151
|
Santos Franco S, Raveh-Amit H, Kobolák J, Alqahtani MH, Mobasheri A, Dinnyes A. The crossroads between cancer stem cells and aging. BMC Cancer 2015; 15 Suppl 1:S1. [PMID: 25708542 PMCID: PMC4331724 DOI: 10.1186/1471-2407-15-s1-s1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The cancer stem cell (CSC) hypothesis suggests that only a subpopulation of cells within a tumour is responsible for the initiation and progression of neoplasia. The original and best evidence for the existence of CSCs came from advances in the field of haematological malignancies. Thus far, putative CSCs have been isolated from various solid and non-solid tumours and shown to possess self-renewal, differentiation, and cancer regeneration properties. Although research in the field is progressing extremely fast, proof of concept for the CSC hypothesis is still lacking and key questions remain unanswered, e.g. the cell of origin for these cells. Nevertheless, it is undisputed that neoplastic transformation is associated with genetic and epigenetic alterations of normal cells, and a better understanding of these complex processes is of utmost importance for developing new anti-cancer therapies. In the present review, we discuss the CSC hypothesis with special emphasis on age-associated alterations that govern carcinogenesis, at least in some types of tumours. We present evidence from the scientific literature for age-related genetic and epigenetic alterations leading to cancer and discuss the main challenges in the field.
Collapse
|
152
|
A differential role for CXCR4 in the regulation of normal versus malignant breast stem cell activity. Oncotarget 2015; 5:599-612. [PMID: 24583601 PMCID: PMC3996659 DOI: 10.18632/oncotarget.1169] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
C-X-C chemokine receptor type 4 (CXCR4) is known to regulate lung, pancreatic and prostate cancer stem cells. In breast cancer, CXCR4 signalling has been reported to be a mediator of metastasis, and is linked to poor prognosis. However its role in normal and malignant breast stem cell function has not been investigated. Anoikis resistant (AR) cells were collected from immortalised (MCF10A, 226L) and malignant (MCF7, T47D, SKBR3) breast cell lines and assessed for stem cell enrichment versus unsorted cells. AR cells had significantly higher mammosphere forming efficiency (MFE) than unsorted cells. The AR normal cells demonstrated increased formation of 3D structures in Matrigel compared to unsorted cells. In vivo, SKBR3 and T47D AR cells had 7- and 130-fold enrichments for tumour formationrespectively, compared with unsorted cells. AR cells contained significantly elevated CXCR4 transcript and protein levels compared to unsorted cells. Importantly, CXCR4 mRNA was higher in stem cell-enriched CD44+/CD24- patient-derived breast cancer cells compared to non-enriched cells. CXCR4 stimulation by its ligand SDF-1 reduced MFE of the normal breast cells lines but increased the MFE in T47D and patient-derived breast cancer cells. CXCR4 inhibition by AMD3100 increased stem cell activity but reduced the self-renewal capacity of the malignant breast cell line T47D. CXCR4+ FACS sorted MCF7 cells demonstrated a significantly increased MFE compared with CXCR4- cells. This significant increase in MFE was further demonstrated in CXCR4 over-expressing MCF7 cells which also had an increase in self-renewal compared to parental cells. A greater reduction in self-renewal following CXCR4 inhibition in the CXCR4 over-expressing cells compared with parental cells was also observed. Our data establish for the first time that CXCR4 signalling has contrasting effects on normal and malignant breast stem cell activity. Here, we demonstrate that CXCR4 signalling specifically regulates breast cancer stem cell activities and may therefore be important in tumour formation at the sites of metastases.
Collapse
|
153
|
Masyuk M, Brand-Saberi B. Recruitment of skeletal muscle progenitors to secondary sites: a role for CXCR4/SDF-1 signalling in skeletal muscle development. Results Probl Cell Differ 2015; 56:1-23. [PMID: 25344664 DOI: 10.1007/978-3-662-44608-9_1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
During embryonic development, myogenesis occurs in different functional muscle groups at different time points depending on the availability of their final destinations. Primary trunk muscle consists of the intrinsic dorsal (M. erector spinae) and ventral (cervical, thoracic, abdominal) muscles. In contrast, secondary trunk muscles are established from progenitor cells that have migrated initially from the somites into the limb buds and thereafter returned to the trunk. Furthermore, craniofacial muscle constitutes a group that originates from four different sources and employs a different set of regulatory molecules. Development of muscle groups at a distance from their origins involves the maintenance of a pool of progenitor cells capable of proliferation and directed cell migration. We review here the data concerning somite-derived progenitor cell migration to the limbs and subsequent retrograde migration in the establishment of secondary trunk muscle in chicken and mouse. We review the function of SDF-1 and CXCR4 in the control of this process referring to our previous work in shoulder muscle and cloacal/perineal muscle development. Some human anatomical variations and malformations of secondary trunk muscles are discussed.
Collapse
Affiliation(s)
- Maryna Masyuk
- Department of Anatomy and Molecular Embryology, Ruhr-Universität Bochum, Universitätsstraße 150, MA 5/161, 44801, Bochum, Germany,
| | | |
Collapse
|
154
|
Sandberg CJ, Vik-Mo EO, Behnan J, Helseth E, Langmoen IA. Transcriptional profiling of adult neural stem-like cells from the human brain. PLoS One 2014; 9:e114739. [PMID: 25514637 PMCID: PMC4267785 DOI: 10.1371/journal.pone.0114739] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 11/13/2014] [Indexed: 12/11/2022] Open
Abstract
There is a great potential for the development of new cell replacement strategies based on adult human neural stem-like cells. However, little is known about the hierarchy of cells and the unique molecular properties of stem- and progenitor cells of the nervous system. Stem cells from the adult human brain can be propagated and expanded in vitro as free floating neurospheres that are capable of self-renewal and differentiation into all three cell types of the central nervous system. Here we report the first global gene expression study of adult human neural stem-like cells originating from five human subventricular zone biopsies (mean age 42, range 33–60). Compared to adult human brain tissue, we identified 1,189 genes that were significantly up- and down-regulated in adult human neural stem-like cells (1% false discovery rate). We found that adult human neural stem-like cells express stem cell markers and have reduced levels of markers that are typical of the mature cells in the nervous system. We report that the genes being highly expressed in adult human neural stem-like cells are associated with developmental processes and the extracellular region of the cell. The calcium signaling pathway and neuroactive ligand-receptor interactions are enriched among the most differentially regulated genes between adult human neural stem-like cells and adult human brain tissue. We confirmed the expression of 10 of the most up-regulated genes in adult human neural stem-like cells in an additional sample set that included adult human neural stem-like cells (n = 6), foetal human neural stem cells (n = 1) and human brain tissues (n = 12). The NGFR, SLITRK6 and KCNS3 receptors were further investigated by immunofluorescence and shown to be heterogeneously expressed in spheres. These receptors could potentially serve as new markers for the identification and characterisation of neural stem- and progenitor cells or as targets for manipulation of cellular fate.
Collapse
Affiliation(s)
- Cecilie Jonsgar Sandberg
- Vilhelm Magnus Lab, Institute for Surgical Research and Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
- * E-mail:
| | - Einar O. Vik-Mo
- Vilhelm Magnus Lab, Institute for Surgical Research and Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
| | - Jinan Behnan
- Vilhelm Magnus Lab, Institute for Surgical Research and Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
| | - Eirik Helseth
- Vilhelm Magnus Lab, Institute for Surgical Research and Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
| | - Iver A. Langmoen
- Vilhelm Magnus Lab, Institute for Surgical Research and Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
- Cancer Stem Cell Innovation Center (CAST), Oslo University Hospital and University of Oslo, Oslo, Norway
- Norwegian Stem Cell Center, Oslo University Hospital and University of Oslo, Oslo, Norway
| |
Collapse
|
155
|
In vitro modeling of the prostate cancer microenvironment. Adv Drug Deliv Rev 2014; 79-80:214-21. [PMID: 24816064 DOI: 10.1016/j.addr.2014.04.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 04/15/2014] [Accepted: 04/29/2014] [Indexed: 01/01/2023]
Abstract
Prostate cancer is the most commonly diagnosed malignancy in men and advanced disease is incurable. Model systems are a fundamental tool for research and many in vitro models of prostate cancer use cancer cell lines in monoculture. Although these have yielded significant insight they are inherently limited by virtue of their two-dimensional (2D) growth and inability to include the influence of tumour microenvironment. These major limitations can be overcome with the development of newer systems that more faithfully recreate and mimic the complex in vivo multi-cellular, three-dimensional (3D) microenvironment. This article presents the current state of in vitro models for prostate cancer, with particular emphasis on 3D systems and the challenges that remain before their potential to advance our understanding of prostate disease and aid in the development and testing of new therapeutic agents can be realised.
Collapse
|
156
|
Differentiation of human breast-milk stem cells to neural stem cells and neurons. Neurol Res Int 2014; 2014:807896. [PMID: 25506428 PMCID: PMC4260437 DOI: 10.1155/2014/807896] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 10/08/2014] [Accepted: 10/20/2014] [Indexed: 12/24/2022] Open
Abstract
Objectives. Human breast milk contains a heterogeneous population of cells that have the potential to provide a noninvasive source of cells for cell therapy in many neurodegenerative diseases without any ethical concern. The objectives of this study were to differentiate the breast milk-derived stem cells (BMDSC) toward neural stem cells and then into the neurons and neuroglia. Materials and Methods. To do this, the BMDSC were isolated from human breast milk and cultured in Dulbecco's modified Eagle medium/F12 (DMEM/F12) containing fibroblast growth factor (bFGF). The cells were then characterized by evaluation of the embryonic and stem cell markers. Then, the cells were exposed to culture medium containing 1% B27 and 2% N2 for 7–10 days followed by medium supplemented with B27, N2, bFGF 10 µg/mL, and endothelial growth factor (EGF) 20 µg/mL. Then, the sphere-forming assay was performed. The spheres were then differentiated into three neural lineages by withdrawing growth factor in the presence of 5% FBS (fetal bovine serum). The immunofluorescence was done for β-tubulin III, O4, and GFAP (glial fibrillary acidic protein). Results. The results indicated that the cells expressed both embryonic and mesenchymal stem cell (MSC) markers. They also showed neurospheres formation that was nestin-positive. The cells were also differentiated into all three neural lineages. Conclusion. The BMDSC can behave in the same way with neural stem cells. They were differentiated into oligodendrocytes, and astrocytes as well as neurons.
Collapse
|
157
|
Deng L, Shang L, Bai S, Chen J, He X, Martin-Trevino R, Chen S, Li XY, Meng X, Yu B, Wang X, Liu Y, McDermott SP, Ariazi AE, Ginestier C, Ibarra I, Ke J, Luther T, Clouthier SG, Xu L, Shan G, Song E, Yao H, Hannon GJ, Weiss SJ, Wicha MS, Liu S. MicroRNA100 inhibits self-renewal of breast cancer stem-like cells and breast tumor development. Cancer Res 2014; 74:6648-60. [PMID: 25217527 PMCID: PMC4370193 DOI: 10.1158/0008-5472.can-13-3710] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
miRNAs are essential for self-renewal and differentiation of normal and malignant stem cells by regulating the expression of key stem cell regulatory genes. Here, we report evidence implicating the miR100 in self-renewal of cancer stem-like cells (CSC). We found that miR100 expression levels relate to the cellular differentiation state, with lowest expression in cells displaying stem cell markers. Utilizing a tetracycline-inducible lentivirus to elevate expression of miR100 in human cells, we found that increasing miR100 levels decreased the production of breast CSCs. This effect was correlated with an inhibition of cancer cell proliferation in vitro and in mouse tumor xenografts due to attenuated expression of the CSC regulatory genes SMARCA5, SMARCD1, and BMPR2. Furthermore, miR100 induction in breast CSCs immediately upon their orthotopic implantation or intracardiac injection completely blocked tumor growth and metastasis formation. Clinically, we observed a significant association between miR100 expression in breast cancer specimens and patient survival. Our results suggest that miR100 is required to direct CSC self-renewal and differentiation.
Collapse
Affiliation(s)
- Lu Deng
- Innovation Center for Cell Biology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Li Shang
- Comprehensive Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Shoumin Bai
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ji Chen
- Innovation Center for Cell Biology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Xueyan He
- Innovation Center for Cell Biology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Rachel Martin-Trevino
- Comprehensive Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Shanshan Chen
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiao-Yan Li
- Division of Molecular Medicine and Genetics, Department of Internal Medicine and Life Sciences Institute, University of Michigan, Ann Arbor, Michigan
| | - Xiaojie Meng
- Departments of Molecular Biosciences and Radiation Oncology, University of Kansas Cancer Center, University of Kansas Medical School, University of Kansas, Lawrence, Kansas
| | - Bin Yu
- Innovation Center for Cell Biology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaolin Wang
- Innovation Center for Cell Biology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Yajing Liu
- Comprehensive Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Sean P McDermott
- Comprehensive Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Alexa E Ariazi
- Comprehensive Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Christophe Ginestier
- Centre de Recherche en Cance'rologie de Marseille, Laboratoire d'Oncologie Mole'culaire, UMR891 Inserm/Institut Paoli-Calmettes, Universite' de la Me'diterrane'e, Marseille, France
| | - Ingrid Ibarra
- Cold Spring Harbor Laboratory, Program in Genetics and Bioinformatics, Cold Spring Harbor, New York, New York
| | - Jia Ke
- Department of Colorectal Surgery, Sixth Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tahra Luther
- Comprehensive Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Shawn G Clouthier
- Comprehensive Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Liang Xu
- Departments of Molecular Biosciences and Radiation Oncology, University of Kansas Cancer Center, University of Kansas Medical School, University of Kansas, Lawrence, Kansas
| | - Ge Shan
- Innovation Center for Cell Biology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Erwei Song
- Department of Breast Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Herui Yao
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Gregory J Hannon
- Cold Spring Harbor Laboratory, Program in Genetics and Bioinformatics, Cold Spring Harbor, New York, New York
| | - Stephen J Weiss
- Division of Molecular Medicine and Genetics, Department of Internal Medicine and Life Sciences Institute, University of Michigan, Ann Arbor, Michigan
| | - Max S Wicha
- Comprehensive Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Suling Liu
- Innovation Center for Cell Biology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
158
|
Endothelial cells provide a notch-dependent pro-tumoral niche for enhancing breast cancer survival, stemness and pro-metastatic properties. PLoS One 2014; 9:e112424. [PMID: 25380486 PMCID: PMC4224483 DOI: 10.1371/journal.pone.0112424] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 10/15/2014] [Indexed: 01/05/2023] Open
Abstract
Treating metastasis has been challenging due to tumors complexity and heterogeneity. This complexity is partly related to the crosstalk between tumor and its microenvironment. Endothelial cells -the building blocks of tumor vasculature- have been shown to have additional roles in cancer progression than angiogenesis and supplying oxygen and nutrients. Here, we show an alternative role for endothelial cells in supporting breast cancer growth and spreading independent of their vascular functions. Using endothelial cells and breast cancer cell lines MDA-MB231 and MCF-7, we developed co-culture systems to study the influence of tumor endothelium on breast tumor development by both in vitro and in vivo approaches. Our results demonstrated that endothelial cells conferred survival advantage to tumor cells under complete starvation and enriched the CD44HighCD24Low/- stem cell population in tumor cells. Moreover, endothelial cells enhanced the pro-metastatic potential of breast cancer cells. The in vitro and in vivo results concordantly confirmed a role for endothelial Jagged1 to promote breast tumor through notch activation. Here, we propose a role for endothelial cells in enhancing breast cancer progression, stemness, and pro-metastatic traits through a perfusion-independent manner. Our findings may be beneficial in developing novel therapeutic approaches.
Collapse
|
159
|
Lagadec C, Vlashi E, Frohnen P, Alhiyari Y, Chan M, Pajonk F. The RNA-binding protein Musashi-1 regulates proteasome subunit expression in breast cancer- and glioma-initiating cells. Stem Cells 2014; 32:135-44. [PMID: 24022895 DOI: 10.1002/stem.1537] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 03/10/2013] [Accepted: 08/17/2013] [Indexed: 01/11/2023]
Abstract
Cancer stem cells (CSCs) or tumor-initiating cells, similar to normal tissue stem cells, rely on developmental pathways, such as the Notch pathway, to maintain their stem cell state. One of the regulators of the Notch pathway is Musashi-1, a mRNA-binding protein. Musashi-1 promotes Notch signaling by binding to the mRNA of Numb, the negative regulator of Notch signaling, thus preventing its translation. CSCs have also been shown to downregulate their 26S proteasome activity in several types of solid tumors, thus making them resistant to proteasome-inhibitors used as anticancer agents in the clinic. Interestingly, the Notch pathway can be inhibited by proteasomal degradation of the Notch intracellular domain (Notch-ICD); therefore, downregulation of the 26S proteasome activity can lead to stabilization of Notch-ICD. Here, we present evidence that the downregulation of the 26S proteasome in CSCs constitutes another level of control by which Musashi-1 promotes signaling through the Notch pathway and maintenance of the stem cell phenotype of this subpopulation of cancer cells. We demonstrate that Musashi-1 mediates the downregulation of the 26S proteasome by binding to the mRNA of NF-YA, the transcriptional factor regulating 26S proteasome subunit expression, thus providing an additional route by which the degradation of Notch-ICD is prevented, and Notch signaling is sustained.
Collapse
Affiliation(s)
- Chann Lagadec
- Department of Radiation Oncology, David Geffen School of Medicine and, University of California Los Angeles, California, USA
| | | | | | | | | | | |
Collapse
|
160
|
Expression of OCT4A: the first step to the next stage of urothelial bladder cancer progression. Int J Mol Sci 2014; 15:16069-82. [PMID: 25216339 PMCID: PMC4200762 DOI: 10.3390/ijms150916069] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 09/02/2014] [Accepted: 09/03/2014] [Indexed: 12/20/2022] Open
Abstract
OCT4 (octamer-binding transcription factor) is a transcription factor responsible for maintaining the pluripotent properties of embryonic stem cells. In this paper, we present the results of studies to investigate the role of the OCT4 splicing variant in urothelial bladder cancer and the relationship between the OCT4 phenotype and the morphological parameters of tumor malignancy. Ninety patients who received a cystectomy for bladder cancer were enrolled. The expression of OCT4 protein was analyzed by immunohistochemistry. The ratio of OCT4-positive cells was the lowest in pT1 (pathological assessment (p)--tumor extent confined to mucosa (T1)) tumors and the highest in pTis (non-papillary tumor extent confined to urothelium) and pT2 (tumor extent including muscularis propria) tumors. Information about the percentage of OCT4A-positive tumor cells could facilitate choosing the treatment mode in borderline pTis-pT1 (crossing the border of the basement membrane; the first stage of progression) and pT1-pT2 (crossing the border of the muscularis propria; the second stage of progression) cases: a higher percentage of OCT4A-positive cells should support more radical therapy. A significantly higher percentage of cases with moderate OCT4 intensity was found in metastasizing (the third stage of progression) cases with >2 positive lymph nodes. The percentage of OCT4-positive cells was significantly higher for cancers with a high grade, higher non-classic differentiation number and greater aggressiveness of invasion. The differentiation, maturation and aggressiveness of tumor invasion appear to depend on the expression of the OCT4 phenotype in cancer cells, similar to the successive stages of malignancy progression in urothelial cancer.
Collapse
|
161
|
Prick J, de Haan G, Green AR, Kent DG. Clonal heterogeneity as a driver of disease variability in the evolution of myeloproliferative neoplasms. Exp Hematol 2014; 42:841-51. [PMID: 25201757 DOI: 10.1016/j.exphem.2014.07.268] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/23/2014] [Accepted: 07/24/2014] [Indexed: 01/01/2023]
Abstract
Myeloproliferative neoplasms (MPNs) are clonal hematological diseases in which cells of the myelo-erythroid lineage are overproduced and patients are predisposed to leukemic transformation. Hematopoietic stem cells are the suspected disease-initiating cells, and these cells must acquire a clonal advantage relative to nonmutant hematopoietic stem cells to perpetuate disease. In 2005, several groups identified a single gain-of-function point mutation in JAK2 that associated with the majority of MPNs, and subsequent studies have led to a comprehensive understanding of the mutational landscape in MPNs. However, confusion still exists as to how a single genetic aberration can be associated with multiple distinct disease entities. Many explanations have been proposed, including JAK2V617F homozygosity, individual patient heterogeneity, and the differential regulation of downstream JAK2 signaling pathways. Several groups have made knock-in mouse models expressing JAK2V617F and have observed divergent phenotypes, each recapitulating some aspects of disease. Intriguingly, most of these models do not observe a strong hematopoietic stem cell self-renewal advantage compared with wild-type littermate controls, raising the question of how a clonal advantage is established in patients with MPNs. This review summarizes the current molecular understanding of MPNs and the diversity of disease phenotypes and proposes that the increased proliferation induced by JAK2V617F applies a selection pressure on the mutant clone that results in highly diverse clonal evolution in individuals.
Collapse
Affiliation(s)
- Janine Prick
- Cambridge Institute for Medical Research and Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom; Laboratory of Ageing Biology and Stem Cells, European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gerald de Haan
- Laboratory of Ageing Biology and Stem Cells, European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Anthony R Green
- Cambridge Institute for Medical Research and Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom; Department of Haematology, University of Cambridge, Cambridge, United Kingdom; Department of Haematology, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - David G Kent
- Cambridge Institute for Medical Research and Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom; Department of Haematology, University of Cambridge, Cambridge, United Kingdom; Department of Haematology, Addenbrooke's Hospital, Cambridge, United Kingdom.
| |
Collapse
|
162
|
The mammary cellular hierarchy and breast cancer. Cell Mol Life Sci 2014; 71:4301-24. [PMID: 25080108 PMCID: PMC4207940 DOI: 10.1007/s00018-014-1674-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Revised: 05/29/2014] [Accepted: 06/23/2014] [Indexed: 12/26/2022]
Abstract
Advances in the study of hematopoietic cell maturation have paved the way to a deeper understanding the stem and progenitor cellular hierarchy in the mammary gland. The mammary epithelium, unlike the hematopoietic cellular hierarchy, sits in a complex niche where communication between epithelial cells and signals from the systemic hormonal milieu, as well as from extra-cellular matrix, influence cell fate decisions and contribute to tissue homeostasis. We review the discovery, definition and regulation of the mammary cellular hierarchy and we describe the development of the concepts that have guided our investigations. We outline recent advances in in vivo lineage tracing that is now challenging many of our assumptions regarding the behavior of mammary stem cells, and we show how understanding these cellular lineages has altered our view of breast cancer.
Collapse
|
163
|
Zhang M, Li Z, Zhang X, Chang Y. Cancer stem cells as a potential therapeutic target in breast cancer. Stem Cell Investig 2014; 1:14. [PMID: 27358860 DOI: 10.3978/j.issn.2306-9759.2014.06.01] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 06/23/2014] [Indexed: 01/06/2023]
Abstract
Cancer stem cells (CSCs) are defined as the small population of cancer cells that have stem cell properties as in hierarchically organized tumors. They are considered as the source of tumor initiation and maintenance. These cells are highly resistant to current cancer treatment and may be responsible for the disease recurrence after therapy as well. Therefore, considerable efforts have been made to elucidate the molecular and pathological properties of the CSCs to develop effective therapies targeting CSCs. A growing body of experimental evidence has revealed that therapeutic targeting CSCs may offer a new strategy for patients with breast cancer (BC). In this review, we summarized the evidence for existence of CSCs, followed by an overview of their molecular biomarkers, signaling pathways and potential therapeutic strategies to target these CSCs in BC.
Collapse
Affiliation(s)
- Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Zhaoming Li
- Department of Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Xudong Zhang
- Department of Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Yu Chang
- Department of Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
164
|
Melchor L, Molyneux G, Mackay A, Magnay FA, Atienza M, Kendrick H, Nava-Rodrigues D, López-García MÁ, Milanezi F, Greenow K, Robertson D, Palacios J, Reis-Filho JS, Smalley MJ. Identification of cellular and genetic drivers of breast cancer heterogeneity in genetically engineered mouse tumour models. J Pathol 2014; 233:124-37. [PMID: 24615332 DOI: 10.1002/path.4345] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 01/24/2014] [Accepted: 02/26/2014] [Indexed: 11/08/2022]
Abstract
The heterogeneous nature of mammary tumours may arise from different initiating genetic lesions occurring in distinct cells of origin. Here, we generated mice in which Brca2, Pten and p53 were depleted in either basal mammary epithelial cells or luminal oestrogen receptor (ER)-negative cells. Basal cell-origin tumours displayed similar histological phenotypes, regardless of the depleted gene. In contrast, luminal ER-negative cells gave rise to diverse phenotypes, depending on the initiating lesions, including both ER-negative and, strikingly, ER-positive invasive ductal carcinomas. Molecular profiling demonstrated that luminal ER-negative cell-origin tumours resembled a range of the molecular subtypes of human breast cancer, including basal-like, luminal B and 'normal-like'. Furthermore, a subset of these tumours resembled the 'claudin-low' tumour subtype. These findings demonstrate that not only do mammary tumour phenotypes depend on the interactions between cell of origin and driver genetic aberrations, but also multiple mammary tumour subtypes, including both ER-positive and -negative disease, can originate from a single epithelial cell type. This is a fundamental advance in our understanding of tumour aetiology.
Collapse
MESH Headings
- Animals
- BRCA2 Protein/deficiency
- BRCA2 Protein/genetics
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Cell Proliferation
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Claudins/metabolism
- Disease Models, Animal
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Female
- Gene Expression Regulation, Neoplastic
- Genetic Predisposition to Disease
- Humans
- Mammary Glands, Animal/metabolism
- Mammary Glands, Animal/pathology
- Mice
- Mice, Knockout
- PTEN Phosphohydrolase/deficiency
- PTEN Phosphohydrolase/genetics
- Phenotype
- Receptors, Estrogen/metabolism
- Time Factors
- Tumor Suppressor Protein p53/deficiency
- Tumor Suppressor Protein p53/genetics
Collapse
Affiliation(s)
- Lorenzo Melchor
- Division of Breast Cancer Research, Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
165
|
Jajosky AN, Coad JE, Vos JA, Martin KH, Senft JR, Wenger SL, Gibson LF. RepSox slows decay of CD34+ acute myeloid leukemia cells and decreases T cell immunoglobulin mucin-3 expression. Stem Cells Transl Med 2014; 3:836-48. [PMID: 24855276 DOI: 10.5966/sctm.2013-0193] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Despite initial response to therapy, most acute myeloid leukemia (AML) patients relapse. To eliminate relapse-causing leukemic stem/progenitor cells (LPCs), patient-specific immune therapies may be required. In vitro cellular engineering may require increasing the "stemness" or immunogenicity of tumor cells and activating or restoring cancer-impaired immune-effector and antigen-presenting cells. Leukapheresis samples provide the cells needed to engineer therapies: LPCs to be targeted, normal hematopoietic stem cells to be spared, and cancer-impaired immune cells to be repaired and activated. This study sought to advance development of LPC-targeted therapies by exploring nongenetic ways to slow the decay and to increase the immunogenicity of primary CD34(+) AML cells. CD34(+) AML cells generally displayed more colony-forming and aldehyde dehydrogenase activity than CD34(-) AML cells. Along with exposure to bone marrow stromal cells and low (1%-5%) oxygen, culture with RepSox (a reprogramming tool and inhibitor of transforming growth factor-β receptor 1) consistently slowed decline of CD34(+) AML and myelodysplastic syndrome (MDS) cells. RepSox-treated AML cells displayed higher CD34, CXCL12, and MYC mRNA levels than dimethyl sulfoxide-treated controls. RepSox also accelerated loss of T cell immunoglobulin mucin-3 (Tim-3), an immune checkpoint receptor that impairs antitumor immunity, from the surface of AML and MDS cells. Our results suggest RepSox may reduce Tim-3 expression by inhibiting transforming growth factor-β signaling and slow decay of CD34(+) AML cells by increasing CXCL12 and MYC, two factors that inhibit AML cell differentiation. By prolonging survival of CD34(+) AML cells and reducing Tim-3, RepSox may promote in vitro immune cell activation and advance development of LPC-targeted therapies.
Collapse
MESH Headings
- Aldehyde Dehydrogenase/metabolism
- Antigens, CD34/genetics
- Antigens, CD34/metabolism
- Biomarkers, Tumor/metabolism
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Cellular Reprogramming/drug effects
- Chemokine CXCL12/genetics
- Chemokine CXCL12/metabolism
- Coculture Techniques
- Dose-Response Relationship, Drug
- Feeder Cells
- Gene Expression Regulation, Leukemic/drug effects
- Hepatitis A Virus Cellular Receptor 2
- Humans
- Leukapheresis
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/immunology
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Oxygen/metabolism
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/metabolism
- Proto-Oncogene Proteins c-myc/genetics
- Proto-Oncogene Proteins c-myc/metabolism
- Pyrazoles/pharmacology
- Pyridines/pharmacology
- Receptor, Transforming Growth Factor-beta Type I
- Receptors, Transforming Growth Factor beta/antagonists & inhibitors
- Receptors, Transforming Growth Factor beta/metabolism
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/pathology
- Time Factors
- Tumor Cells, Cultured
- Tumor Escape
Collapse
Affiliation(s)
- Audrey N Jajosky
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Cancer Cell Biology Program, and Departments of Pathology, Neurobiology and Anatomy, and Microbiology, Immunology and Cell Biology, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | - James E Coad
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Cancer Cell Biology Program, and Departments of Pathology, Neurobiology and Anatomy, and Microbiology, Immunology and Cell Biology, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | - Jeffrey A Vos
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Cancer Cell Biology Program, and Departments of Pathology, Neurobiology and Anatomy, and Microbiology, Immunology and Cell Biology, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | - Karen H Martin
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Cancer Cell Biology Program, and Departments of Pathology, Neurobiology and Anatomy, and Microbiology, Immunology and Cell Biology, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | - Jamie R Senft
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Cancer Cell Biology Program, and Departments of Pathology, Neurobiology and Anatomy, and Microbiology, Immunology and Cell Biology, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | - Sharon L Wenger
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Cancer Cell Biology Program, and Departments of Pathology, Neurobiology and Anatomy, and Microbiology, Immunology and Cell Biology, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | - Laura F Gibson
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Cancer Cell Biology Program, and Departments of Pathology, Neurobiology and Anatomy, and Microbiology, Immunology and Cell Biology, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| |
Collapse
|
166
|
Jaganathan H, Mitra S, Srinivasan S, Dave B, Godin B. Design and in vitro evaluation of layer by layer siRNA nanovectors targeting breast tumor initiating cells. PLoS One 2014; 9:e91986. [PMID: 24694753 PMCID: PMC3973666 DOI: 10.1371/journal.pone.0091986] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 02/15/2014] [Indexed: 12/31/2022] Open
Abstract
Efficient therapeutics and early detection has helped to increase breast cancer survival rates over the years. However, the recurrence of breast cancer remains to be a problem and this may be due to the presence of a small population of cells, called tumor initiating cells (TICs). Breast TICs are resistant to drugs, difficult to detect, and exhibit high self-renewal capabilities. In this study, layer by layer (LBL) small interfering RNA (siRNA) nanovectors (SNVs) were designed to target breast TICs. SNVs were fabricated using alternating layers of poly-L-lysine and siRNA molecules on gold (Au) nanoparticle (NP) surfaces. The stability, cell uptake, and release profile for SNVs were examined. In addition, SNVs reduced TIC-related STAT3 expression levels, CD44+/CD24−/EpCAM+ surface marker levels and the number of mammospheres formed compared to the standard transfection agent. The data from this study show, for the first time, that SNVs in LBL assembly effectively delivers STAT3 siRNA and inhibit the growth of breast TICs in vitro.
Collapse
Affiliation(s)
- Hamsa Jaganathan
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Sucharita Mitra
- Cancer Center of Excellence, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Srimeenakshi Srinivasan
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Bhuvanesh Dave
- Cancer Center of Excellence, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
167
|
Manda K, Kavanagh JN, Buttler D, Prise KM, Hildebrandt G. Low dose effects of ionizing radiation on normal tissue stem cells. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2014; 761:6-14. [PMID: 24566131 DOI: 10.1016/j.mrrev.2014.02.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 02/03/2014] [Accepted: 02/13/2014] [Indexed: 12/18/2022]
Abstract
In recent years, there has been growing evidence for the involvement of stem cells in cancer initiation. As a result of their long life span, stem cells may have an increased propensity to accumulate genetic damage relative to differentiated cells. Therefore, stem cells of normal tissues may be important targets for radiation-induced carcinogenesis. Knowledge of the effects of ionizing radiation (IR) on normal stem cells and on the processes involved in carcinogenesis is very limited. The influence of high doses of IR (>5Gy) on proliferation, cell cycle and induction of senescence has been demonstrated in stem cells. There have been limited studies of the effects of moderate (0.5-5Gy) and low doses (<0.5Gy) of IR on stem cells however, the effect of low dose IR (LD-IR) on normal stem cells as possible targets for radiation-induced carcinogenesis has not been studied in any depth. There may also be important parallels between stem cell responses and those of cancer stem cells, which may highlight potential key common mechanisms of their response and radiosensitivity. This review will provide an overview of the current knowledge of radiation-induced effects on normal stem cells, with particular focus on low and moderate doses of IR.
Collapse
Affiliation(s)
- Katrin Manda
- Department of Radiotherapy and Radiation Oncology, University of Rostock, Suedring 75, 18059 Rostock, Germany.
| | - Joy N Kavanagh
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom.
| | - Dajana Buttler
- Department of Radiotherapy and Radiation Oncology, University of Rostock, Suedring 75, 18059 Rostock, Germany.
| | - Kevin M Prise
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom.
| | - Guido Hildebrandt
- Department of Radiotherapy and Radiation Oncology, University of Rostock, Suedring 75, 18059 Rostock, Germany.
| |
Collapse
|
168
|
Zhao Y, Huang Q, Zhang T, Dong J, Wang A, Lan Q, Gu X, Qin Z. Ultrastructural Studies of Glioma Stem Cells/Progenitor Cells. Ultrastruct Pathol 2014; 32:241-5. [DOI: 10.1080/01913120802289165] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
169
|
Chen Z, Forman LW, Williams RM, Faller DV. Protein kinase C-δ inactivation inhibits the proliferation and survival of cancer stem cells in culture and in vivo. BMC Cancer 2014; 14:90. [PMID: 24528676 PMCID: PMC3927586 DOI: 10.1186/1471-2407-14-90] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 02/06/2014] [Indexed: 12/18/2022] Open
Abstract
Background A subpopulation of tumor cells with distinct stem-like properties (cancer stem-like cells, CSCs) may be responsible for tumor initiation, invasive growth, and possibly dissemination to distant organ sites. CSCs exhibit a spectrum of biological, biochemical, and molecular features that are consistent with a stem-like phenotype, including growth as non-adherent spheres (clonogenic potential), ability to form a new tumor in xenograft assays, unlimited self-renewal, and the capacity for multipotency and lineage-specific differentiation. PKCδ is a novel class serine/threonine kinase of the PKC family, and functions in a number of cellular activities including cell proliferation, survival or apoptosis. PKCδ has previously been validated as a synthetic lethal target in cancer cells of multiple types with aberrant activation of Ras signaling, using both genetic (shRNA and dominant-negative PKCδ mutants) and small molecule inhibitors. In contrast, PKCδ is not required for the proliferation or survival of normal cells, suggesting the potential tumor-specificity of a PKCδ-targeted approach. Methods shRNA knockdown was used validate PKCδ as a target in primary cancer stem cell lines and stem-like cells derived from human tumor cell lines, including breast, pancreatic, prostate and melanoma tumor cells. Novel and potent small molecule PKCδ inhibitors were employed in assays monitoring apoptosis, proliferation and clonogenic capacity of these cancer stem-like populations. Significant differences among data sets were determined using two-tailed Student’s t tests or ANOVA. Results We demonstrate that CSC-like populations derived from multiple types of human primary tumors, from human cancer cell lines, and from transformed human cells, require PKCδ activity and are susceptible to agents which deplete PKCδ protein or activity. Inhibition of PKCδ by specific genetic strategies (shRNA) or by novel small molecule inhibitors is growth inhibitory and cytotoxic to multiple types of human CSCs in culture. PKCδ inhibition efficiently prevents tumor sphere outgrowth from tumor cell cultures, with exposure times as short as six hours. Small-molecule PKCδ inhibitors also inhibit human CSC growth in vivo in a mouse xenograft model. Conclusions These findings suggest that the novel PKC isozyme PKCδ may represent a new molecular target for cancer stem cell populations.
Collapse
Affiliation(s)
| | | | | | - Douglas V Faller
- Cancer Center, Boston University School of Medicine, K-712C, 72 E, Concord St,, Boston, MA 02118, USA.
| |
Collapse
|
170
|
Fong D, Seeber A, Terracciano L, Kasal A, Mazzoleni G, Lehne F, Gastl G, Spizzo G. Expression of EpCAM(MF) and EpCAM(MT) variants in human carcinomas. J Clin Pathol 2014; 67:408-14. [PMID: 24465008 PMCID: PMC3995261 DOI: 10.1136/jclinpath-2013-201932] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Aims Regulated intramembrane proteolysis has been shown to be an important mechanism for oncogenic activation of epithelial cell adhesion molecule (EpCAM) through nuclear translocation of the intracellular domain EpICD. Recent studies have identified new membrane-bound EpCAM variants. To evaluate the prevalence of two membranous EpCAM variants in human tumours, we performed a large-scale expression analysis using specific antibodies against the extracellular domain EpEX (MOC-31 clone) and the intracellular domain EpICD (9-2 clone) of the EpCAM antigen by immunohistochemistry. Material and methods Two multi-tissue microarrays (TMA) series containing 1564 tissue samples each of 53 different histological tumour types were stained and compared. One TMA was stained for EpEX and one for EpICD. Membranous full-length EpCAM (EpCAMMF) expression in tissues was defined by the expression of EpEX and EpICD, while the truncated variant of EpCAM (EpCAMMT) was characterised by a significant loss of membranous EpICD expression compared with EpEX expression. Results We defined tumours with high EpCAMMT expression (ie, cancers of the endometrium and bladder), tumours with intermediate (ie, gastric, pancreatic, colorectal and oesophageal cancer) and tumours with low rates of expression of the EpCAMMT variant (ie, lung, ovarian, gallbladder, breast and prostate cancer). Conclusions Our results indicate that loss of membranous EpICD expression is a common event in human epithelial carcinomas, arguing for the expression of different degrees of EpCAMMF and EpCAMMT variants across the most important tumour entities. Future studies evaluating the prognostic and predictive role of these variants in human malignancies, especially in patients treated with EpCAM-specific antibodies, are clearly warranted.
Collapse
Affiliation(s)
- Dominic Fong
- Tyrolean Cancer Research Institute, , Innsbruck, Austria
| | | | | | | | | | | | | | | |
Collapse
|
171
|
Yu M, Finner A, Shapiro J, Lo B, Barekatain A, McElwee KJ. Hair follicles and their role in skin health. ACTA ACUST UNITED AC 2014. [DOI: 10.1586/17469872.1.6.855] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
172
|
Barton M, Santucci-Pereira J, Russo J. Molecular pathways involved in pregnancy-induced prevention against breast cancer. Front Endocrinol (Lausanne) 2014; 5:213. [PMID: 25540638 PMCID: PMC4261797 DOI: 10.3389/fendo.2014.00213] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 11/25/2014] [Indexed: 12/18/2022] Open
Abstract
Pregnancy produces a protective effect against breast cancer in women who had their first full term pregnancy (FTP) in their middle twenties. The later in life the first delivery occurs, the higher the risk of breast cancer development. Also, transiently during the postpartum period, the risk of developing breast cancer increases. This transient increased risk is taken over by a long-lasting protective period. The genomic profile of parous women has shown pregnancy induces a long-lasting "genomic signature" that explains the preventive effect on breast cancer. This signature reveals that chromatin remodeling is the driver of the differentiation process conferred by FTP. The chromatin remodeling process may be the ultimate step mediating the protection of the breast against developing breast cancer in post-menopausal years.
Collapse
Affiliation(s)
- Maria Barton
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA, USA
| | - Julia Santucci-Pereira
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA, USA
| | - Jose Russo
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA, USA
- *Correspondence: Jose Russo, The Irma H Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center, Temple University Health System, 333 Cottman Avenue, Room P2037, Philadelphia, PA 19111, USA e-mail:
| |
Collapse
|
173
|
Dobbin ZC, Landen CN. Isolation and characterization of potential cancer stem cells from solid human tumors--potential applications. CURRENT PROTOCOLS IN PHARMACOLOGY 2013; 63:14.28.1-14.28.19. [PMID: 24510756 PMCID: PMC4041697 DOI: 10.1002/0471141755.ph1428s63] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cancer stem cells (CSCs) are a subpopulation of cells within a heterogeneous tumor that have enhanced biologic properties, e.g., increased capacity for self-renewal, increased tumorigenicity, enhanced differentiation capacity, and resistance to chemo- and radiotherapies. This unit describes protocols to isolate and characterize potential cancer stem cells from a solid tumor. These involve creating a single-cell suspension from tumor tissue, tagging the cell subpopulations of interest, and sorting them into different populations. The sorted subpopulations can be evaluated for their ability to meet the functional requirements of a CSC, which primarily include increased tumorigenicity in an in vivo xenograft assay. Use of the protocols described in this unit makes it possible to study populations of cells that may have properties of CSCs.
Collapse
Affiliation(s)
- Zachary C. Dobbin
- Division of Gynecologic Oncology, University of Alabama at Birmingham, Wallace Tumor Institute, 1824 6 Avenue South, Birmingham, AL 35233
| | - Charles N. Landen
- Division of Gynecologic Oncology, University of Alabama at Birmingham, Wallace Tumor Institute, 1824 6 Avenue South, Birmingham, AL 35233,Zachary Dobbin: , 205-934-2049, Charles Landen: , 205-934-0473
| |
Collapse
|
174
|
Charpentier M, Martin S. Interplay of Stem Cell Characteristics, EMT, and Microtentacles in Circulating Breast Tumor Cells. Cancers (Basel) 2013; 5:1545-65. [PMID: 24240660 PMCID: PMC3875953 DOI: 10.3390/cancers5041545] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 10/08/2013] [Accepted: 11/04/2013] [Indexed: 12/31/2022] Open
Abstract
Metastasis, not the primary tumor, is responsible for the majority of breast cancer-related deaths. Emerging evidence indicates that breast cancer stem cells (CSCs) and the epithelial-to-mesenchymal transition (EMT) cooperate to produce circulating tumor cells (CTCs) that are highly competent for metastasis. CTCs with both CSC and EMT characteristics have recently been identified in the bloodstream of patients with metastatic disease. Breast CSCs have elevated tumorigenicity required for metastatic outgrowth, while EMT may promote CSC character and endows breast cancer cells with enhanced invasive and migratory potential. Both CSCs and EMT are associated with a more flexible cytoskeleton and with anoikis-resistance, which help breast carcinoma cells survive in circulation. Suspended breast carcinoma cells produce tubulin-based extensions of the plasma membrane, termed microtentacles (McTNs), which aid in reattachment. CSC and EMT-associated upregulation of intermediate filament vimentin and increased detyrosination of α-tubulin promote the formation of McTNs. The combined advantages of CSCs and EMT and their associated cytoskeletal alterations increase metastatic efficiency, but understanding the biology of these CTCs also presents new therapeutic targets to reduce metastasis.
Collapse
Affiliation(s)
- Monica Charpentier
- Program in Molecular Medicine, University of Maryland School of Medicine, 655 W. Baltimore St., Bressler Bldg., Rm 10-20, Baltimore, MD 21201, USA; E-Mail:
- Marlene and Stewart Greenebaum National Cancer Institute Cancer Center, University of Maryland School of Medicine, 655 W. Baltimore St., Bressler Bldg., Rm 10-29, Baltimore, MD 21201, USA
| | - Stuart Martin
- Marlene and Stewart Greenebaum National Cancer Institute Cancer Center, University of Maryland School of Medicine, 655 W. Baltimore St., Bressler Bldg., Rm 10-29, Baltimore, MD 21201, USA
- Department of Physiology, University of Maryland School of Medicine, 655 W. Baltimore St., Bressler Bldg., Rm 10-29, Baltimore, MD 21201, USA
| |
Collapse
|
175
|
Bell D, Hanna EY, Miele L, Roberts D, Weber RS, El-Naggar AK. Expression and significance of notch signaling pathway in salivary adenoid cystic carcinoma. Ann Diagn Pathol 2013; 18:10-3. [PMID: 24238845 DOI: 10.1016/j.anndiagpath.2013.10.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 09/08/2013] [Accepted: 10/03/2013] [Indexed: 12/19/2022]
Abstract
Notch signaling plays a role in stem cell biology, tumor formation, angiogenesis, and cell death. Targeting Notch pathway could serve as a therapeutic strategy in cancer. Little is known about the differential role of various components of the Notch pathway in salivary adenoid cystic carcinoma (AdCC). To investigate the association of the Notch pathway in AdCC carcinogenesis, we analyzed the Notch receptor (Notch-1, Notch-2, Notch-4) and Notch ligand (Jagged-1, Delta) expressions. The results showed elevated expression levels of all 5 proteins in AdCC tissue relative to normal salivary gland tissues. Jagged-1/Notch-2 coexpression was significantly associated with increased patient survival rate. The elevated expression level of these Notch receptors and ligands in AdCC points to Notch signaling as a key player in AdCC pathogenesis. Our data provide evidence for a relationship between Jagged-1/Notch-2 coexpression and better overall patient survival with AdCC. Targeting Notch signaling pathway may provide therapeutic benefits for these patients.
Collapse
Affiliation(s)
- Diana Bell
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX.
| | | | | | | | | | | |
Collapse
|
176
|
Mu Z, Li H, Fernandez SV, Alpaugh KR, Zhang R, Cristofanilli M. EZH2 knockdown suppresses the growth and invasion of human inflammatory breast cancer cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2013; 32:70. [PMID: 24294976 PMCID: PMC3850122 DOI: 10.1186/1756-9966-32-70] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 09/24/2013] [Indexed: 11/17/2022]
Abstract
Introduction Inflammatory breast cancer (IBC) is the most metastatic variant of breast cancer with the poorest survival in all types of breast cancer patients and presently therapeutic targets for IBC are very limited. Enhancer of zeste homolog 2 (EZH2) is frequently expressed in human IBC and its expression positively correlates with worse clinical outcome. However, the molecular basis for EZH2 promoting IBC has not been explored. Here, we investigated the functional role of EZH2 in IBC cells by examining the effects of its knockdown on the formation of tumor spheroids and invasion of these cells in vitro and in vivo in an orthotopic xenograft model. Methods SUM149 and a new IBC cell line-FC-IBC-02 derived from pleural effusion fluid of an IBC patient were used in this study. Specific knockdown of EZH2 was performed using short hairpin RNA (shRNA) specific to the human EZH2 gene. Cell growth and the formation of tumor spheroids were examined in vitro. The effects of EZH2 knockdown on IBC cell migration and invasion were examined by a Boyden chamber assay. For the in vivo tumor growth studies, IBC cells were orthotopically transplanted into the mammary fat pads of immunodeficient mice. Results The results showed that EZH2 is expressed at higher levels in human IBC cell lines compared with normal human mammary epithelial cells, and the knockdown of EZH2 expression significantly suppressed cell growth and tumor spheroid formation of human IBC cells in vitro. In addition, EZH2 knockdown inhibited the migration and invasion of IBC cells. Significantly, EZH2 knockdown suppressed the angiogenesis and tumor growth of IBC cells in vivo. Conclusions Our results provide direct evidence that EZH2 is critical for the formation of tumor spheroids and invasion of human IBC cells and could be a potential target for developing novel therapeutic strategies for human IBC.
Collapse
|
177
|
Ribelles N, Santonja A, Pajares B, Llácer C, Alba E. The seed and soil hypothesis revisited: current state of knowledge of inherited genes on prognosis in breast cancer. Cancer Treat Rev 2013; 40:293-9. [PMID: 24112814 DOI: 10.1016/j.ctrv.2013.09.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 09/03/2013] [Accepted: 09/06/2013] [Indexed: 10/26/2022]
Abstract
The crucial event in the course of malignancies such as breast cancer is its metastatic spread from the primary tumor of origin to distant organs. The natural history of a tumor is determined by the expression of its genes, and in this sense, knowledge has advanced dramatically in recent decades. However, much less is known about the role that the patient plays in the behavior of a tumor. In this article, we review the evidence regarding the genetic background of the host in metastatic tumor dissemination, providing information from epidemiological studies as well as from animal models and human studies. Undoubtedly, the elucidation of possible interpersonal variability in susceptibility to developing metastases would significantly contribute to improve management of cancer patients.
Collapse
Affiliation(s)
- Nuria Ribelles
- Department of Medical Oncology, Hospital Universitario Virgen de la Victoria, Campus Teatinos s/n, 29010 Málaga, Spain.
| | | | | | | | | |
Collapse
|
178
|
Cui TX, Kryczek I, Zhao L, Zhao E, Kuick R, Roh MH, Vatan L, Szeliga W, Mao Y, Thomas DG, Kotarski J, Tarkowski R, Wicha M, Cho K, Giordano T, Liu R, Zou W. Myeloid-derived suppressor cells enhance stemness of cancer cells by inducing microRNA101 and suppressing the corepressor CtBP2. Immunity 2013; 39:611-21. [PMID: 24012420 DOI: 10.1016/j.immuni.2013.08.025] [Citation(s) in RCA: 361] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 06/24/2013] [Indexed: 01/17/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) and cancer stem cells (CSCs) are important cellular components in the cancer microenvironment and may affect cancer phenotype and patient outcome. The nature of MDSCs and their interaction with CSCs in ovarian carcinoma are unclear. We examined the interaction between MDSCs and CSCs in patients with ovarian carcinoma and showed that MDSCs inhibited T cell activation and enhanced CSC gene expression, sphere formation, and cancer metastasis. MDSCs triggered miRNA101 expression in cancer cells. miRNA101 subsequently repressesed the corepressor gene C-terminal binding protein-2 (CtBP2), and CtBP2 directly targeted stem cell core genes resulting in increased cancer cell stemness and increasing metastatic and tumorigenic potential. Increased MDSC density and tumor microRNA101 expression predict poor survival, as does decreased tumor CtBP2 expression, independent of each other. Collectively, our work identifies an immune-associated cellular, molecular, and clinical network involving MDSCs-microRNA101-CtBP2-stem cell core genes, which extrinsically controls cancer stemness and impacts patient outcome.
Collapse
Affiliation(s)
- Tracy X Cui
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
179
|
Effects of ellipticine on ALDH1A1-expressing breast cancer stem cells--an in vitro and in silico study. Tumour Biol 2013; 35:723-37. [PMID: 23982874 DOI: 10.1007/s13277-013-1099-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 08/08/2013] [Indexed: 02/07/2023] Open
Abstract
Targeting breast cancer stem cells (BCSCs) offers a promising strategy for breast cancer treatment. We examined the plant alkaloid ellipticine for its efficacy to inhibit the expression of aldehyde dehydrogenase 1 class A1 (ALDH1A1)-positive BCSCs by in vitro and in silico methods. At 3 mM concentration, ellipticine decreased the expression of ALDH1A1-positive BCSCs by 62% (p = 0.073) in the MCF7 cell line and by 53% (p = 0.024) in the SUM159 cell line compared to vehicle-treated cultures. Ellipticine significantly reduced the formation of mammospheres, whereas paclitaxel enhanced mammosphere formation in both the treated cell lines. Interestingly, when treated with a combination of ellipticine and paclitaxel, the percentage of ALDH1A1-positive BCSCs dropped by several fold in vitro. A homology model of Homo sapiens ALDH1A1 was built using the crystal structure of NAD-bound sheep liver class I aldehyde dehydrogenase [PDB ID: 1BXS] as a template. Molecular simulation and docking studies revealed that the amino acids Asn-117 and Asn-121, Glu-249, Cys-302, and Gln-350, present in the active site of human ALDH1A1, played a vital role in interacting with the drug. The present study suggests that ellipticine reduces the proliferation and self-renewal ability of ALDH1A1-positive BCSCs and can be used in combination with a cytotoxic drug like paclitaxel for potential targeting of BCSCs.
Collapse
|
180
|
N-cadherin/FGFR promotes metastasis through epithelial-to-mesenchymal transition and stem/progenitor cell-like properties. Oncogene 2013; 33:3411-21. [PMID: 23975425 DOI: 10.1038/onc.2013.310] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 05/05/2013] [Accepted: 05/20/2013] [Indexed: 12/15/2022]
Abstract
N-cadherin and HER2/neu were found to be co-expressed in invasive breast carcinomas. To test the contribution of N-cadherin and HER2 in mammary tumor metastasis, we targeted N-cadherin expression in the mammary epithelium of the MMTV-Neu mouse. In the context of ErbB2/Neu, N-cadherin stimulated carcinoma cell invasion, proliferation and metastasis. N-cadherin caused fibroblast growth factor receptor (FGFR) upmodulation, resulting in epithelial-to-mesenchymal transition (EMT) and stem/progenitor like properties, involving Snail and Slug upregulation, mammosphere formation and aldehyde dehydrogenase activity. N-cadherin potentiation of the FGFR stimulated extracellular signal regulated kinase (ERK) and protein kinase B (AKT) phosphorylation resulting in differential effects on metastasis. Although ERK inhibition suppressed cyclin D1 expression, cell proliferation and stem/progenitor cell properties, it did not affect invasion or EMT. Conversely, AKT inhibition suppressed invasion through Akt 2 attenuation, and EMT through Snail inhibition, but had no effect on cyclin D1 expression, cell proliferation or mammosphere formation. These findings suggest N-cadherin/FGFR has a pivotal role in promoting metastasis through differential regulation of ERK and AKT, and underscore the potential for targeting the FGFR in advanced ErbB2-amplified breast tumors.
Collapse
|
181
|
Duong MT, Akli S, Macalou S, Biernacka A, Debeb BG, Yi M, Hunt KK, Keyomarsi K. Hbo1 is a cyclin E/CDK2 substrate that enriches breast cancer stem-like cells. Cancer Res 2013; 73:5556-68. [PMID: 23955388 DOI: 10.1158/0008-5472.can-13-0013] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Expression of cyclin E proteolytic cleavage products, low-molecular weight cyclin E (LMW-E), is associated with poor clinical outcome in patients with breast cancer and it enhances tumorigenecity in mouse models. Here we report that LMW-E expression in human mammary epithelial cells induces an epithelial-to-mesenchymal transition phenotype, increases the CD44(hi)/CD24(lo) population, enhances mammosphere formation, and upregulates aldehyde dehydrogenase expression and activity. We also report that breast tumors expressing LMW-E have a higher proportion of CD44(hi)/CD24(lo) tumor cells as compared with tumors expressing only full-length cyclin E. In order to explore how LMW-E enriches cancer stem cells in breast tumors, we conducted a protein microarray analysis that identified the histone acetyltransferase (HAT) Hbo1 as a novel cyclin E/CDK2 substrate. The LMW-E/CDK2 complex phosphorylated Hbo1 at T88 without affecting its HAT activity. When coexpressed with LMW-E/CDK2, wild-type Hbo1 promoted enrichment of cancer stem-like cells (CSC), whereas the T88 Hbo1 mutant reversed the CSC phenotype. Finally, doxorubicin and salinomycin (a CSC-selective cytotoxic agent) synergized to kill cells expressing LMW-E, but not full-length cyclin E. Collectively, our results suggest that the heightened oncogenecity of LMW-E relates to its ability to promote CSC properties, supporting the design of therapeutic strategies to target this unique function.
Collapse
Affiliation(s)
- Mylinh T Duong
- Departments of Experimental Radiation Oncology, Radiation Oncology, and Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 0066, Houston, TX77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
182
|
Wang X, Lu H, Li T, Yu L, Liu G, Peng X, Zhao J. Krüppel-like factor 8 promotes tumorigenic mammary stem cell induction by targeting miR-146a. Am J Cancer Res 2013; 3:356-373. [PMID: 23977446 PMCID: PMC3744016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Accepted: 07/03/2013] [Indexed: 06/02/2023] Open
Abstract
The properties of stem cells can be induced during the epithelial to mesenchymal transition (EMT). The responsible molecular mechanisms, however, remain largely undefined. Here we report the identification of the microRNA-146a (miR-146a) as a common target of Krüppel-like factor 8 (KLF8) and TGF-β, both of which are known EMT-inducers. Upon KLF8 overexpression or TGF-β treatment, a significant portion of the MCF-10A cells gained stem cell traits as demonstrated by an increased expression of CD44(high)/CD24low, activity of aldehyde dehydrogenase (ALDH), mammosphere formation and chemoresistance. Along with this change, the expression of miR-146a was highly upregulated in the cells. Importantly, we found that miR-146a was aberrantly co-overexpressed with KLF8 in a panel of invasive human breast cancer cell lines. Ectopic expression of KLF8 failed to induce the stem cell traits in the MCF-10A cells if the cells were pre-treated with miR-146a inhibitor, whereas overexpression of miR-146a in the MCF-10A cells alone was sufficient to induce the stem cell traits. Co-staining and luciferase reporter analyses indicated that miR-146a targets the 3'-UTR of the Notch signaling inhibitor NUMB for translational inhibition. Overexpression of KLF8 dramatically potentiated the tumorigenecity of MCF-10A cells expressing the H-Ras oncogene, which was accompanied by a loss of NUMB expression in the tumors. Taken together, this study identifies a novel role and mechanism for KLF8 in inducing pro-tumorigenic mammary stem cells via miR-146a potentially by activating Notch signaling. This mechanism could be exploited as a therapeutic target against drug resistance of breast cancer.
Collapse
Affiliation(s)
- Xianhui Wang
- Burnett School of Biomedical Sciences, University of Central Florida, College of MedicineOrlando, FL
| | - Heng Lu
- Burnett School of Biomedical Sciences, University of Central Florida, College of MedicineOrlando, FL
| | - Tianshu Li
- Burnett School of Biomedical Sciences, University of Central Florida, College of MedicineOrlando, FL
| | - Lin Yu
- Burnett School of Biomedical Sciences, University of Central Florida, College of MedicineOrlando, FL
| | - Gang Liu
- Center for Cell Biology & Cancer Research, Albany Medical CollegeAlbany, NY
| | - Xu Peng
- Department of Systems Biology and Translational Medicine, College of Medicine, Texas A&M Health Science CenterTemple, TX
| | - Jihe Zhao
- Burnett School of Biomedical Sciences, University of Central Florida, College of MedicineOrlando, FL
| |
Collapse
|
183
|
Boecker W, Stenman G, Loening T, Andersson MK, Bankfalvi A, von Holstein S, Heegaard S, Lange A, Berg T, Samoilova V, Tiemann K, Buchwalow I. K5/K14-positive cells contribute to salivary gland-like breast tumors with myoepithelial differentiation. Mod Pathol 2013; 26:1086-100. [PMID: 23558567 DOI: 10.1038/modpathol.2013.45] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 01/14/2013] [Accepted: 01/14/2013] [Indexed: 02/07/2023]
Abstract
Salivary gland-like tumors of the breast show a great variety of architectural patterns and cellular differentiations such as glandular, myoepithelial, squamous, and even mesenchymal phenotypes. However, currently little is known about the evolution and cellular differentiation of these tumors. For that reason, we performed an in situ triple immunofluorescence lineage/differentiation tracing (isTILT) and qRT-PCR study of basal (K5/K14), glandular (K7/K8/18), and epidermal-specific squamous (K10) keratins, p63, and smooth muscle actin (SMA; myoepithelial marker) with the aim to construct and trace different cell lineages and define their cellular hierarchy in tumors with myoepithelial differentiation. isTILT analysis of a series of 28 breast, salivary, and lacrimal gland tumors, including pleomorphic adenomas (n=8), epithelial-myoepithelial tumors (n=9), and adenoid cystic carcinomas (n=11) revealed that all tumor types contained K5/K14-positive progenitor cells in varying frequencies from a few percent up to 15%. These K5/K14-positive tumor cells were found to differentiate to glandular- (K8/18-positive) and myoepithelial-lineage (SMA-positive)-specific cells and were also shown to generate various heterologeous cell differentiations such as squamous and mesenchymal progenies. p63 was co-expressed with K5/K14 in basal-like progenitor cells, myoepithelial, and squamous cells but not in glandular cells. Our results show that the corresponding counterpart tumors of breast and salivary/lacrimal glands have identical cellular compositions. Taken together, our isTILT and RNA-expression data indicate that look-alike tumors of the breast represent a special subgroup of basal-type tumors with benign or usually low malignant potential.
Collapse
Affiliation(s)
- Werner Boecker
- Institute for Hematopathology, Reference Center for Gynaeco- and Breast-pathology, Hamburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Bane A, Viloria-Petit A, Pinnaduwage D, Mulligan AM, O'Malley FP, Andrulis IL. Clinical-pathologic significance of cancer stem cell marker expression in familial breast cancers. Breast Cancer Res Treat 2013; 140:195-205. [PMID: 23813303 PMCID: PMC3706716 DOI: 10.1007/s10549-013-2591-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 05/29/2013] [Indexed: 01/16/2023]
Abstract
Human breast cancer cells with a CD44+/CD24−/low or ALDH1+ phenotype have been demonstrated to be enriched for cancer stem cells (CSCs) using in vitro and in vivo techniques. The aim of this study was to determine the association between CD44+/CD24−/low and ALDH1 expression with clinical–pathologic tumor characteristics, tumor molecular subtype, and survival in a well characterized collection of familial breast cancer cases. 364 familial breast cancers from the Ontario Familial Breast Cancer Registry (58 BRCA1-associated, 64 BRCA2-associated, and 242 familial non-BRCA1/2 cancers) were studied. Each tumor had a centralized pathology review performed. TMA sections of all tumors were analyzed for the expression of ER, PR, HER2, CK5, CK14, EGFR, CD44, CD24, and ALDH1. The Chi square test or Fisher’s exact test was used to analyze the marker associations with clinical–pathologic tumor variables, molecular subtype and genetic subtype. Analyses of the association of overall survival (OS) with marker status were conducted using Kaplan–Meier plots and log-rank tests. The CD44+/CD24−/low and ALDH1+ phenotypes were identified in 16% and 15% of the familial breast cancer cases, respectively, and associated with high-tumor grade, a high-mitotic count, and component features of the medullary type of breast cancer. CD44+/CD24−/low and ALDH1 expression in this series were further associated with the basal-like molecular subtype and the CD44+/CD24−/low phenotype was independently associated with BRCA1 mutational status. The currently accepted breast CSCs markers are present in a minority of familial breast cancers. Whereas the presence of these markers is correlated with several poor prognostic features and the basal-like subtype of breast cancer, they do not predict OS.
Collapse
Affiliation(s)
- Anita Bane
- Department of Pathology and Molecular Medicine, Juravinski Hospital and Cancer Centre, McMaster University, 711 Concession St., Hamilton, ON, Canada.
| | | | | | | | | | | |
Collapse
|
185
|
Giordano A, Gao H, Cohen EN, Anfossi S, Khoury J, Hess K, Krishnamurthy S, Tin S, Cristofanilli M, Hortobagyi GN, Woodward WA, Lucci A, Reuben JM. Clinical relevance of cancer stem cells in bone marrow of early breast cancer patients. Ann Oncol 2013; 24:2515-2521. [PMID: 23798614 DOI: 10.1093/annonc/mdt223] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) are epithelial tumor cells that express CD44(+)CD24(-/lo). CSCs can be further divided into those that have aldehyde dehydrogenase (ALDH) activity (Aldefluor(+)) and those that do not. We hypothesized that if CSCs are responsible for tumor dissemination, their presence in bone marrow (BM) would be prognostic in early stages of breast cancer (EBC) patients. PATIENTS AND METHODS BM aspirates were collected at the time of surgery from 108 patients with EBC. BM was analyzed for CSCs and ALDH activity by flow cytometry. Overall survival and disease-free survival (DFS) were calculated from the date of diagnosis and analyzed with Kaplan-Meier survival plots. Cox multivariate proportional hazards model was also carried out. RESULTS Patients with CSCs in BM had a hazard ratio (HR) of 8.8 for DFS (P = 0.002); patients with Aldefluor(+) CSCs had a HR of 5.9 (P = 0.052) for DFS. All deceased patients (n = 7) had CSCs in BM. In multivariate analysis, the presence of CSCs in BM was a prognostic factor of DFS (HR = 15.8, P = 0.017). CONCLUSIONS The presence of BM metastasis is correlated with CSCs and these CSCs irrespective of ALDH activity are an independent adverse prognostic factor in EBC patients.
Collapse
Affiliation(s)
- A Giordano
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, USA; Department of Endocrinology and Molecular and Clinical Oncology, University of Naples Federico II, Naples, Italy
| | - H Gao
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - E N Cohen
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - S Anfossi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J Khoury
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - K Hess
- Departments of Biostatistics
| | - S Krishnamurthy
- Pathology, The University of Texas MD Anderson Cancer Center, Houston
| | - S Tin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - M Cristofanilli
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia
| | | | | | - A Lucci
- Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J M Reuben
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, USA.
| |
Collapse
|
186
|
Buchwalter G, Hickey MM, Cromer A, Selfors LM, Gunawardane RN, Frishman J, Jeselsohn R, Lim E, Chi D, Fu X, Schiff R, Brown M, Brugge JS. PDEF promotes luminal differentiation and acts as a survival factor for ER-positive breast cancer cells. Cancer Cell 2013; 23:753-67. [PMID: 23764000 PMCID: PMC3711136 DOI: 10.1016/j.ccr.2013.04.026] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 02/19/2013] [Accepted: 04/23/2013] [Indexed: 01/25/2023]
Abstract
Breast cancer is a heterogeneous disease and can be classified based on gene expression profiles that reflect distinct epithelial subtypes. We identify prostate-derived ETS factor (PDEF) as a mediator of mammary luminal epithelial lineage-specific gene expression and as a factor required for tumorigenesis in a subset of breast cancers. PDEF levels strongly correlate with estrogen receptor (ER)-positive luminal breast cancer, and PDEF transcription is inversely regulated by ER and GATA3. Furthermore, PDEF is essential for luminal breast cancer cell survival and is required in models of endocrine resistance. These results offer insights into the function of this ETS factor that are clinically relevant and may be of therapeutic value for patients with breast cancer treated with endocrine therapy.
Collapse
Affiliation(s)
- Gilles Buchwalter
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School
| | - Michele M. Hickey
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| | - Anne Cromer
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| | - Laura M. Selfors
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| | | | - Jason Frishman
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School
| | - Rinath Jeselsohn
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School
| | - Elgene Lim
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School
| | - David Chi
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School
| | - Xiaosong Fu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77054, USA
| | - Rachel Schiff
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77054, USA
| | - Myles Brown
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School
- Correspondence: (J.S.B.), (M.B.)
| | - Joan S. Brugge
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
- Correspondence: (J.S.B.), (M.B.)
| |
Collapse
|
187
|
Zheng H, Kang Y. Multilayer control of the EMT master regulators. Oncogene 2013; 33:1755-63. [PMID: 23604123 DOI: 10.1038/onc.2013.128] [Citation(s) in RCA: 252] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Revised: 02/27/2013] [Accepted: 02/27/2013] [Indexed: 12/23/2022]
Abstract
Metastasis is the leading cause of cancer-associated death in most tumor types. Metastatic dissemination of cancer cells from the primary tumor is believed to be initiated by the reactivation of an embryonic development program referred to as epithelial-mesenchymal transition (EMT), whereby epithelial cells lose apicobasal polarity and cell-cell contacts, and gain mesenchymal phenotypes with increased migratory and invasive capabilities. EMT has also been implicated in the regulation of cancer stem cell property, immune suppression and cancer regression. Several transcription factors have been identified as master regulators of EMT, including the Snail, Zeb and Twist families, and their expression is tightly regulated at different steps of transcription, translation and protein stability control by a variety of cell-intrinsic pathways as well as extracellular cues. Here, we review the recent literature on the signaling pathways and mechanisms that control the expression of these master transcription factors during EMT and cancer progression.
Collapse
Affiliation(s)
- H Zheng
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Y Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| |
Collapse
|
188
|
Borena BM, Bussche L, Burvenich C, Duchateau L, Van de Walle GR. Mammary stem cell research in veterinary science: an update. Stem Cells Dev 2013; 22:1743-51. [PMID: 23360296 DOI: 10.1089/scd.2012.0677] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The mammary gland is an organ with a remarkable regenerative capacity that can undergo multiple cycles of proliferation, lactation, and involution. Growing evidence suggests that these changes are driven by the coordinated division and differentiation of mammary stem cell populations (MaSC). Whereas information regarding MaSC and their role in comparative mammary gland physiology is readily available in human and mice, such information remains scarce in most veterinary mammal species such as cows, horses, sheep, goats, pigs, and dogs. We believe that a better knowledge on the MaSC in these species will not only help to gain more insights into mammary gland (patho) physiology in veterinary medicine, but will also be of value for human medicine. Therefore, this review summarizes the current knowledge on stem cell isolation and characterization in different mammals of veterinary importance.
Collapse
Affiliation(s)
- Bizunesh M Borena
- Department of Comparative Physiology and Biometrics, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | | | | | | | | |
Collapse
|
189
|
Du WW, Fang L, Yang X, Sheng W, Yang BL, Seth A, Zhang Y, Yang BB, Yee AJ. The Role of Versican in Modulating Breast Cancer Cell Self-renewal. Mol Cancer Res 2013; 11:443-55. [DOI: 10.1158/1541-7786.mcr-12-0461] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
190
|
Wilson-Robles HM, Daly M, Pfent C, Sheppard S. Identification and evaluation of putative tumour-initiating cells in canine malignant melanoma cell lines. Vet Comp Oncol 2013; 13:60-9. [DOI: 10.1111/vco.12019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 01/02/2013] [Accepted: 01/03/2013] [Indexed: 12/24/2022]
Affiliation(s)
- H. M. Wilson-Robles
- Department of Small Animal Clinical Sciences; College of Veterinary Medicine, Texas A&M University; College Station TX USA
| | - M. Daly
- Department of Small Animal Clinical Sciences; College of Veterinary Medicine, Texas A&M University; College Station TX USA
| | - C. Pfent
- Department of Veterinary Pathobiology; College of Veterinary Medicine, Texas A&M University; College Station TX USA
| | - S. Sheppard
- Department of Small Animal Clinical Sciences; College of Veterinary Medicine, Texas A&M University; College Station TX USA
| |
Collapse
|
191
|
Mukherjee D, Zhao J. The Role of chemokine receptor CXCR4 in breast cancer metastasis. Am J Cancer Res 2013; 3:46-57. [PMID: 23359227 PMCID: PMC3555200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 12/25/2012] [Indexed: 06/01/2023] Open
Abstract
Breast cancer is one of the leading causes of cancer related deaths worldwide. Breast cancer-related mortality is associated with the development of metastatic potential of primary tumor lesions. The chemokine receptor CXCR4 has been found to be a prognostic marker in various types of cancer, including breast cancer. Recent advances in the field of cancer biology has pointed to the critical role that CXCR4 receptor and its ligand CXCL12 play in the metastasis of various types of cancer, including breast cancer. Breast tumors preferentially metastasize to the lung, bones and lymph nodes, all of which represent organs that secrete high levels of CXCL12. CXCL12 acts as a chemoattractant that drives CXCR4-positive primary tumor cells towards secondary metastatic sites leading to the onset of metastatic lesions. Since its discovery in 2001, the CXCR4 field has progressed at a very fast rate and further studies have pointed to the role of CXCR4 in dissemination of tumor cells from primary sites, transendothelial migration of tumor cells as well as the trafficking and homing of cancer stem cells. This review summarizes the information that has been obtained over the years regarding the role of CXCL12-CXCR4 signaling in breast cancer, discusses its potential application to the development of new therapeutic tools for breast cancer control, and elucidates the potential therapeutic challenges which lie ahead and the future directions that this field can take for the improvement of prognosis in breast cancer patients.
Collapse
Affiliation(s)
- Debarati Mukherjee
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine Orlando, FL 32827, USA
| | | |
Collapse
|
192
|
Kim SY, Kang JW, Song X, Kim BK, Yoo YD, Kwon YT, Lee YJ. Role of the IL-6-JAK1-STAT3-Oct-4 pathway in the conversion of non-stem cancer cells into cancer stem-like cells. Cell Signal 2013; 25:961-9. [PMID: 23333246 DOI: 10.1016/j.cellsig.2013.01.007] [Citation(s) in RCA: 222] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 01/08/2013] [Indexed: 01/06/2023]
Abstract
Previous studies have demonstrated that a small subset of cancer cells is capable of tumor initiation. The existence of tumor initiating cancer stem cells (CSCs) has several implications in terms of future cancer treatment and therapies. However, recently, several researchers proposed that differentiated cancer cells (non-CSCs) can convert to stem-like cells to maintain equilibrium. These results imply that removing CSCs may prompt non-CSCs in the tumor to convert into stem cells to maintain the equilibrium. Interleukin-6 (IL-6) has been found to play an important role in the inducible formation of CSCs and their dynamic equilibrium with non-stem cells. In this study, we used CSC-like human breast cancer cells and their alternate subset non-CSCs to investigate how IL-6 regulates the conversion of non-CSCs to CSCs. MDA-MB-231 and MDA-MB-453 CSC-like cells formed mammospheres well, whereas most of non-stem cells died by anoikis and only part of the remaining non-stem cells produced viable mammospheres. Similar results were observed in xenograft tumor formation. Data from cytokine array assay show that IL-6 was secreted from non-CSCs when cells were cultured in ultra-low attachment plates. IL-6 regulates CSC-associated OCT-4 gene expression through the IL-6-JAK1-STAT3 signal transduction pathway in non-CSCs. Inhibiting this pathway by treatment with anti-IL-6 antibody (1 μg/ml) or niclosamide (0.5-2 μM)/LLL12 (5-10 μM) effectively prevented OCT-4 gene expression. These results suggest that the IL-6-JAK1-STAT3 signal transduction pathway plays an important role in the conversion of non-CSCs into CSCs through regulation of OCT-4 gene expression.
Collapse
Affiliation(s)
- Seog-Young Kim
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | |
Collapse
|
193
|
|
194
|
Role of the microenvironment in ovarian cancer stem cell maintenance. BIOMED RESEARCH INTERNATIONAL 2012; 2013:630782. [PMID: 23484135 PMCID: PMC3591167 DOI: 10.1155/2013/630782] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 11/08/2012] [Accepted: 11/08/2012] [Indexed: 12/20/2022]
Abstract
Despite recent progresses in cancer therapy and increased knowledge in cancer biology, ovarian cancer remains a challenging condition. Among the latest concepts developed in cancer biology, cancer stem cells and the role of microenvironment in tumor progression seem to be related. Indeed, cancer stem cells have been described in several solid tumors including ovarian cancers. These particular cells have the ability to self-renew and reconstitute a heterogeneous tumor. They are characterized by specific surface markers and display resistance to therapeutic regimens. During development, specific molecular cues from the tumor microenvironment can play a role in maintaining and expanding stemness of cancer cells. The tumor stroma contains several compartments: cellular component, cytokine network, and extracellular matrix. These different compartments interact to form a permissive niche for the cancer stem cells. Understanding the molecular cues underlying this crosstalk will allow the design of new therapeutic regimens targeting the niche. In this paper, we will discuss the mechanisms implicated in the interaction between ovarian cancer stem cells and their microenvironment.
Collapse
|
195
|
Stoyanova T, Goldstein AS, Cai H, Drake JM, Huang J, Witte ON. Regulated proteolysis of Trop2 drives epithelial hyperplasia and stem cell self-renewal via β-catenin signaling. Genes Dev 2012; 26:2271-85. [PMID: 23070813 DOI: 10.1101/gad.196451.112] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The cell surface protein Trop2 is expressed on immature stem/progenitor-like cells and is overexpressed in many epithelial cancers. However the biological function of Trop2 in tissue maintenance and tumorigenesis remains unclear. In this study, we demonstrate that Trop2 is a regulator of self-renewal, proliferation, and transformation. Trop2 controls these processes through a mechanism of regulated intramembrane proteolysis that leads to cleavage of Trop2, creating two products: the extracellular domain and the intracellular domain. The intracellular domain of Trop2 is released from the membrane and accumulates in the nucleus. Heightened expression of the Trop2 intracellular domain promotes stem/progenitor self-renewal through signaling via β-catenin and is sufficient to initiate precursor lesions to prostate cancer in vivo. Importantly, we demonstrate that loss of β-catenin or Trop2 loss-of-function cleavage mutants abrogates Trop2-driven self-renewal and hyperplasia in the prostate. These findings suggest that heightened expression of Trop2 is selected for in epithelial cancers to enhance the stem-like properties of self-renewal and proliferation. Defining the mechanism of Trop2 function in self-renewal and transformation is essential to identify new therapeutic strategies to block Trop2 activation in cancer.
Collapse
Affiliation(s)
- Tanya Stoyanova
- Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California 90095, USA
| | | | | | | | | | | |
Collapse
|
196
|
Gopinath S, Malla R, Alapati K, Gorantla B, Gujrati M, Dinh DH, Rao JS. Cathepsin B and uPAR regulate self-renewal of glioma-initiating cells through GLI-regulated Sox2 and Bmi1 expression. Carcinogenesis 2012; 34:550-9. [PMID: 23222817 DOI: 10.1093/carcin/bgs375] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cancer-initiating cells comprise a heterogeneous population of undifferentiated cells with the capacity for self-renewal and high proliferative potential. We investigated the role of uPAR and cathepsin B in the maintenance of stem cell nature in glioma-initiating cells (GICs). Simultaneous knockdown of uPAR and cathepsin B significantly reduced the expression of CD133, Nestin, Sox2 and Bmi1 at the protein level and GLI1 and GLI2 at the messenger RNA level. Also, knockdown of uPAR and cathepsin B resulted in a reduction in the number of GICs as well as sphere size. These changes are mediated by Sox2 and Bmi1, downstream of hedgehog signaling. Addition of cyclopamine reduced the expression of Sox2 and Bmi1 along with GLI1 and GLI2 expression, induced differentiation and reduced subsphere formation of GICs thereby indicating that hedgehog signaling acts upstream of Sox2 and Bmi1. Further confirmation was obtained from increased luciferase expression under the control of a GLI-bound Sox2 and Bmi1 luciferase promoter. Simultaneous knockdown of uPAR and cathepsin B also reduced the expression of Nestin Sox2 and Bmi1 in vivo. Thus, our study highlights the importance of uPAR and cathepsin B in the regulation of malignant stem cell self-renewal through hedgehog components, Bmi1 and Sox2.
Collapse
Affiliation(s)
- Sreelatha Gopinath
- Department of Cancer Biology & Pharmacology, University of Illinois College of Medicine at Peoria, One Illini Drive, Peoria, IL 61656, USA
| | | | | | | | | | | | | |
Collapse
|
197
|
Vira D, Basak SK, Veena MS, Wang MB, Batra RK, Srivatsan ES. Cancer stem cells, microRNAs, and therapeutic strategies including natural products. Cancer Metastasis Rev 2012; 31:733-51. [PMID: 22752409 DOI: 10.1007/s10555-012-9382-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Embryonic stem cells divide continuously and differentiate into organs through the expression of specific transcription factors at specific time periods. Differentiated adult stem cells on the other hand remain in quiescent state and divide by receiving cues from the environment (extracellular matrix or niche), as in the case of wound healing from tissue injury or inflammation. Similarly, it is believed that cancer stem cells (CSCs), forming a smaller fraction of the tumor bulk, also remain in a quiescent state. These cells are capable of initiating and propagating neoplastic growth upon receiving environmental cues, such as overexpression of growth factors, cytokines, and chemokines. Candidate CSCs express distinct biomarkers that can be utilized for their identification and isolation. This review focuses on the known and candidate cancer stem cell markers identified in various solid tumors and the promising future of disease management and therapy targeted at these markers. The review also provides details on the differential expression of microRNAs (miRNAs), and the miRNA- and natural product-based therapies that could be applied for the treatment of cancer stem cells.
Collapse
Affiliation(s)
- Darshni Vira
- Department of Surgery, VAGLAHS West Los Angeles, Los Angeles, CA 90073, USA
| | | | | | | | | | | |
Collapse
|
198
|
Karamboulas C, Ailles L. Developmental signaling pathways in cancer stem cells of solid tumors. Biochim Biophys Acta Gen Subj 2012. [PMID: 23196196 DOI: 10.1016/j.bbagen.2012.11.008] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND The intricate regulation of several signaling pathways is essential for embryonic development and adult tissue homeostasis. Cancers commonly display aberrant activity within these pathways. A population of cells identified in several cancers, termed cancer stem cells (CSCs) show similar properties to normal stem cells and evidence suggests that altered developmental signaling pathways play an important role in maintaining CSCs and thereby the tumor itself. SCOPE OF REVIEW This review will focus on the roles of the Notch, Wnt and Hedgehog pathways in the brain, breast and colon cancers. We describe the roles these pathways play in normal tissue homeostasis through the regulation of stem cell fate in these three tissues, and the experimental evidence indicating that the role of these pathways in cancers of these is directly linked to CSCs. MAJOR CONCLUSIONS A large body of evidence is accumulating to indicate that the deregulation of Notch, Wnt and Hedgehog pathways play important roles in both normal and cancer stem cells. We are only beginning to understand how these pathways interact, how they are coordinated during normal development and adult tissue homeostasis, and how they are deregulated during cancer. However, it is becoming increasingly clear that if we are to target CSCs therapeutically, it will likely be necessary to develop combination therapies. GENERAL SIGNIFICANCE If CSCs are the driving force behind tumor maintenance and growth then understanding the molecular mechanisms regulating CSCs is essential. Such knowledge will contribute to better targeted therapies that could significantly enhance cancer treatments and patient survival. This article is part of a Special Issue entitled Biochemistry of Stem Cells.
Collapse
Affiliation(s)
- Christina Karamboulas
- Ontario Cancer Institute, Campbell Family Cancer Research Institute, University Health Network, Toronto, Ontario, Canada M5G 1L7
| | | |
Collapse
|
199
|
Kim SY, Rhee JG, Song X, Prochownik EV, Spitz DR, Lee YJ. Breast cancer stem cell-like cells are more sensitive to ionizing radiation than non-stem cells: role of ATM. PLoS One 2012. [PMID: 23185620 PMCID: PMC3503893 DOI: 10.1371/journal.pone.0050423] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
There are contradictory observations about the different radiosensitivities of cancer stem cells and cancer non-stem cells. To resolve these contradictory observations, we studied radiosensitivities by employing breast cancer stem cell (CSC)-like MDA-MB231 and MDA-MB453 cells as well as their corresponding non-stem cells. CSC-like cells proliferate without differentiating and have characteristics of tumor-initiating cells [1]. These cells were exposed to γ-rays (1.25–8.75 Gy) and survival curves were determined by colony formation. A final slope, D0, of the survival curve for each cell line was determined to measure radiosensitivity. The D0 of CSC-like and non-stem MDA-MB-453 cells were 1.16 Gy and 1.55 Gy, respectively. Similar results were observed in MDA-MB-231 cells (0.94 Gy vs. 1.56 Gy). After determination of radiosensitivity, we investigated intrinsic cellular determinants which influence radiosensitivity including cell cycle distribution, free-radical scavengers and DNA repair. We observed that even though cell cycle status and antioxidant content may contribute to differential radiosensitivity, differential DNA repair capacity may be a greater determinant of radiosensitivity. Unlike non-stem cells, CSC-like cells have little/no sublethal damage repair, a low intracellular level of ataxia telangiectasia mutated (ATM) and delay of γ-H2AX foci removal (DNA strand break repair). These results suggest that low DNA repair capacity is responsible for the high radiosensitivity of these CSC-like cells.
Collapse
Affiliation(s)
- Seog-Young Kim
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | | | | | | | | | | |
Collapse
|
200
|
Marsden CG, Wright MJ, Carrier L, Moroz K, Rowan BG. Disseminated breast cancer cells acquire a highly malignant and aggressive metastatic phenotype during metastatic latency in the bone. PLoS One 2012; 7:e47587. [PMID: 23173031 PMCID: PMC3500091 DOI: 10.1371/journal.pone.0047587] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 09/18/2012] [Indexed: 12/19/2022] Open
Abstract
Background Disseminated tumor cells (DTCs) in the bone marrow may exist in a dormant state for extended periods of time, maintaining the ability to proliferate upon activation, engraft at new sites, and form detectable metastases. However, understanding of the behavior and biology of dormant breast cancer cells in the bone marrow niche remains limited, as well as their potential involvement in tumor recurrence and metastasis. Therefore, the purpose of this study was to investigate the tumorigenicity and metastatic potential of dormant disseminated breast cancer cells (prior to activation) in the bone marrow. Methodology/Principal Findings Total bone marrow, isolated from mice previously injected with tumorspheres into the mammary fat pad, was injected into the mammary fat pad of NUDE mice. As a negative control, bone marrow isolated from non-injected mice was injected into the mammary fat pad of NUDE mice. The resultant tumors were analyzed by immunohistochemistry for expression of epithelial and mesenchymal markers. Mouse lungs, livers, and kidneys were analyzed by H+E staining to detect metastases. The injection of bone marrow isolated from mice previously injected with tumorspheres into the mammary fat pad, resulted in large tumor formation in the mammary fat pad 2 months post-injection. However, the injection of bone marrow isolated from non-injected mice did not result in tumor formation in the mammary fat pad. The DTC-derived tumors exhibited accelerated development of metastatic lesions within the lung, liver and kidney. The resultant tumors and the majority of metastatic lesions within the lung and liver exhibited a mesenchymal-like phenotype. Conclusions/Significance Dormant DTCs within the bone marrow are highly malignant upon injection into the mammary fat pad, with the accelerated development of metastatic lesions within the lung, liver and kidney. These results suggest the acquisition of a more aggressive phenotype of DTCs during metastatic latency within the bone marrow microenvironment.
Collapse
Affiliation(s)
- Carolyn G. Marsden
- Department of Structural and Cellular Biology, The Louisiana Cancer Research Consortium, Tulane University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Mary Jo Wright
- Department of Surgery, The Louisiana Cancer Research Consortium, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Latonya Carrier
- Department of Structural and Cellular Biology, The Louisiana Cancer Research Consortium, Tulane University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Krzysztof Moroz
- Section of Surgical Pathology and Cytopathology, Louisiana Cancer Research Consortium, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Brian G. Rowan
- Department of Structural and Cellular Biology, The Louisiana Cancer Research Consortium, Tulane University Health Sciences Center, New Orleans, Louisiana, United States of America
- * E-mail:
| |
Collapse
|