To investigate whether miRNA‑302a (miR‑302a) is involved in mediating chemo‑resistance to paclitaxel in prostate cancer, a series of in vitro analyses were performed in paclitaxel‑resistant prostate cancer PC‑3PR cells and non‑resistant prostate cancer PC‑3 cells. It was demonstrated that the expression of miR‑302a was upregulated in PC‑3PR cells. Notably, ectopic expression of miR‑302a also increased resistance to paclitaxel in wild‑type PC‑3 cells. By contrast, silencing of miR‑302a in PC‑3PR cells sensitized the cells to paclitaxel. Gene and protein expression analyses suggested that the miR‑302a target gene breast cancer resistance protein (BCRP) may mediate chemo‑resistance to paclitaxel in PC‑3PR cells. In conclusion, the data suggested that elevated miR‑302a levels, in part, mediate sensitivity to paclitaxel in prostate cancer through the aberrant regulation of its downstream targets, AOF2, BCRP and permeability glycoprotein 1. These data have implications for the development of novel therapeutics in prostate cancer that may improve sensitivity to chemotherapeutics.
Collapse
Affiliation(s)
- Yuqi Wu
- Department of Urology, Carson International Cancer Centre, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy Centre, Shenzhen University, Shenzhen, Guangdong 518000, P.R. China
| | - Li Hu
- Department of Urology, Carson International Cancer Centre, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy Centre, Shenzhen University, Shenzhen, Guangdong 518000, P.R. China
| | - Zizhen Qin
- Department of Urology, Carson International Cancer Centre, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy Centre, Shenzhen University, Shenzhen, Guangdong 518000, P.R. China
| | - Xiangwei Wang
- Department of Urology, Carson International Cancer Centre, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy Centre, Shenzhen University, Shenzhen, Guangdong 518000, P.R. China
| |
Collapse
|
106
|
Jagust P, de Luxán-Delgado B, Parejo-Alonso B, Sancho P. Metabolism-Based Therapeutic Strategies Targeting Cancer Stem Cells. Front Pharmacol 2019; 10:203. [PMID: 30967773 PMCID: PMC6438930 DOI: 10.3389/fphar.2019.00203] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 02/18/2019] [Indexed: 02/02/2023] Open
Abstract
Cancer heterogeneity constitutes the major source of disease progression and therapy failure. Tumors comprise functionally diverse subpopulations, with cancer stem cells (CSCs) as the source of this heterogeneity. Since these cells bear in vivo tumorigenicity and metastatic potential, survive chemotherapy and drive relapse, its elimination may be the only way to achieve long-term survival in patients. Thanks to the great advances in the field over the last few years, we know now that cellular metabolism and stemness are highly intertwined in normal development and cancer. Indeed, CSCs show distinct metabolic features as compared with their more differentiated progenies, though their dominant metabolic phenotype varies across tumor entities, patients and even subclones within a tumor. Following initial works focused on glucose metabolism, current studies have unveiled particularities of CSC metabolism in terms of redox state, lipid metabolism and use of alternative fuels, such as amino acids or ketone bodies. In this review, we describe the different metabolic phenotypes attributed to CSCs with special focus on metabolism-based therapeutic strategies tested in preclinical and clinical settings.
Collapse
Affiliation(s)
- Petra Jagust
- Centre for Stem Cells in Cancer and Ageing, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Beatriz de Luxán-Delgado
- Centre for Stem Cells in Cancer and Ageing, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Beatriz Parejo-Alonso
- Traslational Research Unit, Hospital Universitario Miguel Servet, Aragon Institute for Health Research (IIS Aragon), Zaragoza, Spain
| | - Patricia Sancho
- Centre for Stem Cells in Cancer and Ageing, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.,Traslational Research Unit, Hospital Universitario Miguel Servet, Aragon Institute for Health Research (IIS Aragon), Zaragoza, Spain
| |
Collapse
|
107
|
Manzo G. Similarities Between Embryo Development and Cancer Process Suggest New Strategies for Research and Therapy of Tumors: A New Point of View. Front Cell Dev Biol 2019; 7:20. [PMID: 30899759 PMCID: PMC6416183 DOI: 10.3389/fcell.2019.00020] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 02/05/2019] [Indexed: 12/25/2022] Open
Abstract
Here, I propose that cancer stem cells (CSCs) would be equivalent to para-embryonic stem cells (p-ESCs), derived from adult cells de-re-programmed to a ground state. p-ESCs would differ from ESCs by the absence of genomic homeostasis. A p-ESC would constitute the cancer cell of origin (i-CSC or CSC0), capable of generating an initial tumor, corresponding to a pre-implantation blastocyst. In a niche with proper signals, it would engraft as a primary tumor, corresponding to a post-implantation blastocyst. i-CSC progeny would form primary pluripotent and slow self-renewing CSCs (CSC1s), blocked in an undifferentiated state, corresponding to epiblast cells; CSC1s would be tumor-initiating cells (TICs). CSC1s would generate secondary CSCs (CSC2s), corresponding to hypoblast cells; CSC2s would be tumor growth cells (TGCs). CSC1s/CSC2s would generate tertiary CSCs (CSC3s), with a mesenchymal phenotype; CSC3s would be tumor migrating cells (TMCs), corresponding to mesodermal precursors at primitive streak. CSC3s with more favorable conditions (normoxia), by asymmetrical division, would differentiate into cancer progenitor cells (CPCs), and these into cancer differentiated cells (CDCs), thus generating a defined cell hierarchy and tumor progression, mimicking somito-histo-organogenesis. CSC3s with less favorable conditions (hypoxia) would delaminate and migrate as quiescent circulating micro-metastases, mimicking mesenchymal cells in gastrula morphogenetic movements. In metastatic niches, these CSC3s would install and remain dormant in the presence of epithelial/mesenchymal transition (EMT) signals and hypoxia. But, in the presence of mesenchymal/epithelial transition (MET) signals and normoxia, they would revert to self-renewing CSC1s, reproducing the same cell hierarchy of the primary tumor as macro-metastases. Further similarities between ontogenesis and oncogenesis involving crucial factors, such as ID, HSP70, HLA-G, CD44, LIF, and STAT3, are strongly evident at molecular, physiological and immunological levels. Much experimental data about these factors led to considering the cancer process as ectopic rudimentary ontogenesis, where CSCs have privileged immunological conditions. These would consent to CSC development in an adverse environment, just like an embryo, which is tolerated, accepted and favored by the maternal organism in spite of its paternal semi-allogeneicity. From all these considerations, novel research directions, potential innovative tumor therapy and prophylaxis strategies might, theoretically, result.
Collapse
Affiliation(s)
- Giovanni Manzo
- General Pathology, “La Sapienza” University of Rome, Retired, Botrugno, Italy
| |
Collapse
|
108
|
Granda-Díaz R, Menéndez ST, Pedregal Mallo D, Hermida-Prado F, Rodríguez R, Suárez-Fernández L, Vallina A, Sánchez-Canteli M, Rodríguez A, Fernández-García MS, Rodrigo JP, García-Pedrero JM. The Novel Role of SOX2 as an Early Predictor of Cancer Risk in Patients with Laryngeal Precancerous Lesions. Cancers (Basel) 2019; 11:cancers11030286. [PMID: 30823625 PMCID: PMC6468607 DOI: 10.3390/cancers11030286] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/19/2019] [Accepted: 02/22/2019] [Indexed: 01/09/2023] Open
Abstract
The SOX2 gene located at 3q26 is frequently amplified and overexpressed in multiple cancers, including head and neck squamous cell carcinomas (HNSCC). The tumor-promoting activity and involvement of SOX2 in tumor progression has been extensively demonstrated, thereby emerging as a promising therapeutic target. However, the role of SOX2 in early stages of tumorigenesis and its possible contribution to malignant transformation remain unexplored. This study investigates for the first time SOX2 protein expression by immunohistochemistry and gene amplification by real-time PCR using a large series of 94 laryngeal precancerous lesions. Correlations with the histopathological classification and the risk of progression to invasive carcinoma were established. Nuclear SOX2 expression was frequently detected in 38 (40%) laryngeal dysplasias, whereas stromal cells and normal adjacent epithelia showed negative expression. SOX2 gene amplification was detected in 18 (33%) of 55 laryngeal dysplasias. Univariate Cox analysis showed that SOX2 gene amplification (p = 0.046) and protein expression (p < 0.001) but not histological grading (p = 0.432) were significantly associated with laryngeal cancer risk. In multivariate stepwise analysis including age, tobacco, histology, SOX2 gene amplification and SOX2 expression, SOX2 expression (HR = 3.531, 95% CI 1.144 to 10.904; p = 0.028) was the only significant independent predictor of laryngeal cancer development. These findings underscore the relevant role of SOX2 in early tumorigenesis and a novel clinical application of SOX2 expression as independent predictor of laryngeal cancer risk in patients with precancerous lesions beyond current WHO histological grading. Therefore, targeting SOX2 could lead to effective strategies for both cancer prevention and treatment.
Collapse
Affiliation(s)
- Rocío Granda-Díaz
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias, University of Oviedo, Avda. Roma, 33011 Oviedo, Spain.
- Ciber de Cáncer, CIBERONC, Av. Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| | - Sofía T Menéndez
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias, University of Oviedo, Avda. Roma, 33011 Oviedo, Spain.
- Ciber de Cáncer, CIBERONC, Av. Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| | - Daniel Pedregal Mallo
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias, University of Oviedo, Avda. Roma, 33011 Oviedo, Spain.
| | - Francisco Hermida-Prado
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias, University of Oviedo, Avda. Roma, 33011 Oviedo, Spain.
| | - René Rodríguez
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias, University of Oviedo, Avda. Roma, 33011 Oviedo, Spain.
- Ciber de Cáncer, CIBERONC, Av. Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| | - Laura Suárez-Fernández
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias, University of Oviedo, Avda. Roma, 33011 Oviedo, Spain.
| | - Aitana Vallina
- Department of Pathology, Hospital Universitario Central de Asturias, Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, Avda. Roma, 33011 Oviedo, Spain.
| | - Mario Sánchez-Canteli
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias, University of Oviedo, Avda. Roma, 33011 Oviedo, Spain.
| | - Aida Rodríguez
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias, University of Oviedo, Avda. Roma, 33011 Oviedo, Spain.
| | - M Soledad Fernández-García
- Department of Pathology, Hospital Universitario Central de Asturias, Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, Avda. Roma, 33011 Oviedo, Spain.
| | - Juan P Rodrigo
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias, University of Oviedo, Avda. Roma, 33011 Oviedo, Spain.
- Ciber de Cáncer, CIBERONC, Av. Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| | - Juana M García-Pedrero
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias, University of Oviedo, Avda. Roma, 33011 Oviedo, Spain.
- Ciber de Cáncer, CIBERONC, Av. Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| |
Collapse
|
109
|
Chen EYY, Chen JS, Ying SY. The microRNA and the perspectives of miR-302. Heliyon 2019; 5:e01167. [PMID: 30723835 PMCID: PMC6351428 DOI: 10.1016/j.heliyon.2019.e01167] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/18/2018] [Accepted: 01/22/2019] [Indexed: 12/24/2022] Open
Abstract
MiRNAs are naturally occurring, small, non-coding RNA molecules that post-transcriptionally regulate the expression of a large number of genes involved in various biological processes, either through mRNA degradation or through translation inhibition. MiRNAs play important roles in many aspects of physiology and pathology throughout the body, particularly in cancer, which have made miRNAs attractive tools and targets for translational research. The types of non-coding RNAs, biogenesis of miRNAs, circulating miRNAs, and direct delivery of miRNA were briefly reviewed. As a case of point, the role and perspective of miR-302, a family of ES-specific miRNA, on cancer, iPSCs, heart disease were presented.
Collapse
Affiliation(s)
- Emily Yen Yu Chen
- Department of Integrative Anatomical Sciences, Keck School of Medicine, BMT-403, University of Southern California, 1333 San Pablo Street, Los Angeles, CA 90033, USA
- WJWU & LYNN Institute for Stem Cell Research, Santa Fe Springs, CA 90670, USA
| | - Jack S. Chen
- WJWU & LYNN Institute for Stem Cell Research, Santa Fe Springs, CA 90670, USA
| | - Shao-Yao Ying
- Department of Integrative Anatomical Sciences, Keck School of Medicine, BMT-403, University of Southern California, 1333 San Pablo Street, Los Angeles, CA 90033, USA
| |
Collapse
|
110
|
Price ZK, Lokman NA, Ricciardelli C. Differing Roles of Hyaluronan Molecular Weight on Cancer Cell Behavior and Chemotherapy Resistance. Cancers (Basel) 2018; 10:E482. [PMID: 30513961 PMCID: PMC6316154 DOI: 10.3390/cancers10120482] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 02/07/2023] Open
Abstract
Hyaluronan (HA), a glycosaminoglycan located in the extracellular matrix, is important in embryo development, inflammation, wound healing and cancer. There is an extensive body of research demonstrating the role of HA in all stages of cancer, from initiation to relapse and therapy resistance. HA interacts with multiple cell surface receptors, including CD44, receptor for hyaluronan mediated motility (RHAMM) and intracellular signaling pathways, including receptor tyrosine kinase pathways, to promote the survival and proliferation of cancer cells. Additionally, HA promotes the formation of cancer stem cell (CSC) populations, which are hypothesized to be responsible for the initiation of tumors and therapy resistance. Recent studies have identified that the molecular weight of HA plays differing roles on both normal and cancer cell behavior. This review explores the role of HA in cancer progression and therapy resistance and how its molecular weight is important in regulating CSC populations, epithelial to mesenchymal transition (EMT), ATP binding cassette (ABC) transporter expression and receptor tyrosine kinase pathways.
Collapse
Affiliation(s)
- Zoe K Price
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, South Australia 5000, Australia.
| | - Noor A Lokman
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, South Australia 5000, Australia.
| | - Carmela Ricciardelli
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, South Australia 5000, Australia.
| |
Collapse
|
111
|
Motegi T, Tomiyasu H, Goto-Koshino Y, Takahashi M, Hiyoshi-Kanemoto S, Fujino Y, Ohno K, Tsuimoto H. Prognostic value of CD44 variant isoform expression in dogs with multicentric high-grade B-cell lymphoma. Am J Vet Res 2018; 79:961-969. [PMID: 30153061 DOI: 10.2460/ajvr.79.9.961] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To determine the prognostic value of CD44 variant isoform expression in dogs with multicentric high-grade B-cell lymphoma (BCL). ANIMALS 45 dogs with multicentric BCL and 10 healthy control Beagles. PROCEDURES The medical record database of a veterinary teaching hospital was searched to identify dogs with BCL that were treated between November 2005 and April 2015. Information regarding overall response to chemotherapy, progression-free survival (PFS) time, and overall survival time was extracted from each record. Archived lymph node aspirate specimens from dogs with BCL and lymph node aspirate specimens from the 10 control dogs underwent real-time PCR analysis to determine mRNA expression of CD44 variant isoforms of exons 3, 6, and 7 and the CD44 whole isoform. For each isoform, mRNA expression was compared between dogs with BCL and control dogs. The mean relative expression of each isoform was used to classify dogs with BCL into either a high- or low-expression group, and overall response rate, PFS time, and overall survival time (ie, indices of prognosis) were compared between the 2 groups. RESULTS For all isoforms evaluated, mean relative mRNA expression for dogs with BCL was numerically lower than that for control dogs. Dogs with BCL and high CD44 isoform expression had a lower overall response rate, median PFS time, and median overall survival time, compared with dogs with BCL and low CD44 isoform expression. CONCLUSIONS AND CLINICAL RELEVANCE Results indicated that, for dogs with BCL, high expression of exons 3, 6, and 7 was associated with a poor prognosis.
Collapse
|
112
|
Lee IC, Wu YC, Hung WS. Hyaluronic Acid-Based Multilayer Films Regulate Hypoxic Multicellular Aggregation of Pancreatic Cancer Cells with Distinct Cancer Stem-Cell-like Properties. ACS APPLIED MATERIALS & INTERFACES 2018; 10:38769-38779. [PMID: 30395429 DOI: 10.1021/acsami.8b14006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
In vitro spherical cancer models have been widely used in cancer stem cell (CSC) research, and the ability of CSCs to form multicellular colonies is recognized as a morphological marker. However, although several spherical/colony models share a common three-dimensional (3D) conformation, each model displays its own intrinsic properties. Thus, the CSC phenotypes with distinct multicellular aggregate morphologies must be defined and clarified. Here, a novel 3D model was designed to regulate the type of pancreatic CSC colonies that form using niche mimetic hyaluronic acid (HA)-based multilayer nanofilms and hypoxia. The multicellular aggregate morphology, CSC phenotypes, CSC-related marker expression, cell cycle, invasion, and drug resistance were determined. On the basis of the results of a cell morphology analysis, colonies formed on multilayer nanofilms in response to both normoxia and hypoxia, but round and island-type colonies, were investigated. Immunostaining results revealed a significantly higher expression of stem cell markers, such as OCT4, CXCR4, and CD44v6, in colonies that formed on multilayer nanofilms. These colonies also expressed higher levels of E-cadherin, hypoxia-inducible factor-1α, and vimentin, particularly the round-type colonies that formed on HA-based multilayer nanofilms, [poly(allylamine) (PAH)/HA]3, indicating that these colonies exhibit hybrid and metastable epithelial/mesenchymal phenotypes. Moreover, the cell cycle and invasion tests revealed that most of the cells in colonies growing on multilayer nanofilms showed a quiescent, slow cycling phenotype but displayed higher invasion after induction. Furthermore, a hypoxic environment strongly influences the drug resistance. This study describes a useful tool to investigate the diverse phenotypes of pancreatic CSC colonies and to study their regulatory factors that may benefit CSC research.
Collapse
Affiliation(s)
- I-Chi Lee
- Graduate Institute of Biomedical Engineering , Chang-Gung University , Taoyuan 33302 , Taiwan
- Neurosurgery Department , Chang Gung Memorial Hospital , Linkou 33305 , Taiwan
| | - Yu-Chieh Wu
- Graduate Institute of Biomedical Engineering , Chang-Gung University , Taoyuan 33302 , Taiwan
| | - Wei-Shan Hung
- Graduate Institute of Biomedical Engineering , Chang-Gung University , Taoyuan 33302 , Taiwan
| |
Collapse
|
113
|
Izumikawa T, Itano N. Metabolic Reprogramming and Hyaluronan Production in Cancer Stem Cells. TRENDS GLYCOSCI GLYC 2018. [DOI: 10.4052/tigg.1713.1e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Tomomi Izumikawa
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University
| | - Naoki Itano
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University
| |
Collapse
|
114
|
Wang LQ, Yu P, Li B, Guo YH, Liang ZR, Zheng LL, Yang JH, Xu H, Liu S, Zheng LS, Zhou H, Qu LH. miR-372 and miR-373 enhance the stemness of colorectal cancer cells by repressing differentiation signaling pathways. Mol Oncol 2018; 12:1949-1964. [PMID: 30171794 PMCID: PMC6210048 DOI: 10.1002/1878-0261.12376] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 07/28/2018] [Accepted: 08/10/2018] [Indexed: 01/05/2023] Open
Abstract
miR‐372/373, a cluster of stem cell‐specific microRNAs transactivated by the Wnt pathway, has been reported to be dysregulated in various cancers, particularly colorectal cancer (CRC); however, the unique role of these microRNAs in cancer remains to be discovered. In the present study, we characterized the upregulation in expression of miR‐372/373 in CRC tissues from The Cancer Genome Atlas data, and then showed that overexpression of miR‐372/373 enhanced the stemness of CRC cells by enriching the CD26/CD24‐positive cell population and promoting self‐renewal, chemotherapy resistance and the invasive potential of CRC cells. To clarify the mechanism underlying microRNA‐induced stemness, we profiled 45 cell signaling pathways in CRC cells overexpressing miR‐372/373 and found that stemness‐related pathways, such as Nanog and Hedgehog, were upregulated. Instead, differentiation‐related pathways, such as NFκB, MAPK/Erk and VDR, were markedly repressed by miR‐372/373. Numerous new targets of miR‐372/373 were identified, including SPOP, VDR and SETD7, all of which are factors important for cell differentiation. Furthermore, in contrast to the increase in miR‐372/373 expression in CRC tissues, the expression levels of SPOP and VDR mRNA were significantly downregulated in these tissues, indicative of the poor differentiation status of CRC. Taken together, our findings suggest that miR‐372/373 enhance CRC cell stemness by repressing the expression of differentiation genes. These results provide new insights for understanding the function and mechanisms of stem cell‐specific microRNAs in the development of metastasis and drug resistance in CRC.
Collapse
Affiliation(s)
- Lu-Qin Wang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Peng Yu
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Bin Li
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yan-Hua Guo
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zi-Rui Liang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ling-Ling Zheng
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jian-Hua Yang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Hui Xu
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Shun Liu
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Li-Si Zheng
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Hui Zhou
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Liang-Hu Qu
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
115
|
Karamanos NK, Piperigkou Z, Theocharis AD, Watanabe H, Franchi M, Baud S, Brézillon S, Götte M, Passi A, Vigetti D, Ricard-Blum S, Sanderson RD, Neill T, Iozzo RV. Proteoglycan Chemical Diversity Drives Multifunctional Cell Regulation and Therapeutics. Chem Rev 2018; 118:9152-9232. [PMID: 30204432 DOI: 10.1021/acs.chemrev.8b00354] [Citation(s) in RCA: 257] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Nikos K. Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras 26110, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras 26110, Greece
| | - Achilleas D. Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, Aichi 480-1195, Japan
| | - Marco Franchi
- Department for Life Quality Studies, University of Bologna, Rimini 47100, Italy
| | - Stéphanie Baud
- Université de Reims Champagne-Ardenne, Laboratoire SiRMa, CNRS UMR MEDyC 7369, Faculté de Médecine, 51 rue Cognacq Jay, Reims 51100, France
| | - Stéphane Brézillon
- Université de Reims Champagne-Ardenne, Laboratoire de Biochimie Médicale et Biologie Moléculaire, CNRS UMR MEDyC 7369, Faculté de Médecine, 51 rue Cognacq Jay, Reims 51100, France
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster 48149, Germany
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese 21100, Italy
| | - Davide Vigetti
- Department of Medicine and Surgery, University of Insubria, Varese 21100, Italy
| | - Sylvie Ricard-Blum
- University Claude Bernard Lyon 1, CNRS, UMR 5246, Institute of Molecular and Supramolecular Chemistry and Biochemistry, Villeurbanne 69622, France
| | - Ralph D. Sanderson
- Department of Pathology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Thomas Neill
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 10107, United States
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 10107, United States
| |
Collapse
|
116
|
Chopra S, Deodhar K, Pai V, Pant S, Rathod N, Goda JS, Sudhalkar N, Pandey P, Waghmare S, Engineer R, Mahantshetty U, Ghosh J, Gupta S, Shrivastava S. Cancer Stem Cells, CD44, and Outcomes Following Chemoradiation in Locally Advanced Cervical Cancer: Results From a Prospective Study. Int J Radiat Oncol Biol Phys 2018; 103:161-168. [PMID: 30213750 DOI: 10.1016/j.ijrobp.2018.09.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 08/20/2018] [Accepted: 09/03/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE Although cancer stem cells (CSCs) have been reported across solid tumors, there is a dearth of data regarding CSC and its impact on outcomes of cervical cancer. METHODS AND MATERIALS From October 2013 to December 2015, patients with squamous cancer of the cervix (stage IB2-IVA) were included. Pretreatment and posttreatment biopsy was obtained and immunohistochemistry was performed for SOX-2, OCT-4, Nanog, CD44, and Podoplanin. All patients received concurrent radiation and brachytherapy to an equivalent dose of 80 to 84 Gy to point A with concurrent weekly cisplatin. Correlation of CSC expression was performed with known prognostic factors. The effect of stem cell expression on disease outcomes was tested within multivariate analysis. RESULTS One hundred fifty patients were included. The median dose to point A was 83 Gy (46-89 Gy) and a median of 4 cycles (range, 0-6 cycles) of chemotherapy was administered. At baseline, moderate to strong immunohistochemical expression of SOX-2, OCT-4, Nanog, CD44, and Podoplanin was observed in 12.8%, 4.8%, 24.4%, 15.5%, and 1.3% of patients, respectively. At median follow-up of 30 months (range, 3-51 months), locoregional and distant relapse was observed in 12.2% and 23.1% of patients, of whom 4.7% had both local and distant relapse. The 3-year disease-free survival rate was 87%. On multivariate analysis, moderate to high CSC expression and CD44 low status (hazard ratio [HR] = 8.8; 95% confidence interval [CI], 1.0-77.2; P < .04) independently predicted for locoregional relapse-free survival. International Federation of Gynecology and Obstetrics stage (HR = 2.6; 95% CI, 1.3-5.4; P = .004) and presence of residual tumor after external radiation (HR = 3.5; 95% CI, 1.8-6.5; P = .0001) predicted for a detriment in disease-free survival. CONCLUSIONS The presence of stem cell proteins and loss of CD44 independently predicts for reduced locoregional control in locally advanced cervical cancer. Further investigation into the interaction of stem cell and CD44 biology is warranted.
Collapse
Affiliation(s)
- Supriya Chopra
- Department of Radiation Oncology, Advanced Centre for Treatment, Education and Research in Cancer, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India.
| | - Kedar Deodhar
- Department of Pathology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Venkatesh Pai
- Clinical Biology Laboratory, Department of Radiation Oncology, Advanced Centre for Treatment, Education and Research in Cancer, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Sidharth Pant
- Department of Radiation Oncology, Advanced Centre for Treatment, Education and Research in Cancer, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Nidul Rathod
- Clinical Biology Laboratory, Department of Radiation Oncology, Advanced Centre for Treatment, Education and Research in Cancer, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Jayant S Goda
- Department of Radiation Oncology, Advanced Centre for Treatment, Education and Research in Cancer, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Niyati Sudhalkar
- Clinical Biology Laboratory, Department of Radiation Oncology, Advanced Centre for Treatment, Education and Research in Cancer, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Puloma Pandey
- Clinical Biology Laboratory, Department of Radiation Oncology, Advanced Centre for Treatment, Education and Research in Cancer, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Sanjeev Waghmare
- Stem Cell Biology, Advanced Centre for Treatment, Education and Research in Cancer, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Reena Engineer
- Department of Radiation Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Umesh Mahantshetty
- Department of Radiation Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Jaya Ghosh
- Medical Oncology, Advanced Centre for Treatment, Education and Research in Cancer, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Sudeep Gupta
- Medical Oncology, Advanced Centre for Treatment, Education and Research in Cancer, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Shyam Shrivastava
- Department of Radiation Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| |
Collapse
|
117
|
Kim YD, Kim HS, Lee J, Choi JK, Han E, Jeong JE, Cho YS. ESRP1-Induced CD44 v3 Is Important for Controlling Pluripotency in Human Pluripotent Stem Cells. Stem Cells 2018; 36:1525-1534. [DOI: 10.1002/stem.2864] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 05/15/2018] [Accepted: 05/19/2018] [Indexed: 01/12/2023]
Affiliation(s)
- Young-Dae Kim
- Stem Cell Research Laboratory; Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology; Daejeon 34141 Republic of Korea
| | - Han-Seop Kim
- Stem Cell Research Laboratory; Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology; Daejeon 34141 Republic of Korea
| | - Jungwoon Lee
- Stem Cell Research Laboratory; Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology; Daejeon 34141 Republic of Korea
| | - Jung-Kyun Choi
- Stem Cell Research Laboratory; Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology; Daejeon 34141 Republic of Korea
- Department of Bioscience; KRIBB School, University of Science & Technology; Daejeon Republic of Korea
| | - Enna Han
- Stem Cell Research Laboratory; Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology; Daejeon 34141 Republic of Korea
- Department of Bioscience; KRIBB School, University of Science & Technology; Daejeon Republic of Korea
| | - Ji E. Jeong
- Stem Cell Research Laboratory; Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology; Daejeon 34141 Republic of Korea
| | - Yee S. Cho
- Stem Cell Research Laboratory; Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology; Daejeon 34141 Republic of Korea
- Department of Bioscience; KRIBB School, University of Science & Technology; Daejeon Republic of Korea
| |
Collapse
|
118
|
Stem-like and highly invasive prostate cancer cells expressing CD44v8-10 marker originate from CD44-negative cells. Oncotarget 2018; 9:30905-30918. [PMID: 30112117 PMCID: PMC6089404 DOI: 10.18632/oncotarget.25773] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 06/22/2018] [Indexed: 12/28/2022] Open
Abstract
In human prostate cancer (PCa), the neuroendocrine cells, expressing the prostate cancer stem cell (CSC) marker CD44, may be resistant to androgen ablation and promote tumor recurrence. During the study of heterogeneity of the highly aggressive neuroendocrine PCa cell lines PC3 and DU-145, we isolated and expanded in vitro a minor subpopulation of very small cells lacking CD44 (CD44neg). Unexpectedly, these sorted CD44neg cells rapidly and spontaneously converted to a stable CD44high phenotype specifically expressing the CD44v8-10 isoform which the sorted CD44high subpopulation failed to express. Surprisingly and potentially interesting, in these cells expression of CD44v8-10 was found to be induced in stem cell medium. CD44 variant isoforms are known to be more expressed in CSC and metastatic cells than CD44 standard isoform. In agreement, functional analysis of the two sorted and cultured subpopulations has shown that the CD44v8-10pos PC3 cells, resulting from the conversion of the CD44neg subpopulation, were more invasive in vitro and had a higher clonogenic potential than the sorted CD44high cells, in that they produced mainly holoclones, known to be enriched in stem-like cells. Of interest, the CD44v8-10 is more expressed in human PCa biopsies than in normal gland. The discovery of CD44v8-10pos cells with stem-like and invasive features, derived from a minoritarian CD44neg cell population in PCa, alerts on the high plasticity of stem-like markers and urges for prudency on the approaches to targeting the putative CSC.
Collapse
|
119
|
Xiang Y, Zhou X. Octamer-binding transcription factor 4 correlates with complex karyotype, FLT3-ITD mutation and poorer risk stratification, and predicts unfavourable prognosis in patients with acute myeloid leukaemia. Hematology 2018; 23:721-728. [PMID: 29950146 DOI: 10.1080/10245332.2018.1482050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Yongsheng Xiang
- Department of Hematology, The First People’s Hospital of Jingmen, Jingmen, People’s Republic of China
| | - Xiaofen Zhou
- Department of Hematology, Xiangyang Central Hospital, The Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, People’s Republic of China
| |
Collapse
|
120
|
Redfern AD, Spalding LJ, Thompson EW. The Kraken Wakes: induced EMT as a driver of tumour aggression and poor outcome. Clin Exp Metastasis 2018; 35:285-308. [PMID: 29948647 DOI: 10.1007/s10585-018-9906-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 05/23/2018] [Indexed: 02/06/2023]
Abstract
Epithelial mesenchymal transition (EMT) describes the shift of cells from an epithelial form to a contact independent, migratory, mesenchymal form. In cancer the change is linked to invasion and metastasis. Tumour conditions, including hypoxia, acidosis and a range of treatments can trigger EMT, which is implicated in the subsequent development of resistance to those same treatments. Consequently, the degree to which EMT occurs may underpin the entire course of tumour progression and treatment response in a patient. In this review we look past the protective effect of EMT against the initial treatment, to the role of the mesenchymal state, once triggered, in promoting disease growth, spread and future treatment insensitivity. In patients a correlation was found between the propensity of a treatment to induce EMT and failure of that treatment to provide a survival benefit, implicating EMT induction in accelerated tumour progression after treatment cessation. Looking to the mechanisms driving this detrimental effect; increased proliferation, suppressed apoptosis, stem cell induction, augmented angiogenesis, enhanced metastatic dissemination, and immune tolerance, can all result from treatment-induced EMT and could worsen outcome. Evidence also suggests EMT induction with earlier therapies attenuates benefits of later treatments. Looking beyond epithelial tumours, de-differentiation also has therapy-attenuating effects and reversal thereof may yield similar rewards. A range of potential therapies are in development that may address the diverse mechanisms and molecular control systems involved in EMT-induced accelerated progression. Considering the broad reaching effects of mesenchymal shift identified, successful deployment of such treatments could substantially improve patient outcomes.
Collapse
Affiliation(s)
- Andrew D Redfern
- School of Medicine, University of Western Australia (UWA), Harry Perkins Building, Fiona Stanley Hospital Campus, Robin Warren Drive, Murdoch, WA, 6150, Australia.
| | - Lisa J Spalding
- School of Medicine, University of Western Australia (UWA), Harry Perkins Building, Fiona Stanley Hospital Campus, Robin Warren Drive, Murdoch, WA, 6150, Australia
| | - Erik W Thompson
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Australia.,Translational Research Institute, Woolloongabba, Australia.,Department of Surgery, University of Melbourne, Melbourne, Australia
| |
Collapse
|
121
|
The biology and role of CD44 in cancer progression: therapeutic implications. J Hematol Oncol 2018; 11:64. [PMID: 29747682 PMCID: PMC5946470 DOI: 10.1186/s13045-018-0605-5] [Citation(s) in RCA: 852] [Impact Index Per Article: 121.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 04/26/2018] [Indexed: 02/07/2023] Open
Abstract
CD44, a non-kinase transmembrane glycoprotein, is overexpressed in several cell types including cancer stem cells and frequently shows alternative spliced variants that are thought to play a role in cancer development and progression. Hyaluronan, the main ligand for CD44, binds to and activates CD44 resulting in activation of cell signaling pathways that induces cell proliferation, increases cell survival, modulates cytoskeletal changes, and enhances cellular motility. The different functional roles of CD44 standard (CD44s) and specific CD44 variant (CD44v) isoforms are not fully understood. CD44v contain additional peptide motifs that can interact with and sequester growth factors and cytokines at the cell surface thereby functioning as coreceptors to facilitate cell signaling. Moreover, CD44v were expressed in metastasized tumors, whereas switching between CD44v and CD44s may play a role in regulating epithelial to mesenchymal transition (EMT) and in the adaptive plasticity of cancer cells. Here, we review current data on the structural and functional properties of CD44, the known roles for CD44 in tumorigencity, the regulation of CD44 expression, and the potential for targeting CD44 for cancer therapy.
Collapse
|
122
|
Wang L, Xu T, Cui M. Are ovarian cancer stem cells the target for innovative immunotherapy? Onco Targets Ther 2018; 11:2615-2626. [PMID: 29780254 PMCID: PMC5951213 DOI: 10.2147/ott.s155458] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cancer stem cells (CSCs), a subpopulation of cancer cells with the ability of self-renewal and differentiation, are believed to be responsible for tumor generation, progression, metastasis, and relapse. Ovarian cancer, the most malignant gynecological cancer, has consistent pathology behavior with CSC model, which suggests that therapies based on ovarian cancer stem cells (OCSCs) can gain a more successful prognosis. Much evidence has proved that epigenetic mechanism played an important role in tumor formation and sustainment. Since CSCs are generally resistant to conventional therapies (chemotherapy and radiotherapy), immunotherapy is a more effective method that has been implemented in the clinic. Chimeric antigen receptor (CAR)-T cell, an adoptive cellular immunotherapy, which results in apparent elimination of tumor in both hematologic and solid cancers, could be used for ovarian cancer. This review covers the basic conception of CSCs and OCSCs, the implication of epigenetic mechanism underlying cancer evolution considering CSC model, the immunotherapies reported for ovarian cancer targeting OCSCs currently, and the relationship between immune system and hierarchy cancer organized by CSCs. Particularly, the promising prospects and potential pitfalls of targeting OCSC surface markers to design CAR-T cellular immunotherapy are discussed here.
Collapse
Affiliation(s)
- Liang Wang
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Tianmin Xu
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Manhua Cui
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| |
Collapse
|
123
|
Mishra A, Sriram H, Chandarana P, Tanavde V, Kumar RV, Gopinath A, Govindarajan R, Ramaswamy S, Sadasivam S. Decreased expression of cell adhesion genes in cancer stem-like cells isolated from primary oral squamous cell carcinomas. Tumour Biol 2018; 40:1010428318780859. [PMID: 29888653 DOI: 10.1177/1010428318780859] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The goal of this study was to isolate cancer stem-like cells marked by high expression of CD44, a putative cancer stem cell marker, from primary oral squamous cell carcinomas and identify distinctive gene expression patterns in these cells. From 1 October 2013 to 4 September 2015, 76 stage III-IV primary oral squamous cell carcinoma of the gingivobuccal sulcus were resected. In all, 13 tumours were analysed by immunohistochemistry to visualise CD44-expressing cells. Expression of CD44 within The Cancer Genome Atlas-Head and Neck Squamous Cell Carcinoma RNA-sequencing data was also assessed. Seventy resected tumours were dissociated into single cells and stained with antibodies to CD44 as well as CD45 and CD31 (together referred as Lineage/Lin). From 45 of these, CD44+Lin- and CD44-Lin- subpopulations were successfully isolated using fluorescence-activated cell sorting, and good-quality RNA was obtained from 14 such sorted pairs. Libraries from five pairs were sequenced and the results analysed using bioinformatics tools. Reverse transcription quantitative polymerase chain reaction was performed to experimentally validate the differential expression of selected candidate genes identified from the transcriptome sequencing in the same 5 and an additional 9 tumours. CD44 was expressed on the surface of poorly differentiated tumour cells, and within the The Cancer Genome Atlas-Head and Neck Squamous Cell Carcinoma samples, its messenger RNA levels were higher in tumours compared to normal. Transcriptomics revealed that 102 genes were upregulated and 85 genes were downregulated in CD44+Lin- compared to CD44-Lin- cells in at least 3 of the 5 tumours sequenced. The upregulated genes included those involved in immune regulation, while the downregulated genes were enriched for genes involved in cell adhesion. Decreased expression of PCDH18, MGP, SPARCL1 and KRTDAP was confirmed by reverse transcription quantitative polymerase chain reaction. Lower expression of the cell-cell adhesion molecule PCDH18 correlated with poorer overall survival in the The Cancer Genome Atlas-Head and Neck Squamous Cell Carcinoma data highlighting it as a potential negative prognostic factor in this cancer.
Collapse
Affiliation(s)
- Amrendra Mishra
- 1 Institute for Stem Cell Biology and Regenerative Medicine, National Centre for Biological Sciences, UAS-GKVK Campus, Bengaluru, India
- 2 Hannover Biomedical Research School, Hannover Medical School, Hannover, Germany
| | - Harshini Sriram
- 1 Institute for Stem Cell Biology and Regenerative Medicine, National Centre for Biological Sciences, UAS-GKVK Campus, Bengaluru, India
| | | | - Vivek Tanavde
- 3 iBioAnalysis Pvt. Ltd., Ahmedabad, India
- 4 Division of Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Ahmedabad, India
- 5 Bioinformatics Institute, Agency for Science Technology and Research (A*STAR), Singapore
| | - Rekha V Kumar
- 6 Kidwai Memorial Institute of Oncology, Bengaluru, India
| | | | | | - S Ramaswamy
- 1 Institute for Stem Cell Biology and Regenerative Medicine, National Centre for Biological Sciences, UAS-GKVK Campus, Bengaluru, India
| | - Subhashini Sadasivam
- 1 Institute for Stem Cell Biology and Regenerative Medicine, National Centre for Biological Sciences, UAS-GKVK Campus, Bengaluru, India
| |
Collapse
|
124
|
Cai Z, Cao Y, Luo Y, Hu H, Ling H. Signalling mechanism(s) of epithelial-mesenchymal transition and cancer stem cells in tumour therapeutic resistance. Clin Chim Acta 2018; 483:156-163. [PMID: 29709449 DOI: 10.1016/j.cca.2018.04.033] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 04/24/2018] [Accepted: 04/25/2018] [Indexed: 02/06/2023]
Abstract
Epithelial-mesenchymal transition (EMT) leads to tumour progression, including tumour metastasis, disease recurrence and therapy resistance. Cancer stem cells (CSCs) are a small group of cells that have the ability to undergo self-renewal and heterogeneous differentiation, which play a key role in the occurrence and development of cancer. EMT can promote tumour cells to develop stem cell characteristics, which makes tumours more difficult to treat. Therefore, exploring the role of EMT and CSCs in the metastasis of cancer is of great significance to guide tumour treatment and prognosis. In this review, we discuss EMT and CSCs in cancer progression and therapeutic resistance, with a special focus on the common characteristics and relationships between these processes, to explore the crucial relationships in the development of improved anti-tumour therapies. AREAS COVERED In this brief review article, the author has searched PubMed and Wikipedia for original research and reviewed articles to gather current information on the association of CSCs and EMT with therapeutic resistance characteristics, cancer growth and metastasis, which are believed to be regulated by the TGF-β, Wnt, Hedgehog (Hh), β-catenin, STAT3, Notch, and Nanog signalling pathways and other factors (miRNAs, microenvironment and additional cytokines).
Collapse
Affiliation(s)
- Zhihong Cai
- Key Laboratory of Tumor Cellular & Molecular Pathology (University of South China),College of Hunan Province, Cancer Research Institute, University of South China, Hengyang, PR China
| | - Yijing Cao
- Key Laboratory of Tumor Cellular & Molecular Pathology (University of South China),College of Hunan Province, Cancer Research Institute, University of South China, Hengyang, PR China
| | - Yichen Luo
- Key Laboratory of Tumor Cellular & Molecular Pathology (University of South China),College of Hunan Province, Cancer Research Institute, University of South China, Hengyang, PR China
| | - Haobin Hu
- Key Laboratory of Tumor Cellular & Molecular Pathology (University of South China),College of Hunan Province, Cancer Research Institute, University of South China, Hengyang, PR China
| | - Hui Ling
- Key Laboratory of Tumor Cellular & Molecular Pathology (University of South China),College of Hunan Province, Cancer Research Institute, University of South China, Hengyang, PR China.
| |
Collapse
|
125
|
Rios de la Rosa JM, Tirella A, Tirelli N. Receptor-Targeted Drug Delivery and the (Many) Problems We Know of: The Case of CD44 and Hyaluronic Acid. ACTA ACUST UNITED AC 2018. [DOI: 10.1002/adbi.201800049] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Julio M. Rios de la Rosa
- NorthWest Centre for Advanced Drug Delivery (NoWCADD); School of Health Sciences; University of Manchester; Oxford Road Manchester M13 9PT UK
| | - Annalisa Tirella
- NorthWest Centre for Advanced Drug Delivery (NoWCADD); School of Health Sciences; University of Manchester; Oxford Road Manchester M13 9PT UK
| | - Nicola Tirelli
- NorthWest Centre for Advanced Drug Delivery (NoWCADD); School of Health Sciences; University of Manchester; Oxford Road Manchester M13 9PT UK
- Laboratory of Polymers and Biomaterials; Fondazione Istituto Italiano di Tecnologia; Genova 16163 Italy
| |
Collapse
|
126
|
Piperigkou Z, Götte M, Theocharis AD, Karamanos NK. Insights into the key roles of epigenetics in matrix macromolecules-associated wound healing. Adv Drug Deliv Rev 2018; 129:16-36. [PMID: 29079535 DOI: 10.1016/j.addr.2017.10.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/14/2017] [Accepted: 10/20/2017] [Indexed: 02/08/2023]
Abstract
Extracellular matrix (ECM) is a dynamic network of macromolecules, playing a regulatory role in cell functions, tissue regeneration and remodeling. Wound healing is a tissue repair process necessary for the maintenance of the functionality of tissues and organs. This highly orchestrated process is divided into four temporally overlapping phases, including hemostasis, inflammation, proliferation and tissue remodeling. The dynamic interplay between ECM and resident cells exerts its critical role in many aspects of wound healing, including cell proliferation, migration, differentiation, survival, matrix degradation and biosynthesis. Several epigenetic regulatory factors, such as the endogenous non-coding microRNAs (miRNAs), are the drivers of the wound healing response. microRNAs have pivotal roles in regulating ECM composition during wound healing and dermal regeneration. Their expression is associated with the distinct phases of wound healing and they serve as target biomarkers and targets for systematic regulation of wound repair. In this article we critically present the importance of epigenetics with particular emphasis on miRNAs regulating ECM components (i.e. glycoproteins, proteoglycans and matrix proteases) that are key players in wound healing. The clinical relevance of miRNA targeting as well as the delivery strategies designed for clinical applications are also presented and discussed.
Collapse
|
127
|
Abstract
Bone morphogenetic proteins (BMPs) are a diverse class of molecules with over 20 growth factor proteins that belong to the transforming growth factor-β (TGF-β) family and are highly associated with bone formation and disease development. Aberrant expression of various BMPs has been reported in several cancer tissues. Biological function studies have elicited the dual role of BMPs in both cancer development and suppression. Furthermore, a variety of BMP antagonists, ligands, and receptors have been shown to reduce or enhance tumorigenesis and metastasis. Knockout mouse models of BMP signaling components have also revealed that the suppression of BMP signaling impairs cancer metastasis. Herein, we highlight the basic clinical background and involvement of BMPs in modulating cancer progression and their dynamic interactions (e.g., with microRNAs) in the tumor microenvironment in addition to their mutations and roles in chemoprevention. We also suggest that BMPs should be considered a powerful putative therapeutic target in tumorigenesis and bone metastasis.
Collapse
Affiliation(s)
- Duc-Hiep Bach
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 151-742, Republic of Korea
| | - Hyen Joo Park
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 151-742, Republic of Korea
| | - Sang Kook Lee
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 151-742, Republic of Korea
| |
Collapse
|
128
|
Wuebben EL, Rizzino A. The dark side of SOX2: cancer - a comprehensive overview. Oncotarget 2018; 8:44917-44943. [PMID: 28388544 PMCID: PMC5546531 DOI: 10.18632/oncotarget.16570] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 03/16/2017] [Indexed: 12/14/2022] Open
Abstract
The pluripotency-associated transcription factor SOX2 is essential during mammalian embryogenesis and later in life, but SOX2 expression can also be highly detrimental. Over the past 10 years, SOX2 has been shown to be expressed in at least 25 different cancers. This review provides a comprehensive overview of the roles of SOX2 in cancer and focuses on two broad topics. The first delves into the expression and function of SOX2 in cancer focusing on the connection between SOX2 levels and tumor grade as well as patient survival. As part of this discussion, we address the developing connection between SOX2 expression and tumor drug resistance. We also call attention to an under-appreciated property of SOX2, its levels in actively proliferating tumor cells appear to be optimized to maximize tumor growth - too little or too much SOX2 dramatically alters tumor growth. The second topic of this review focuses on the exquisite array of molecular mechanisms that control the expression and transcriptional activity of SOX2. In addition to its complex regulation at the transcriptional level, SOX2 expression and activity are controlled carefully by microRNAs, long non-coding RNAs, and post-translational modifications. In the Conclusion and Future Perspectives section, we point out that there are still important unanswered questions. Addressing these questions is expected to lead to new insights into the functions of SOX2 in cancer, which will help design novels strategies for more effectively treating some of the most deadly cancers.
Collapse
Affiliation(s)
- Erin L Wuebben
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Angie Rizzino
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
129
|
Schomberg J, Ziogas A, Anton-Culver H, Norden-Krichmar T. Identification of a gene expression signature predicting survival in oral cavity squamous cell carcinoma using Monte Carlo cross validation. Oral Oncol 2018; 78:72-79. [PMID: 29496061 DOI: 10.1016/j.oraloncology.2018.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/20/2017] [Accepted: 01/18/2018] [Indexed: 11/30/2022]
Abstract
OBJECTIVES This study aims to identify a robust signature that performs well in predicting overall survival across tumor phenotypes and treatment strata, and validates the application of Monte Carlo cross validation (MCCV) as a means of identifying molecular signatures when utilizing small and highly heterogeneous datasets. MATERIALS AND METHODS RNA sequence gene expression data for 264 patient tumors were acquired from The Cancer Genome Atlas (TCGA). 100 iterations of Monte Carlo cross validation were applied to differential expression and Cox model validation. The association between the gene signature risk score and overall survival was measured using Kaplan-Meier survival curves, univariate, and multivariable Cox regression analyses. RESULTS Pathway analysis findings indicate that ligand-gated ion channel pathways are the most significantly enriched with the genes in the aggregated signature. The aggregated signature described in this study is predictive of overall survival in oral cancer patients across demographic and treatment strata. CONCLUSION This study reinforces previous findings supporting the role of ion channel gating, interleukin, calcitonin receptor, and keratinization pathways in tumor progression and treatment response in oral cancer. These results strengthen the argument that differential expression of genes within these pathways reduces tumor susceptibility to treatment. Conducting differential gene expression (DGE) with Monte Carlo cross validation, as this study describes, offers a potential solution to decreasing the variability in DGE results across future studies that are reliant upon highly heterogeneous datasets. This improves the ability of studies reliant upon similarly structured datasets to reach results that are reproducible.
Collapse
Affiliation(s)
- John Schomberg
- Department of Epidemiology School of Medicine, University of California, Irvine, Irvine, CA 92617, United States
| | - Argyrios Ziogas
- Department of Epidemiology School of Medicine, University of California, Irvine, Irvine, CA 92617, United States
| | - Hoda Anton-Culver
- Department of Epidemiology School of Medicine, University of California, Irvine, Irvine, CA 92617, United States.
| | - Trina Norden-Krichmar
- Department of Epidemiology School of Medicine, University of California, Irvine, Irvine, CA 92617, United States
| |
Collapse
|
130
|
Ning X, Wang C, Zhang M, Wang K. Ectopic Expression of miR-147 Inhibits Stem Cell Marker and Epithelial-Mesenchymal Transition (EMT)-Related Protein Expression in Colon Cancer Cells. Oncol Res 2018; 27:399-406. [PMID: 29426374 PMCID: PMC7848281 DOI: 10.3727/096504018x15179675206495] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Colon cancer is one of the most common cancers in the world. Epithelial-to-mesenchymal transition (EMT) is a crucial step in tumor progression and is also involved in the acquisition of stem cell-like properties. Some miRNAs have been shown to function as either tumor suppressors or oncogenes in colon cancer. Here we investigated the role of miR-147 in the regulation of the stem cell-like traits of colon cancer cells. We observed that miR-147 was downregulated in several colon cancer cell lines, and overexpressed miR-147 decreased the expression of cancer stem cell (CSC) markers OCT4, SOX2, and NANOG in the colon cancer cell lines HCT116 and SW480. Overexpressed miR-147 inhibited EMT by increasing the expression of epithelial markers E-cadherin and α-catenin while decreasing the expression of mesenchymal markers fibronectin and vimentin. Moreover, activation of EMT by TGF-β1 treatment significantly counteracted the inhibitive effect of miR-147 on the expression of CSC markers OCT4, SOX2, and NANOG, supporting the idea that overexpressing miR-147 inhibited stem cell-like traits by suppressing EMT in colon cancer. In addition, we found that overexpressed miR-147 downregulated the expression of β-catenin, c-myc, and survivin, which were related to the Wnt/β-catenin pathway. Moreover, treatment of miR-147 mimic-transfected cells with the Wnt/β-catenin pathway activator LiCl attenuated the inhibitive effect of the miR-147 mimic on the EMT and stem cell-like traits of colon cancer cells, indicating that ectopic expression of miR-147 inhibited stem cell-like traits in colon cancer cells by suppressing EMT via the Wnt/β-catenin pathway. In summary, our present study highlighted the crucial role of miR-147 in the inhibition of the stem cell-like traits of colon cancer cells and indicated that miR-147 could be a promising therapeutic target for colon cancer treatment.
Collapse
Affiliation(s)
- Xiaofei Ning
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jining Medical College, Jining, P.R. China
| | - Cong Wang
- Department of Medical Ultrasonography, Affiliated Hospital of Jining Medical College, Jining, P.R. China
| | - Meng Zhang
- Department of Medical Ultrasonography, Affiliated Hospital of Jining Medical College, Jining, P.R. China
| | - Kecheng Wang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jining Medical College, Jining, P.R. China
| |
Collapse
|
131
|
Wang D, Xiang T, Zhao Z, Lin K, Yin P, Jiang L, Liang Z, Zhu B. Autocrine interleukin-23 promotes self-renewal of CD133+ ovarian cancer stem-like cells. Oncotarget 2018; 7:76006-76020. [PMID: 27738346 PMCID: PMC5342794 DOI: 10.18632/oncotarget.12579] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 08/24/2016] [Indexed: 12/18/2022] Open
Abstract
Cancer stem cells (CSCs) are a group of cells which possess the ability of self-renewing and unlimited proliferation. And these CSCs are thought to be the cause of metastasis, recurrence and resistance. Recent study has found that pro-inflammatory cytokine and chemotactic factor mediate the self-renewing and differentiation of most of CSCs. Thus we speculate that ovarian cancer stem cells (OCSCs) can also maintain the ability of self-renewing and differentiation by releasing inflammatory factor. This report we discuss the biological characteristics and the specific molecular mechanism mediated by interleukin-23 (IL-23) and its receptor on the self-renewing of OCSCs. We found that OCSCs had high expression of IL-23 and IL-23R. IL-23 could promote the self-renewal ability of OCSCs and played a very important role to maintain the stable expression of stem cell markers in vitro. Moreover, we verified that IL-23 could maintain the potential tumorigenic of OCSCs in vivo and mediate the self-renewal ability and the formation of tumor in OCSCs by activating the signal pathways of STAT3 and NF-κB. In addition, human low differentiation tissues showed overexpression of IL-23. And IL-23 positively correlated to the expression level of CD133, Nanog and Oct4. In conclusion, Our discoveries demonstrate that autocrine IL-23 contribute to ovarian cancer malignancy through promoting the self-renewal of CD133+ ovarian cancer stem-like cells, and this suggests that IL-23 and its signaling pathway might serve as therapeutic targets for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Dan Wang
- Department of Obstetrics and Gynecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Tong Xiang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.,Department of Oncology, No. 421 Hospital of PLA, Guangzhou 510318, China
| | - Zhongquan Zhao
- Department of Oncology, Fuzhou General Hospital, Fuzhou, Fujian 350025, China
| | - Kailong Lin
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Pin Yin
- Department of Obstetrics and Gynecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Lupin Jiang
- Department of Obstetrics and Gynecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Zhiqing Liang
- Department of Obstetrics and Gynecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| |
Collapse
|
132
|
Zhao Q, Zhou H, Liu Q, Cao Y, Wang G, Hu A, Ruan L, Wang S, Bo Q, Chen W, Hu C, Xu D, Tao F, Cao J, Ge Y, Yu Z, Li L, Wang H. Prognostic value of the expression of cancer stem cell-related markers CD133 and CD44 in hepatocellular carcinoma: From patients to patient-derived tumor xenograft models. Oncotarget 2018; 7:47431-47443. [PMID: 27329727 PMCID: PMC5216952 DOI: 10.18632/oncotarget.10164] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 06/06/2016] [Indexed: 12/31/2022] Open
Abstract
High expression of cancer stem cell (CSC) markers is related to poor prognosis of patients with hepatocellular carcinoma (HCC). However, the expression of these markers in patient-derived xenograft (PDX) models and the relationship of the expression levels of these markers between HCC patients and their PDX models at subsequent low passages are unclear. To investigate the prognostic impact of putative CSC markers in patients with HCC and in related PDX models, the expression of CD133, CD90, CD44, ALDH1, CK7, CK19, OCT4, SOX2, vimentin, nestin, CD13 and EpCam were assessed by quantitative reverse transcription-PCR (qRT-PCR) and then were validated using immunohistochemistry in tumor or peritumoral tissues from patients and tumor tissues from PDX models. Cumulative survival analysis of the patients and animals was conducted using the Kaplan-Meier method and the log-rank test. Only the expression levels of CD133 and CD44 were higher in tumor tissues than in the peritumoral tissues of HCC patients by qRT-PCR. High consistency of the prognostic value of the expression of CD133/CD44 was observed in HCC patients and the PDX models. High expression levels of CD133 and CD44 were positively related to the poor prognosis of HCC patients and to that in the PDX models. PDX HCC models in the present study have been suggested to be predictive of disease outcome, which could shed light on personalized medicine and the mechanisms of CSC marker expression on prognosis.
Collapse
Affiliation(s)
- Qihong Zhao
- Department of Food and Nutrition Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Heng Zhou
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Qifei Liu
- Department of Food and Nutrition Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Ye Cao
- Department of Food and Nutrition Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Gang Wang
- Department of Oncology, Affiliated Provincial Hospital of Anhui Medical University, Hefei, China
| | - Anla Hu
- Department of Food and Nutrition Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Liang Ruan
- Department of Food and Nutrition Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Sufang Wang
- Department of Food and Nutrition Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Qingli Bo
- Department of Food and Nutrition Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Wenjun Chen
- Department of Food and Nutrition Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Chuanlai Hu
- Department of Food and Nutrition Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Dexiang Xu
- Department of Food and Nutrition Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Fangbiao Tao
- Department of Food and Nutrition Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Jiyu Cao
- Department of Food and Nutrition Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Yongsheng Ge
- Department of General Surgery, Affiliated Provincial Hospital of Anhui Medical University, Hefei, China
| | - Zongfan Yu
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Li Li
- Department of Food and Nutrition Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Hua Wang
- School of Pharmacy, Anhui Medical University, Hefei, China.,Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Institute for Liver Disease, Anhui Medical University, Hefei, China
| |
Collapse
|
133
|
Qadan MA, Piuzzi NS, Boehm C, Bova W, Moos M, Midura RJ, Hascall VC, Malcuit C, Muschler GF. Variation in primary and culture-expanded cells derived from connective tissue progenitors in human bone marrow space, bone trabecular surface and adipose tissue. Cytotherapy 2018; 20:343-360. [PMID: 29396254 DOI: 10.1016/j.jcyt.2017.11.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 11/27/2017] [Accepted: 11/29/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND AIMS Connective tissue progenitors (CTPs) embody the heterogeneous stem and progenitor cell populations present in native tissue. CTPs are essential to the formation and remodeling of connective tissue and represent key targets for tissue-engineering and cell-based therapies. To better understand and characterize CTPs, we aimed to compare the (i) concentration and prevalence, (ii) early in vitro biological behavior and (iii) expression of surface-markers and transcription factors among cells derived from marrow space (MS), trabecular surface (TS), and adipose tissues (AT). METHODS Cancellous-bone and subcutaneous-adipose tissues were collected from 8 patients. Cells were isolated and cultured. Colony formation was assayed using Colonyze software based on ASTM standards. Cell concentration ([Cell]), CTP concentration ([CTP]) and CTP prevalence (PCTP) were determined. Attributes of culture-expanded cells were compared based on (i) effective proliferation rate and (ii) expression of surface-markers CD73, CD90, CD105, SSEA-4, SSEA-3, SSEA-1/CD15, Cripto-1, E-Cadherin/CD324, Ep-CAM/CD326, CD146, hyaluronan and transcription factors Oct3/4, Sox-2 and Nanog using flow cytometry. RESULTS Mean [Cell], [CTP] and PCTP were significantly different between MS and TS samples (P = 0.03, P = 0.008 and P= 0.0003), respectively. AT-derived cells generated the highest mean total cell yield at day 6 of culture-4-fold greater than TS and more than 40-fold greater than MS per million cells plated. TS colonies grew with higher mean density than MS colonies (290 ± 11 versus 150 ± 11 cell per mm2; P = 0.0002). Expression of classical-mesenchymal stromal cell (MSC) markers was consistently recorded (>95%) from all tissue sources, whereas all the other markers were highly variable. CONCLUSIONS The prevalence and biological potential of CTPs are different between patients and tissue sources and lack variation in classical MSC markers. Other markers are more likely to discriminate differences between cell populations in biological performance. Understanding the underlying reasons for variation in the concentration, prevalence, marker expression and biological potential of CTPs between patients and source tissues and determining the means of managing this variation will contribute to the rational development of cell-based clinical diagnostics and targeted cell-based therapies.
Collapse
Affiliation(s)
- Maha A Qadan
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA; School of Biomedical Sciences, Kent State University, Kent, Ohio, USA; Department of Biotechnology and Genetic Engineering, Philadelphia University, Amman, Jordan
| | - Nicolas S Piuzzi
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA; Department of Orthopaedic Surgery, Cleveland Clinic Foundation, Cleveland, Ohio, USA; Instituto Universitario del Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Cynthia Boehm
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Wesley Bova
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Malcolm Moos
- FDA/Center for Biologics Evaluation and Research, Division of Cellular and Gene Therapies, Office of Cellular, Tissue, and Gene Therapies, Silver Spring, Maryland, USA
| | - Ronald J Midura
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Vincent C Hascall
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | | | - George F Muschler
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA; Department of Orthopaedic Surgery, Cleveland Clinic Foundation, Cleveland, Ohio, USA.
| |
Collapse
|
134
|
Ying SY, Fang W, Lin SL. The miR-302-Mediated Induction of Pluripotent Stem Cells (iPSC): Multiple Synergistic Reprogramming Mechanisms. Methods Mol Biol 2018; 1733:283-304. [PMID: 29435941 DOI: 10.1007/978-1-4939-7601-0_23] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Pluripotency represents a unique feature of embryonic stem cells (ESCs). To generate ESC-like-induced pluripotent stem cells (iPSCs) derived from somatic cells, the cell genome needs to be reset and reprogrammed to express the ESC-specific transcriptome. Numerous studies have shown that genomic DNA demethylation is required for epigenetic reprogramming of somatic cell nuclei to form iPSCs; yet, the mechanism remains largely unclear. In ESCs, the reprogramming process goes through two critical stages: germline and zygotic demethylation, both of which erase genomic DNA methylation sites and hence allow for different gene expression patterns to be reset into a pluripotent state. Recently, miR-302, an ESC-specific microRNA (miRNA), was found to play an essential role in four aspects of this reprogramming mechanism-(1) initiating global genomic DNA demethylation, (2) activating ESC-specific gene expression, (3) inhibiting developmental signaling, and (4) preventing stem cell tumorigenicity. In this review, we will summarize miR-302 functions in all four reprogramming aspects and further discuss how these findings may improve the efficiency and safety of the current iPSC technology.
Collapse
Affiliation(s)
- Shao-Yao Ying
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - William Fang
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shi-Lung Lin
- Division of Regenerative Medicine, WJWU & LYNN (W&L) Institute for Stem Cell Research, Santa Fe Springs, CA, USA
| |
Collapse
|
135
|
Abstract
MicroRNAs (miRNAs), widely distributed, small regulatory RNA genes, target both messenger RNA (mRNA) degradation and suppression of protein translation based on sequence complementarity between the miRNA and its targeted mRNA. Different names have been used to describe various types of miRNA. During evolution, RNA retroviruses or transgenes invaded the eukaryotic genome and were inserted itself in the noncoding regions of DNA, conceivably acting as transposon-like jumping genes, providing defense from viral invasion and fine-tuning of gene expression as a secondary level of gene modulation in eukaryotes. When a transposon is inserted in the intron, it becomes an intronic miRNA, taking advantage of the protein synthesis machinery, i.e., mRNA transcription and splicing, as a means for processing and maturation. MiRNAs have been found to play an important, but not life-threatening, role in embryonic development. They might play a pivotal role in diverse biological systems in various organisms, facilitating a quick response and accurate plotting of body physiology and structures. Based on these unique properties, manufactured intronic miRNAs have been developed for in vitro evaluation of gene function, in vivo gene therapy, and generation of transgenic animal models. The biogenesis of miRNAs, circulating miRNAs, miRNAs and cancer, iPSCs, and heart disease are presented in this chapter, highlighting some recent studies on these topics.
Collapse
Affiliation(s)
- Shao-Yao Ying
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Donald C Chang
- WJWU & LYNN Institute for Stem Cell Research, Santa Fe Springs, CA, USA
| | - Shi-Lung Lin
- Division of Regenerative Medicine, WJWU & LYNN Institute for Stem Cell Research, Santa Fe Springs, CA, USA
| |
Collapse
|
136
|
Abstract
Solid tumors are composed of mutually interacting cancer cells and tumor microenvironment. Many environmental components, such as extracellular matrix (ECM), mesenchymal stem cells, endothelial and immune cells, and various growth factors and cytokines, provide signals, either stimulatory or inhibitory, to cancer cells and determine their fates. Meanwhile, cancer cells can also educate surrounding cells or tissues to undergo changes that are in favorable of tumor progression. CD44, as a transmembrane receptor for hyaluronic acid (HA) and many other ECM components and a coreceptor for growth factors and cytokines, is a critical cell surface molecule that can sense, integrate, and transduce cellular microenvironmental signals to membrane-associated cytoskeletal proteins or to cell nucleus to regulate a variety of gene expressions that govern cell behaviors. Mounting evidence suggests that CD44, particularly CD44v isoforms, are cancer stem cell (CSC) markers and critical regulators of cancer stemness, including self-renewal, tumor initiation, and metastasis. Thus, CD44 is widely used alone or in combination with other cell surface markers to isolate or enrich CSCs through fluorescence-activated cell sorting of dissociated single cells that originate from the patient, xenograft tumor tissues, or tumor cell cultures. Sorted cells are cultured in a specialized culture medium for spheroid formation or inoculated into immunodeficient mice for the analysis of tumorigenic or metastatic potential. In this chapter, detailed experimental methods regarding CD44+ tumor cell isolation, spheroid culture, and characterization will be described.
Collapse
Affiliation(s)
- Liang Wang
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Unit 1466, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Xiangsheng Zuo
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Unit 956, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Keping Xie
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Unit 1466, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Daoyan Wei
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Unit 1466, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| |
Collapse
|
137
|
Wang Z, Zhao K, Hackert T, Zöller M. CD44/CD44v6 a Reliable Companion in Cancer-Initiating Cell Maintenance and Tumor Progression. Front Cell Dev Biol 2018; 6:97. [PMID: 30211160 PMCID: PMC6122270 DOI: 10.3389/fcell.2018.00097] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/08/2018] [Indexed: 12/19/2022] Open
Abstract
Metastasis is the leading cause of cancer death, tumor progression proceeding through emigration from the primary tumor, gaining access to the circulation, leaving the circulation, settling in distant organs and growing in the foreign environment. The capacity of a tumor to metastasize relies on a small subpopulation of cells in the primary tumor, so called cancer-initiating cells (CIC). CIC are characterized by sets of markers, mostly membrane anchored adhesion molecules, CD44v6 being the most frequently recovered marker. Knockdown and knockout models accompanied by loss of tumor progression despite unaltered primary tumor growth unraveled that these markers are indispensable for CIC. The unexpected contribution of marker molecules to CIC-related activities prompted research on underlying molecular mechanisms. This review outlines the contribution of CD44, particularly CD44v6 to CIC activities. A first focus is given to the impact of CD44/CD44v6 to inherent CIC features, including the crosstalk with the niche, apoptosis-resistance, and epithelial mesenchymal transition. Following the steps of the metastatic cascade, we report on supporting activities of CD44/CD44v6 in migration and invasion. These CD44/CD44v6 activities rely on the association with membrane-integrated and cytosolic signaling molecules and proteases and transcriptional regulation. They are not restricted to, but most pronounced in CIC and are tightly regulated by feedback loops. Finally, we discuss on the engagement of CD44/CD44v6 in exosome biogenesis, loading and delivery. exosomes being the main acteurs in the long-distance crosstalk of CIC with the host. In brief, by supporting the communication with the niche and promoting apoptosis resistance CD44/CD44v6 plays an important role in CIC maintenance. The multifaceted interplay between CD44/CD44v6, signal transducing molecules and proteases facilitates the metastasizing tumor cell journey through the body. By its engagement in exosome biogenesis CD44/CD44v6 contributes to disseminated tumor cell settlement and growth in distant organs. Thus, CD44/CD44v6 likely is the most central CIC biomarker.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Oncology, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong, China
| | - Kun Zhao
- Pancreas Section, University Hospital of Surgery, Heidelberg, Germany
| | - Thilo Hackert
- Pancreas Section, University Hospital of Surgery, Heidelberg, Germany
| | - Margot Zöller
- Pancreas Section, University Hospital of Surgery, Heidelberg, Germany
- *Correspondence: Margot Zöller
| |
Collapse
|
138
|
Bourguignon LYW. Hyaluronan-Mediated CD44 Signaling Activates Cancer Stem Cells in Head and Neck Cancer. CURRENT CANCER RESEARCH 2018:525-544. [DOI: 10.1007/978-3-319-78762-6_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
139
|
Wuebben EL, Wilder PJ, Cox JL, Grunkemeyer JA, Caffrey T, Hollingsworth MA, Rizzino A. SOX2 functions as a molecular rheostat to control the growth, tumorigenicity and drug responses of pancreatic ductal adenocarcinoma cells. Oncotarget 2017; 7:34890-906. [PMID: 27145457 PMCID: PMC5085197 DOI: 10.18632/oncotarget.8994] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 04/16/2016] [Indexed: 12/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly deadly malignancy. Expression of the stem cell transcription factor SOX2 increases during progression of PDAC. Knockdown of SOX2 in PDAC cell lines decreases growth in vitro; whereas, stable overexpression of SOX2 in one PDAC cell line reportedly increases growth in vitro. Here, we reexamined the role of SOX2 in PDAC cells, because inducible SOX2 overexpression in other tumor cell types inhibits growth. In this study, four PDAC cell lines were engineered for inducible overexpression of SOX2 or inducible knockdown of SOX2. Remarkably, inducible overexpression of SOX2 in PDAC cells inhibits growth in vitro and reduces tumorigenicity. Additionally, inducible knockdown of SOX2 in PDAC cells reduces growth in vitro and in vivo. Thus, growth and tumorigenicity of PDAC cells is highly dependent on the expression of optimal levels of SOX2 – a hallmark of molecular rheostats. We also determined that SOX2 alters the responses of PDAC cells to drugs used in PDAC clinical trials. Increasing SOX2 reduces growth inhibition mediated by MEK and AKT inhibitors; whereas knockdown of SOX2 further reduces growth when PDAC cells are treated with these inhibitors. Thus, targeting SOX2, or its mode of action, could improve the treatment of PDAC.
Collapse
Affiliation(s)
- Erin L Wuebben
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198-5950, USA
| | - Phillip J Wilder
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198-5950, USA
| | - Jesse L Cox
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska 68198-3135, USA
| | - James A Grunkemeyer
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198-5950, USA
| | - Thomas Caffrey
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198-5950, USA
| | - Michael A Hollingsworth
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198-5950, USA
| | - Angie Rizzino
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198-5950, USA.,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska 68198-5870, USA
| |
Collapse
|
140
|
Abstract
Cancer stem cells (CSC) are a prominent component of the tumor bulk and extensive research has now identified them as the subpopulation responsible for tumor relapse and resistance to anti-cancer treatments. Surrounding the bulk formed of tumor cells, an extracellular matrix contributes to cancer growth; the main component of the tumor micro-environment is hyaluronan, a large disaccharide forming a molecular network surrounding the cells. The hyaluronan-dependent coat can regulate cell division and motility in cancer progression and metastasis. One of the receptors of hyaluronan is CD44, a surface protein frequently used as a CSC marker. Indeed, tumor cells with high levels of CD44 appear to exhibit CSC properties and are characterized by elevated relapse rate. The CD44-hyaluronan-dependent interactions are Janus-faced: on one side, they have been shown to be crucial in both malignancy and resistance to therapy; on the other, they represent a potential value for future therapies, as disturbing the CD44-hyaluronan axis would not only impair the pericellular matrix but also the subpopulation of self-renewing oncogenic cells. Here, we will review the key roles of HA and CD44 in CSC maintenance and propagation and will show that CSC-like spheroids from a rabdhomyosarcoma cell line, namely RD, have a prominent pericellular coat necessary for sphere formation and for elevated migration. Thus, a better understanding of the hyaluronan-CD44 interactions holds the potential for ameliorating current cancer therapies and eradicating CSC.
Collapse
|
141
|
Rodrigo JP, Villaronga MÁ, Menéndez ST, Hermida-Prado F, Quer M, Vilaseca I, Allonca E, Pedregal Mallo D, Astudillo A, García-Pedrero JM. A Novel Role For Nanog As An Early Cancer Risk Marker In Patients With Laryngeal Precancerous Lesions. Sci Rep 2017; 7:11110. [PMID: 28894270 PMCID: PMC5594002 DOI: 10.1038/s41598-017-11709-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 08/29/2017] [Indexed: 11/24/2022] Open
Abstract
NANOG is a master regulator of embryonic stem cell pluripotency, found to be frequently aberrantly expressed in a variety of cancers, including laryngeal carcinomas. This study investigates for the first time the role of NANOG expression in early stages of laryngeal tumourigenesis and its potential utility as cancer risk marker. NANOG protein expression was evaluated by immunohistochemistry using two large independent cohorts of patients with laryngeal precancerous lesions, and correlated with clinicopathological parameters and laryngeal cancer risk. NANOG expression was detected by immunohistochemistry in 49 (60%) of 82 laryngeal dysplasias, whereas expression was negligible in patient-matched normal epithelia. Strong NANOG expression was found in 22 (27%) lesions and was established as cut-off point, showing the most robust association with laryngeal cancer risk (P = 0.003) superior to the histological classification (P = 0.320) the current gold standard in the clinical practice. Similar trends were obtained using a multicenter validation cohort of 86 patients with laryngeal dysplasia. Our findings uncover a novel role for NANOG expression in laryngeal tumourigenesis, and its unprecedented application as biomarker for cancer risk assessment.
Collapse
Affiliation(s)
- Juan P Rodrigo
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, Oviedo, CIBERONC, Spain.
| | - M Ángeles Villaronga
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, Oviedo, CIBERONC, Spain
| | - Sofía T Menéndez
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, Oviedo, CIBERONC, Spain
| | - Francisco Hermida-Prado
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, Oviedo, CIBERONC, Spain
| | - Miquel Quer
- Department of Otolaryngology, Hospital Santa Creu i Sant Pau, Barcelona, Spain
| | - Isabel Vilaseca
- Department of Otolaryngology, Hospital Clínic, Barcelona, Spain
| | - Eva Allonca
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, Oviedo, CIBERONC, Spain
| | - Daniel Pedregal Mallo
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, Oviedo, CIBERONC, Spain
| | - Aurora Astudillo
- Department of Pathology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Juana M García-Pedrero
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, Oviedo, CIBERONC, Spain.
| |
Collapse
|
142
|
Bourguignon LYW, Earle C, Shiina M. Activation of Matrix Hyaluronan-Mediated CD44 Signaling, Epigenetic Regulation and Chemoresistance in Head and Neck Cancer Stem Cells. Int J Mol Sci 2017; 18:ijms18091849. [PMID: 28837080 PMCID: PMC5618498 DOI: 10.3390/ijms18091849] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/21/2017] [Accepted: 08/22/2017] [Indexed: 12/21/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a solid tumor composed by a genotypically and phenotypically heterogeneous population of neoplastic cells types. High recurrence rate and regional metastases lead to major morbidity and mortality. Recently, many studies have focused on cellular and molecular mechanisms of tumor progression that can help to predict prognosis and to choose the best therapeutic approach for HNSCC patients. Hyaluronan (HA), an important glycosaminoglycan component of the extracellular matrix (ECM), and its major cell surface receptor, CD44, have been suggested to be important cellular mediators influencing tumor progression and treatment resistance in head and neck cancer. HNSCC contains a small subpopulation of cells that exhibit a hallmark of CD44-expressing cancer stem cell (CSC) properties with self-renewal, multipotency, and a unique potential for tumor initiation. HA has been shown to stimulate a variety of CSC functions including self-renewal, clone formation and differentiation. This review article will present current evidence for the existence of a unique small population of CD44v3highALDHhigh-expressing CSCs in HNSCC. A special focus will be placed on the role of HA/CD44-induced oncogenic signaling and histone methyltransferase, DOT1L activities in regulating histone modifications (via epigenetic changes) and miRNA activation. Many of these events are essential for the CSC properties such as Nanog/Oct4/Sox2 expression, spheroid/clone formation, self-renewal, tumor cell migration/invasion, survival and chemotherapeutic drug resistance in HA-activated head and neck cancer. These newly-discovered HA/CD44-mediated oncogenic signaling pathways delineate unique tumor dynamics with implications for defining the drivers of HNSCC progression processes. Most importantly, the important knowledge obtained from HA/CD44-regulated CSC signaling and functional activation could provide new information regarding the design of novel drug targets to overcome current therapeutic drug resistance which will have significant treatment implications for head and neck cancer patients.
Collapse
Affiliation(s)
- Lilly Y W Bourguignon
- San Francisco Veterans Affairs Medical Center and Department of Medicine, University of California at San Francisco & Endocrine Unit (111N2), 4150 Clement Street, San Francisco, CA 94121, USA.
| | - Christine Earle
- San Francisco Veterans Affairs Medical Center and Department of Medicine, University of California at San Francisco & Endocrine Unit (111N2), 4150 Clement Street, San Francisco, CA 94121, USA.
| | - Marisa Shiina
- San Francisco Veterans Affairs Medical Center and Department of Medicine, University of California at San Francisco & Endocrine Unit (111N2), 4150 Clement Street, San Francisco, CA 94121, USA.
| |
Collapse
|
143
|
Liang Y, Goyette S, Hyder SM. Cholesterol biosynthesis inhibitor RO 48-8071 reduces progesterone receptor expression and inhibits progestin-dependent stem cell-like cell growth in hormone-dependent human breast cancer cells. BREAST CANCER-TARGETS AND THERAPY 2017; 9:487-494. [PMID: 28744156 PMCID: PMC5511027 DOI: 10.2147/bctt.s140265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Clinical trials and studies have shown that postmenopausal women undergoing combination hormone replacement therapy containing estrogen and progestin have an increased risk of breast cancer compared with women taking estrogen or placebo alone. Using animal models, we have previously shown that synthetic progestins, including medroxyprogesterone acetate (MPA), which is widely used clinically, accelerate breast cancer tumor growth and promote metastasis. Furthermore, we have found that MPA elevates CD44 protein expression and aldehyde dehydrogenase (ALDH) activity, two markers of cancer stem cells (CSCs), and increases mammosphere formation, another hallmark of stem cells, in hormone-dependent T47-D human breast cancer cells. Herein, we show that RO 48-8071 (RO), an inhibitor of cholesterol synthesis, reduced MPA-induced CD44 protein expression in two hormone-dependent human breast cancer cell lines, T47-D and BT-474. Because we have previously shown that MPA induction of CD44 is progesterone receptor (PR) dependent, we examined RO’s effects on PR protein and mRNA expressions in T47-D cells. PR mRNA levels remained unchanged after RO treatment; however, RO significantly reduced the protein expression of both PR receptor isoforms, PR-A and PR-B. Using the proteasome inhibitor MG-132, we demonstrated that RO decreases PR protein expression in T47-D cells via the proteasomal degradation pathway. Importantly, treatment of T47-D cells with RO abolished MPA-induced mammosphere formation. Based on our observations, we contend that RO may represent a novel means of preventing MPA-induced CSC expansion. RO could be used clinically to both treat and prevent hormone-dependent breast cancers, which represent the majority of human breast cancers. RO may also have clinical utility in reducing resistance to antihormone therapy.
Collapse
Affiliation(s)
- Yayun Liang
- Department of Biomedical Sciences.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Sandy Goyette
- Department of Biomedical Sciences.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Salman M Hyder
- Department of Biomedical Sciences.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| |
Collapse
|
144
|
Abstract
Cancer stem cells (CSCs) have been identified in oral cavity squamous cell carcinoma (OCSCC). CSCs possess the ability for perpetual self-renewal and proliferation, producing downstream progenitor cells and cancer cells that drive tumor growth. Studies of many cancer types including OCSCC have identified CSCs using specific markers, but it is still unclear as to where in the stem cell hierarchy these markers fall. This is compounded further by the presence of multiple CSC subtypes within OCSCC, making investigation reliant on the use of multiple markers. This review examines the current knowledge in CSC markers OCT4, SOX2, NANOG, ALDH1, phosphorylated STAT3, CD44, CD24, CD133, and Musashi-1, specifically focusing on their use and validity in OCSCC CSC research and how they may be organized into the CSC hierarchy. OCSCC CSCs also express components of the renin–angiotensin system (RAS), which suggests CSCs may be novel therapeutic targets by modulation of the RAS using existing medications.
Collapse
Affiliation(s)
- Ranui Baillie
- Gillies McIndoe Research Institute, Wellington, New Zealand
| | - Swee T Tan
- Gillies McIndoe Research Institute, Wellington, New Zealand.,Wellington Regional Plastic, Maxillofacial and Burns Unit, Hutt Hospital, Wellington, New Zealand
| | | |
Collapse
|
145
|
Yamagata K, Fujiwara A, Onodera D, Motoki T. Lutein Regulates the Expression of Apoptosis-related Genes and Stem Cell Markers in A549 Human Lung Cancer Cells. Nat Prod Commun 2017. [DOI: 10.1177/1934578x1701200616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The BCL2 family has both pro-apoptotic and anti-apoptotic functions. Furthermore, stem cell markers such as Oct4, SOX2, and NANOG enhance cancer cells’ self-renewal, resistance to anti-cancer drugs and clonal growth. Therefore, selective inhibition of BCL2 genes and downregulated expression of stem cell markers should reduce the survival of cancer cells. Previous studies have reported that lutein, a carotenoid pigment present in fruits and vegetables, can inhibit cancer cells. However, the inhibitory effects of lutein on cancer cells have not been investigated sufficiently. In this study, we used gene expression analysis by polymerase chain reaction (PCR) and Western blotting to show that lutein regulates the expression of genes involved in apoptosis and several stem cell marker genes in a human lung cancer cell line, A549. Lutein induced gene expression of pro-apoptotic BAX and CAS3 and reduced the level of the anti-apoptotic gene BCL2. Furthermore, protein expression of BCL2 and BAX was regulated by treatment with lutein. Lutein also inhibited SOX2 and NANOG gene expression in A549, but not POU5F1. In addition, lutein reduced gene expression of SLCA11, but induced CD44 and CD133 gene expression. These results indicated that lutein inhibits several events associated with apoptosis regulation in a BCL2 family-dependent pathway.
Collapse
Affiliation(s)
- Kazuo Yamagata
- Department of Food Bioscience and Biotechnology, College of Bioresource Sciences, Nihon University (NUBS), 1866, Kameino, Fujisawa, Kanagawa, Japan
| | - Ayame Fujiwara
- Department of Food Bioscience and Biotechnology, College of Bioresource Sciences, Nihon University (NUBS), 1866, Kameino, Fujisawa, Kanagawa, Japan
| | - Daiki Onodera
- Department of Food Bioscience and Biotechnology, College of Bioresource Sciences, Nihon University (NUBS), 1866, Kameino, Fujisawa, Kanagawa, Japan
| | - Tagami Motoki
- Department of Internal Medicine, Sanraku Hospital, Kanda-Surugadai 2–5, Chiyoda-ku, Tokyo, Japan
| |
Collapse
|
146
|
Goyette S, Liang Y, Mafuvadze B, Cook MT, Munir M, Hyder SM. Natural and synthetic progestins enrich cancer stem cell-like cells in hormone-responsive human breast cancer cell populations in vitro. BREAST CANCER-TARGETS AND THERAPY 2017; 9:347-357. [PMID: 28579829 PMCID: PMC5446973 DOI: 10.2147/bctt.s135371] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Clinical trials and studies have shown that combination estrogen/progestin hormone replacement therapy, but not estrogen therapy alone or placebo, increases breast cancer risk in postmenopausal women. Using animal models, we have previously shown that both natural and synthetic progestins (including medroxyprogesterone acetate [MPA], a synthetic progestin used widely in the clinical setting) accelerate the development of breast tumors in vivo and increase their metastasis to lymph nodes. Based on these observations, we have hypothesized that progestin-induced breast cancer tumor growth and metastasis may be mediated by an enrichment of the cancer stem cell (CSC) pool. In this study, we used T47-D and BT-474 hormone-responsive human breast cancer cells to examine the effects of progestin on phenotypic and functional markers of CSCs in vitro. Both natural and synthetic progestins (10 nM) significantly increased protein expression of CD44, an important CSC marker in tumor cells. MPA increased the levels of both CD44 variants v3 and v6 associated with stem cell functions. This induction of CD44 was blocked by the antiprogestin RU-486, suggesting that this process is progesterone receptor (PR) dependent. CD44 induction was chiefly progestin dependent. Because RU-486 can bind other steroid receptors, we treated PR-negative T47-DCO-Y cells with MPA and found that MPA failed to induce CD44 protein expression, confirming that PR is essential for progestin-mediated CD44 induction in T47-D cells. Further, MPA treatment of T47-D cells significantly increased the activity of aldehyde dehydrogenase (ALDH), another CSC marker. Finally, two synthetic progestins, MPA and norethindrone, significantly increased the ability of T47-D cells to form mammospheres, suggesting that enrichment of the CD44high, ALDHbright subpopulation of cancer cells induced by MPA exposure is of functional significance. Based on our observations, we contend that exposure of breast cancer cells to synthetic progestins leads to an enrichment of the CSC pool, supporting the development of progestin-accelerated tumors in vivo.
Collapse
Affiliation(s)
- Sandy Goyette
- Department of Biomedical Sciences.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Yayun Liang
- Department of Biomedical Sciences.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Benford Mafuvadze
- Department of Biomedical Sciences.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Matthew T Cook
- Department of Biomedical Sciences.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Moiz Munir
- Department of Biomedical Sciences.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Salman M Hyder
- Department of Biomedical Sciences.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| |
Collapse
|
147
|
Liu CW, Hua KT, Li KC, Kao HF, Hong RL, Ko JY, Hsiao M, Kuo ML, Tan CT. Histone Methyltransferase G9a Drives Chemotherapy Resistance by Regulating the Glutamate-Cysteine Ligase Catalytic Subunit in Head and Neck Squamous Cell Carcinoma. Mol Cancer Ther 2017; 16:1421-1434. [PMID: 28265008 DOI: 10.1158/1535-7163.mct-16-0567-t] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 10/17/2016] [Accepted: 02/20/2017] [Indexed: 11/16/2022]
Abstract
Transient chemotherapeutic response is a major obstacle to treating head and neck squamous cell carcinomas (HNSCC). Histone methyltransferase G9a has recently been shown to be abundantly expressed in HNSCC, and is required to maintain the malignant phenotype. In this study, we found that high G9a expression is significantly associated with poor chemotherapeutic response and disease-free survival in HNSCC patients. Similarly, G9a expression and enzymatic activity were elevated in cisplatin-resistant HNSCC cells. Genetic or pharmacologic inhibition of G9a sensitized the resistant cells to cisplatin, increasing cellular apoptosis. Mechanistic investigations indicated that G9a contributes to transcriptional activation of the glutamate-cysteine ligase catalytic subunit (GCLC), which results in upregulation of cellular glutathione (GSH) and drug resistance. In addition, we observed a significant positive correlation between G9a and GCLC expression in tumors of HNSCC patients. Taken together, our findings provide evidence that G9a protects HNSCC cells against chemotherapy by increasing the synthesis of GSH, and imply G9a as a promising target for overcoming cisplatin resistance in HNSCC. Mol Cancer Ther; 16(7); 1421-34. ©2017 AACR.
Collapse
Affiliation(s)
- Chia-Wen Liu
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Otolaryngology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Kuo-Tai Hua
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kai-Chun Li
- Graduate Institute of Biomedical Sciences, College of Life Sciences, National Taiwan University, Taipei, Taiwan
| | - Hsiang-Fong Kao
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan.,National Taiwan University Cancer Center, Taipei, Taiwan
| | - Ruey-Long Hong
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Jenq-Yuh Ko
- Department of Otolaryngology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Michael Hsiao
- The Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Min-Liang Kuo
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Biomedical Sciences, College of Life Sciences, National Taiwan University, Taipei, Taiwan
| | - Ching-Ting Tan
- Department of Otolaryngology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan. .,National Taiwan University Cancer Center, Taipei, Taiwan
| |
Collapse
|
148
|
Autocrine and Paracrine Mechanisms Promoting Chemoresistance in Cholangiocarcinoma. Int J Mol Sci 2017; 18:ijms18010149. [PMID: 28098760 PMCID: PMC5297782 DOI: 10.3390/ijms18010149] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 12/19/2016] [Accepted: 01/06/2017] [Indexed: 02/07/2023] Open
Abstract
Resistance to conventional chemotherapeutic agents, a typical feature of cholangiocarcinoma, prevents the efficacy of the therapeutic arsenal usually used to combat malignancy in humans. Mechanisms of chemoresistance by neoplastic cholangiocytes include evasion of drug-induced apoptosis mediated by autocrine and paracrine cues released in the tumor microenvironment. Here, recent evidence regarding molecular mechanisms of chemoresistance is reviewed, as well as associations between well-developed chemoresistance and activation of the cancer stem cell compartment. It is concluded that improved understanding of the complex interplay between apoptosis signaling and the promotion of cell survival represent potentially productive areas for active investigation, with the ultimate aim of encouraging future studies to unveil new, effective strategies able to overcome current limitations on treatment.
Collapse
|
149
|
Naik PP, Das DN, Panda PK, Mukhopadhyay S, Sinha N, Praharaj PP, Agarwal R, Bhutia SK. Implications of cancer stem cells in developing therapeutic resistance in oral cancer. Oral Oncol 2016; 62:122-135. [PMID: 27865365 DOI: 10.1016/j.oraloncology.2016.10.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 10/05/2016] [Accepted: 10/15/2016] [Indexed: 12/13/2022]
Abstract
Conventional therapeutics are often frequented with recurrences, refraction and regimen resistance in oral cavity cancers which are predominantly manifested by cancer stem cells (CSCs). During oncoevolution, cancer cells may undergo structural and functional reprogramming wherein they evolve as highly tolerant CSC phenotypes with greater survival advantages. The CSCs possess inherent and exclusive properties including self-renewal, hierarchical differentiation, and tumorigenicity that serve as the basis of chemo-radio-resistance in oral cancer. However, the key mechanisms underlying the CSC-mediated therapy resistance need to be further elucidated. A spectrum of dysfunctional cellular pathways including the developmental signaling, apoptosis, autophagy, cell cycle regulation, DNA damage responses and epigenetic regulations protect the CSCs from conventional therapies. Moreover, tumor niche shelters CSCs and creates an immunosuppressive environment favoring the survival of CSCs. Maintenance of lower redox status, epithelial-to-mesenchymal transition (EMT), metabolic reprogramming and altered drug responses are the accessory features that aid in the process of chemo-radio-resistance in oral CSCs. This review deals with the functional and molecular basis of cancer cell pluripotency-associated resistance highlighting the abrupt fundamental cellular processes; targeting these events may hold a great promise in the successful treatment of oral cancer.
Collapse
Affiliation(s)
- Prajna Paramita Naik
- Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Durgesh Nandini Das
- Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Prashanta Kumar Panda
- Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Subhadip Mukhopadhyay
- Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Niharika Sinha
- Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | | | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO, United States; University of Colorado Cancer Center, University of Colorado Denver, Aurora, CO, United States.
| | - Sujit Kumar Bhutia
- Department of Life Science, National Institute of Technology, Rourkela, Odisha, India.
| |
Collapse
|
150
|
Roles and targeting of the HAS/hyaluronan/CD44 molecular system in cancer. Matrix Biol 2016; 59:3-22. [PMID: 27746219 DOI: 10.1016/j.matbio.2016.10.001] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/02/2016] [Accepted: 10/03/2016] [Indexed: 02/07/2023]
Abstract
Synthesis, deposition, and interactions of hyaluronan (HA) with its cellular receptor CD44 are crucial events that regulate the onset and progression of tumors. The intracellular signaling pathways initiated by HA interactions with CD44 leading to tumorigenic responses are complex. Moreover, HA molecules may perform dual functions depending on their concentration and size. Overexpression of variant isoforms of CD44 (CD44v) is most commonly linked to cancer progression, whereas their loss is associated with inhibition of tumor growth. In this review, we highlight that the regulation of HA synthases (HASes) by post-translational modifications, such as O-GlcNAcylation and ubiquitination, environmental factors and the action of microRNAs is important for HA synthesis and secretion in the tumor microenvironment. Moreover, we focus on the roles and interactions of CD44 with various proteins that reside extra- and intracellularly, as well as on cellular membranes with particular reference to the CD44-HA axis in cancer stem cell functions, and the importance of CD44/CD44v6 targeting to inhibit tumorigenesis.
Collapse
|