1
|
Ordaz-Ramos A, Diaz-Blancas J, Martínez-Cruz A, Castro-Oropeza R, Zampedri C, Romero-Rodríguez DP, Rodriguez-Dorantes M, Melendez-Zajgla J, Maldonado V, Vazquez-Santillan K. RANKL regulates differentially breast cancer stem cell properties through its RANK and LGR4 receptors. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119888. [PMID: 39662745 DOI: 10.1016/j.bbamcr.2024.119888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 10/31/2024] [Accepted: 12/02/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Breast cancer stem cells (BCSC) are a subpopulation responsible for cancer resistance and relapse. The receptor activator of nuclear factor kappa-Β ligand (RANKL) is a cytokine capable of activating RANK and LGR4 receptors. RANKL/RANK signaling maintains the self-renewal of BCSCs, however, the effect of RANKL via LGR4 remains unclear. Evidence from osteoclasts suggests that RANKL/LGR4 axis disrupts RANK signaling, leading to opposing cellular responses. Anti-RANKL inhibitors are potential agents for eradicating CSCs, but their effect on RANKL/LGR4 signal has not been demonstrated. OBJECTIVE This project aimed to elucidate the role of RANKL in regulating stemness depending on the expression of its receptors. METHODS We use in vitro and in vivo approaches to evaluate the effects of RANKL inhibition in stemness in low or high-LGR4 expressing cells. Furthermore, we analyze the effects of RANKL stimulation on the stemness of LGR4 or RANK overexpressing cells. Additionally, we evaluated the impact of RANKL/LGR4 signaling in the activity of Wnt/β-catenin and NF-κB signaling pathways. RESULTS Our findings indicated that elevated RANKL expression is related to a favorable prognosis in patients with high LGR4 levels. Furthermore, RANKL inhibition decreased BCSC properties in LGR4-low cell lines, while it promoted migration in LGR4-high cells. Additionally, the RANKL/RANK axis activated NF-κB signaling and enhanced BCSCs in RANK-overexpressing cells. In contrast, in LGR4-overexpressing cells, RANKL failed to activate NF-κB but instead inhibited the Wnt/β-catenin pathway, leading to a reduction in BCSCs. CONCLUSION Our findings suggest that RANKL exerts different responses according to the expression of its receptors.
Collapse
Affiliation(s)
- Alejandro Ordaz-Ramos
- Innovation and Precision Medicine Laboratory, Instituto Nacional de Medicina Genómica, Periférico Sur No.4809, Col Arenal Tepepan, Tlalpan, Mexico City C.P. 14610, Mexico; Posgrado en Ciencias Biológicas, Unidad de Posgrado, Edificio D, 1° Piso, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, Mexico City C.P. 04510, Mexico
| | - Jorge Diaz-Blancas
- Epigenetics Laboratory, Instituto Nacional de Medicina Genómica, Periférico Sur No.4809, Col Arenal Tepepan, Tlalpan, Mexico City C.P. 14610, Mexico
| | - Aketzalli Martínez-Cruz
- Innovation and Precision Medicine Laboratory, Instituto Nacional de Medicina Genómica, Periférico Sur No.4809, Col Arenal Tepepan, Tlalpan, Mexico City C.P. 14610, Mexico
| | - Rosario Castro-Oropeza
- Molecular Oncology Laboratory, Unidad de Investigación Médica en Enfermedades Oncológicas, Hospital de Oncologia, Centro Medico Nacional Siglo XXI, IMSS, Avenida Cuahuhtemoc 330, Col Doctores, Cuauhtemoc, Mexico City C.P. 06720, Mexico
| | - Cecilia Zampedri
- Multidisciplinary Zebrafish Laboratory, Department of Bioengineer, Escuela de Ingenieria y Ciencias, Instituto Tecnologico y de Estudios Superiores Monterrey, Mexico City, Mexico
| | - Damaris P Romero-Rodríguez
- Flow Citometry Laboratory, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosió Villegas", Calzada de Tlalpan 4502, Col Belisario Dominguez Secc 16, Tlalpan, C.P. 14080, Mexico; Laboratorio Nacional Conahcyt de Investigación y Diagnóstico por Inmunocitofluorometría (LANCIDI), Mexico City, Mexico
| | - Mauricio Rodriguez-Dorantes
- Oncogenomics Laboratory, Instituto Nacional de Medicina Genómica, Periférico Sur No.4809, Col Arenal Tepepan, Tlalpan, Mexico City C.P. 14610, Mexico
| | - Jorge Melendez-Zajgla
- Functional Cancer Genomics Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14160, Mexico
| | - Vilma Maldonado
- Epigenetics Laboratory, Instituto Nacional de Medicina Genómica, Periférico Sur No.4809, Col Arenal Tepepan, Tlalpan, Mexico City C.P. 14610, Mexico
| | - Karla Vazquez-Santillan
- Innovation and Precision Medicine Laboratory, Instituto Nacional de Medicina Genómica, Periférico Sur No.4809, Col Arenal Tepepan, Tlalpan, Mexico City C.P. 14610, Mexico.
| |
Collapse
|
2
|
Shimpi AA, Williams ED, Ling L, Tamir T, White FM, Fischbach C. Phosphoproteomic Changes Induced by Cell-Derived Matrix and Their Effect on Tumor Cell Migration and Cytoskeleton Remodeling. ACS Biomater Sci Eng 2023; 9:6835-6848. [PMID: 38015076 DOI: 10.1021/acsbiomaterials.3c01034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Increased fibrotic extracellular matrix (ECM) deposition promotes tumor invasion, which is the first step of the metastatic cascade. Yet, the underlying mechanisms are poorly understood as conventional studies of tumor cell migration are often performed in 2D cultures lacking the compositional and structural complexity of native ECM. Moreover, these studies frequently focus on select candidate pathways potentially overlooking other relevant changes in cell signaling. Here, we combine a cell-derived matrix (CDM) model with phosphotyrosine phosphoproteomic analysis to investigate tumor cell migration on fibrotic ECM relative to standard tissue culture plastic (TCP). Our results suggest that tumor cells cultured on CDMs migrate faster and in a more directional manner than their counterparts on TCP. These changes in migration correlate with decreased cell spreading and increased cell elongation. While the formation of phosphorylated focal adhesion kinase (pFAK)+ adhesion complexes did not vary between TCP and CDMs, time-dependent phosphoproteomic analysis identified that the SRC family kinase LYN may be differentially regulated. Pharmacological inhibition of LYN decreased tumor cell migration and cytoskeletal rearrangement on CDMs and also on TCP, suggesting that LYN regulates tumor cell migration on CDMs in combination with other mechanisms. These data highlight how the combination of physicochemically complex in vitro systems with phosphoproteomics can help identify signaling mechanisms by which the fibrotic ECM regulates tumor cell migration.
Collapse
Affiliation(s)
- Adrian A Shimpi
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Erik D Williams
- Department of Information Science, Cornell University, Ithaca, New York 14853, United States
| | - Lu Ling
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Tigist Tamir
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 023139, United States
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 023139, United States
| | - Forest M White
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 023139, United States
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 023139, United States
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
3
|
Tan ML, Jenkins-Johnston N, Huang S, Schutrum B, Vadhin S, Adhikari A, Williams RM, Zipfel WR, Lammerding J, Varner JD, Fischbach C. Endothelial cells metabolically regulate breast cancer invasion toward a microvessel. APL Bioeng 2023; 7:046116. [PMID: 38058993 PMCID: PMC10697723 DOI: 10.1063/5.0171109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/06/2023] [Indexed: 12/08/2023] Open
Abstract
Breast cancer metastasis is initiated by invasion of tumor cells into the collagen type I-rich stroma to reach adjacent blood vessels. Prior work has identified that metabolic plasticity is a key requirement of tumor cell invasion into collagen. However, it remains largely unclear how blood vessels affect this relationship. Here, we developed a microfluidic platform to analyze how tumor cells invade collagen in the presence and absence of a microvascular channel. We demonstrate that endothelial cells secrete pro-migratory factors that direct tumor cell invasion toward the microvessel. Analysis of tumor cell metabolism using metabolic imaging, metabolomics, and computational flux balance analysis revealed that these changes are accompanied by increased rates of glycolysis and oxygen consumption caused by broad alterations of glucose metabolism. Indeed, restricting glucose availability decreased endothelial cell-induced tumor cell invasion. Our results suggest that endothelial cells promote tumor invasion into the stroma due, in part, to reprogramming tumor cell metabolism.
Collapse
Affiliation(s)
- Matthew L. Tan
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Niaa Jenkins-Johnston
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Sarah Huang
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Brittany Schutrum
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Sandra Vadhin
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Abhinav Adhikari
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Rebecca M. Williams
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Warren R. Zipfel
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Jan Lammerding
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Jeffrey D. Varner
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, USA
| | | |
Collapse
|
4
|
Cui J, Christin JR, Reisz JA, Cendali FI, Sanawar R, Coutinho De Miranda M, D’Alessandro A, Guo W. Targeting ABCA12-controlled ceramide homeostasis inhibits breast cancer stem cell function and chemoresistance. SCIENCE ADVANCES 2023; 9:eadh1891. [PMID: 38039374 PMCID: PMC10691781 DOI: 10.1126/sciadv.adh1891] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 11/01/2023] [Indexed: 12/03/2023]
Abstract
Cancer stem cells (CSCs) drive tumor growth, metastasis, and chemoresistance. While emerging evidence suggests that CSCs have a unique dependency on lipid metabolism, the functions and regulation of distinct lipid species in CSCs remain poorly understood. Here, we developed a stem cell factor SOX9-based reporter for isolating CSCs in primary tumors and metastases of spontaneous mammary tumor models. Transcriptomic analyses uncover that SOX9high CSCs up-regulate the ABCA12 lipid transporter. ABCA12 down-regulation impairs cancer stemness and chemoresistance. Lipidomic analyses reveal that ABCA12 maintains cancer stemness and chemoresistance by reducing intracellular ceramide abundance, identifying a CSC-associated function of ABCA subfamily transporter. Ceramide suppresses cancer stemness by inhibiting the YAP-SOX9 signaling pathway in CSCs. Increasing ceramide levels in tumors enhances their sensitivity to chemotherapy and prevents the enrichment of SOX9high CSCs. In addition, SOX9high and ABCA12high cancer cells contribute to chemoresistance in human patient-derived xenografts. These findings identify a CSC-suppressing lipid metabolism pathway that can be exploited to inhibit CSCs and overcome chemoresistance.
Collapse
Affiliation(s)
- Jihong Cui
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - John R. Christin
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Julie A. Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Francesca Isabelle Cendali
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Rahul Sanawar
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Marcelo Coutinho De Miranda
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Wenjun Guo
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Montefiore Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
5
|
Yamashita N, Withers H, Morimoto Y, Bhattacharya A, Haratake N, Daimon T, Fushimi A, Nakashoji A, Thorner AR, Isenhart E, Rosario S, Long MD, Kufe D. MUC1-C integrates aerobic glycolysis with suppression of oxidative phosphorylation in triple-negative breast cancer stem cells. iScience 2023; 26:108168. [PMID: 37915591 PMCID: PMC10616323 DOI: 10.1016/j.isci.2023.108168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/17/2023] [Accepted: 10/05/2023] [Indexed: 11/03/2023] Open
Abstract
Activation of the MUC1-C protein promotes lineage plasticity, epigenetic reprogramming, and the cancer stem cell (CSC) state. The present studies performed on enriched populations of triple-negative breast cancer (TNBC) CSCs demonstrate that MUC1-C is essential for integrating activation of glycolytic pathway genes with self-renewal and tumorigenicity. MUC1-C further integrates the glycolytic pathway with suppression of mitochondrial DNA (mtDNA) genes encoding components of mitochondrial Complexes I-V. The repression of mtDNA genes is explained by MUC1-C-mediated (i) downregulation of the mitochondrial transcription factor A (TFAM) required for mtDNA transcription and (ii) induction of the mitochondrial transcription termination factor 3 (mTERF3). In support of pathogenesis that suppresses mitochondrial ROS production, targeting MUC1-C increases (i) mtDNA gene transcription, (ii) superoxide levels, and (iii) loss of self-renewal capacity. These findings and scRNA-seq analysis of CSC subpopulations indicate that MUC1-C regulates self-renewal and redox balance by integrating activation of glycolysis with suppression of oxidative phosphorylation.
Collapse
Affiliation(s)
- Nami Yamashita
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Henry Withers
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | | | - Naoki Haratake
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Tatsuaki Daimon
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Atsushi Fushimi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Ayako Nakashoji
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Aaron R. Thorner
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Emily Isenhart
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Spencer Rosario
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Mark D. Long
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Donald Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
6
|
Choi S, Whitman MA, Shimpi AA, Sempertegui ND, Chiou AE, Druso JE, Verma A, Lux SC, Cheng Z, Paszek M, Elemento O, Estroff LA, Fischbach C. Bone-matrix mineralization dampens integrin-mediated mechanosignalling and metastatic progression in breast cancer. Nat Biomed Eng 2023; 7:1455-1472. [PMID: 37550422 DOI: 10.1038/s41551-023-01077-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/30/2023] [Indexed: 08/09/2023]
Abstract
In patients with breast cancer, lower bone mineral density increases the risk of bone metastasis. Although the relationship between bone-matrix mineralization and tumour-cell phenotype in breast cancer is not well understood, mineralization-induced rigidity is thought to drive metastatic progression via increased cell-adhesion forces. Here, by using collagen-based matrices with adjustable intrafibrillar mineralization, we show that, unexpectedly, matrix mineralization dampens integrin-mediated mechanosignalling and induces a less proliferative stem-cell-like phenotype in breast cancer cells. In mice with xenografted decellularized physiological bone matrices seeded with human breast tumour cells, the presence of bone mineral reduced tumour growth and upregulated a gene-expression signature that is associated with longer metastasis-free survival in patients with breast cancer. Our findings suggest that bone-matrix changes in osteogenic niches regulate metastatic progression in breast cancer and that in vitro models of bone metastasis should integrate organic and inorganic matrix components to mimic physiological and pathologic mineralization.
Collapse
Affiliation(s)
- Siyoung Choi
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Matthew A Whitman
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Adrian A Shimpi
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Nicole D Sempertegui
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Aaron E Chiou
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Joseph E Druso
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Akanksha Verma
- Caryl and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Stephanie C Lux
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Zhu Cheng
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA
| | - Matthew Paszek
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA
| | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Lara A Estroff
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, USA.
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY, USA.
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
7
|
Bednarek R, Wojkowska DW, Braun M, Watala C, Salifu MO, Swiatkowska M, Babinska A. Triple negative breast cancer metastasis is hindered by a peptide antagonist of F11R/JAM‑A protein. Cancer Cell Int 2023; 23:160. [PMID: 37563645 PMCID: PMC10416405 DOI: 10.1186/s12935-023-03023-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/06/2023] [Indexed: 08/12/2023] Open
Abstract
BACKGROUND The F11R/JAM-A cell adhesion protein was examined as the therapeutic target in triple negative breast cancer (TNBC) with the use of the peptide antagonist to F11R/JAM-A, that previously inhibited the early stages of breast cancer metastasis in vitro. METHODS The online in silico analysis was performed by TNMPlot, UALCAN, and KM plotter. The in vitro experiments were performed to verify the effect of peptide 4D (P4D) on human endothelial cell lines EA.hy926 and HMEC-1 as well as on human TNBC cell line MDA-MB-231. The cell morphology upon P4D treatment was verified by light microscopy, while the cell functions were assessed by colony forming assay, MTT cell viability assay, BrdU cell proliferation assay, and Transepithelial/Endothelial Electrical Resistance measurements. The in vivo experiments on 4T1 murine breast cancer model were followed by histopathological analysis and a series of quantitative analyses of murine tissues. RESULTS By in silico analysis we have found the elevated gene expression in breast cancer with particular emphasis on TNBC. The elevated F11R expression in TNBC was related with poorer survival prognosis. Peptide 4D has altered the morphology and increased the permeability of endothelial monolayers. The colony formation, viability, and proliferation of MDA-MB-231 cells were decreased. P4D inhibited the metastasis in 4T1 breast cancer murine model in a statistically significant manner that was demonstrated by the resampling bootstrap technique. CONCLUSIONS The P4D peptide antagonist to F11R/JAM-A is able to hinder the metastasis in TNBC. This assumption needs to be confirmed by additional 4T1 mouse model study performed on larger group size, before making the decision on human clinical trials.
Collapse
Affiliation(s)
- Radosław Bednarek
- Department of Cytobiology and Proteomics, Chair of Biomedical Sciences, Medical University of Lodz, ul. Mazowiecka 6/8, 92-215, Lodz, Poland.
| | - Dagmara W Wojkowska
- Department of Haemostasis and Haemostatic Disorders, Medical University of Lodz, Lodz, Poland
| | - Marcin Braun
- Department of Pathology, Chair of Oncology, Medical University of Lodz, Lodz, Poland
| | - Cezary Watala
- Department of Haemostasis and Haemostatic Disorders, Medical University of Lodz, Lodz, Poland
| | - Moro O Salifu
- Department of Medicine, Downstate Medical Center, State University of New York, Brooklyn, NY, USA
| | - Maria Swiatkowska
- Department of Cytobiology and Proteomics, Chair of Biomedical Sciences, Medical University of Lodz, ul. Mazowiecka 6/8, 92-215, Lodz, Poland
| | - Anna Babinska
- Department of Medicine, Downstate Medical Center, State University of New York, Brooklyn, NY, USA
| |
Collapse
|
8
|
Shimpi AA, Tan ML, Vilkhovoy M, Dai D, Roberts LM, Kuo J, Huang L, Varner JD, Paszek M, Fischbach C. Convergent Approaches to Delineate the Metabolic Regulation of Tumor Invasion by Hyaluronic Acid Biosynthesis. Adv Healthc Mater 2023; 12:e2202224. [PMID: 36479976 PMCID: PMC10238572 DOI: 10.1002/adhm.202202224] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/21/2022] [Indexed: 12/13/2022]
Abstract
Metastasis is the leading cause of breast cancer-related deaths and is often driven by invasion and cancer-stem like cells (CSCs). Both the CSC phenotype and invasion are associated with increased hyaluronic acid (HA) production. How these independent observations are connected, and which role metabolism plays in this process, remains unclear due to the lack of convergent approaches integrating engineered model systems, computational tools, and cancer biology. Using microfluidic invasion models, metabolomics, computational flux balance analysis, and bioinformatic analysis of patient data, the functional links between the stem-like, invasive, and metabolic phenotype of breast cancer cells as a function of HA biosynthesis are investigated. These results suggest that CSCs are more invasive than non-CSCs and that broad metabolic changes caused by overproduction of HA play a role in this process. Accordingly, overexpression of hyaluronic acid synthases (HAS) 2 or 3 induces a metabolic phenotype that promotes cancer cell stemness and invasion in vitro and upregulates a transcriptomic signature predictive of increased invasion and worse patient survival. This study suggests that HA overproduction leads to metabolic adaptations to satisfy the energy demands for 3D invasion of breast CSCs highlighting the importance of engineered model systems and multidisciplinary approaches in cancer research.
Collapse
Affiliation(s)
- Adrian A. Shimpi
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, 14853, USA
| | - Matthew L. Tan
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, 14853, USA
| | - Michael Vilkhovoy
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, 14853, USA
| | - David Dai
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, 14853, USA
| | - L. Monet Roberts
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, 14853, USA
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, 14853, USA
| | - Joe Kuo
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, 14853, USA
| | - Lingting Huang
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, 14853, USA
| | - Jeffrey D. Varner
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, 14853, USA
| | - Matthew Paszek
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, 14853, USA
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, New York, 14853, USA
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, 14853, USA
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, New York, 14853, USA
| |
Collapse
|
9
|
Tian Y, Zhang P, Mou Y, Yang W, Zhang J, Li Q, Dou X. Silencing Notch4 promotes tumorigenesis and inhibits metastasis of triple-negative breast cancer via Nanog and Cdc42. Cell Death Discov 2023; 9:148. [PMID: 37149651 PMCID: PMC10164131 DOI: 10.1038/s41420-023-01450-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/18/2023] [Accepted: 04/24/2023] [Indexed: 05/08/2023] Open
Abstract
Elucidation of individual Notch protein biology in specific cancer is crucial to develop safe, effective, and tumor-selective Notch-targeting therapeutic reagents for clinical use [1]. Here, we explored the Notch4 function in triple-negative breast cancer (TNBC). We found that silencing Notch4 enhanced tumorigenic ability in TNBC cells via upregulating Nanog expression, a pluripotency factor of embryonic stem cells. Intriguingly, silencing Notch4 in TNBC cells suppressed metastasis via downregulating Cdc42 expression, a key molecular for cell polarity formation. Notably, downregulation of Cdc42 expression affected Vimentin distribution, but not Vimentin expression to inhibit EMT shift. Collectively, our results show that silencing Notch4 enhances tumorigenesis and inhibits metastasis in TNBC, indicating that targeting Notch4 may not be a potential strategy for drug discovery in TNBC.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China
- Clinical Research Center, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China
| | - Peipei Zhang
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China
- Clinical Research Center, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China
| | - Yajun Mou
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China
- Clinical Research Center, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China
| | - Wenxiu Yang
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China
| | - Junhong Zhang
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China
| | - Qing Li
- Department of Orthopedics, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China
| | - Xiaowei Dou
- Clinical Research Center, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China.
| |
Collapse
|
10
|
Le Minh G, Esquea EM, Dhameliya TT, Merzy J, Lee MH, Ball LE, Reginato MJ. Kruppel-like factor 8 regulates triple negative breast cancer stem cell-like activity. Front Oncol 2023; 13:1141834. [PMID: 37152043 PMCID: PMC10155275 DOI: 10.3389/fonc.2023.1141834] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/29/2023] [Indexed: 05/09/2023] Open
Abstract
Introduction Breast tumor development is regulated by a sub-population of breast cancer cells, termed cancer stem-like cells (CSC), which are capable of self-renewing and differentiating, and are involved in promoting breast cancer invasion, metastasis, drug resistance and relapse. CSCs are highly adaptable, capable of reprogramming their own metabolism and signaling activity in response to stimuli within the tumor microenvironment. Recently, the nutrient sensor O-GlcNAc transferase (OGT) and O-GlcNAcylation was shown to be enriched in CSC populations, where it promotes the stemness and tumorigenesis of breast cancer cells in vitro and in vivo. This enrichment was associated with upregulation of the transcription factor Kruppel-like-factor 8 (KLF8) suggesting a potential role of KLF8 in regulating CSCs properties. Methods Triple-negative breast cancer cells were genetically modified to generate KLF8 overexpressing or KLF8 knock-down cells. Cancer cells, control or with altered KLF8 expression were analyzed to assess mammosphere formation efficiency, CSCs frequency and expression of CSCs factors. Tumor growth in vivo of control or KLF8 knock-down cells was assessed by fat-pad injection of these cell in immunocompromised mice. Results Here, we show that KLF8 is required and sufficient for regulating CSC phenotypes and regulating transcription factors SOX2, NANOG, OCT4 and c-MYC. KLF8 levels are associated with chemoresistance in triple negative breast cancer patients and overexpression in breast cancer cells increased paclitaxel resistance. KLF8 and OGT co-regulate each other to form a feed-forward loop to promote CSCs phenotype and mammosphere formation of breast cancer cells. Discussion These results suggest a critical role of KLF8 and OGT in promoting CSCs and cancer progression, that may serve as potential targets for developing strategy to target CSCs specifically.
Collapse
Affiliation(s)
- Giang Le Minh
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Emily M. Esquea
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Tejsi T. Dhameliya
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Jessica Merzy
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Mi-Hye Lee
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
| | - Lauren E. Ball
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
| | - Mauricio J. Reginato
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, United States
- Translational and Cellular Oncology Program, Sidney Kimmel Cancer Center of Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
11
|
Heussner RK, Zhang H, Qian G, Baker MJ, Provenzano PP. Differential contractility regulates cancer stem cell migration. Biophys J 2023; 122:1198-1210. [PMID: 36772795 PMCID: PMC10111274 DOI: 10.1016/j.bpj.2023.02.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 10/22/2022] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
Cancer stem cells (CSCs) are known to have a high capacity for tumor initiation and the formation of metastases. We have previously shown that in collagen constructs mimetic of aligned extracellular matrix architectures observed in carcinomas, breast CSCs demonstrate enhanced directional and total motility compared with more differentiated carcinoma populations. Here, we show that CSCs maintain increased motility in diverse environments including on 2D elastic polyacrylamide gels of various stiffness, 3D randomly oriented collagen matrices, and ectopic cerebral slices representative of a common metastatic site. A consistent twofold increase of CSC motility across platforms suggests a general shift in cell migration mechanics between well-differentiated carcinoma cells and their stem-like counterparts. To further elucidate the source of differences in migration, we demonstrate that CSCs are less contractile than the whole population (WP) and develop fewer and smaller focal adhesions and show that enhanced CSC migration can be tuned via contractile forces. The WP can be shifted to a CSC-like migratory phenotype using partial myosin II inhibition. Inversely, CSCs can be shifted to a less migratory WP-like phenotype using microtubule-destabilizing drugs that increase contractility or by directly enhancing contractile forces. This work begins to reveal the mechanistic differences driving CSC migration and raises important implications regarding the potentially disparate effects of microtubule-targeting agents on the motility of different cell populations.
Collapse
Affiliation(s)
- Rachel K Heussner
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota; University of Minnesota Physical Sciences in Oncology Center, Minneapolis, Minnesota
| | - Hongrong Zhang
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota; University of Minnesota Physical Sciences in Oncology Center, Minneapolis, Minnesota; University of Minnesota Center for Multiparametric Imaging of Tumor Immune Microenvironments, Minneapolis, Minnesota
| | - Guhan Qian
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota; University of Minnesota Physical Sciences in Oncology Center, Minneapolis, Minnesota; University of Minnesota Center for Multiparametric Imaging of Tumor Immune Microenvironments, Minneapolis, Minnesota
| | - Mikayla J Baker
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota; University of Minnesota Physical Sciences in Oncology Center, Minneapolis, Minnesota; University of Minnesota Center for Multiparametric Imaging of Tumor Immune Microenvironments, Minneapolis, Minnesota
| | - Paolo P Provenzano
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota; University of Minnesota Physical Sciences in Oncology Center, Minneapolis, Minnesota; University of Minnesota Center for Multiparametric Imaging of Tumor Immune Microenvironments, Minneapolis, Minnesota; Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota; Department of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota; Institute for Engineering in Medicine, University of Minnesota, Minneapolis, Minnesota; Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
12
|
Zhou L, Liu L, Chang MA, Ma C, Chen W, Chen P. Spatiotemporal dissection of tumor microenvironment via in situ sensing and monitoring in tumor-on-a-chip. Biosens Bioelectron 2023; 225:115064. [PMID: 36680970 PMCID: PMC9918721 DOI: 10.1016/j.bios.2023.115064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
Real-time monitoring in the tumor microenvironment provides critical insights of cancer progression and mechanistic understanding of responses to cancer treatments. However, clinical challenges and significant questions remain regarding assessment of limited clinical tissue samples, establishment of validated, controllable pre-clinical cancer models, monitoring of static versus dynamic markers, and the translation of insights gained from in vitro tumor microenvironments to systematic investigation and understanding in clinical practice. State-of-art tumor-on-a-chip strategies will be reviewed herein, and emerging real-time sensing and monitoring platforms for on-chip analysis of tumor microenvironment will also be examined. The integration of the sensors with tumor-on-a-chip platforms to provide spatiotemporal information of the tumor microenvironment and the associated challenges will be further evaluated. Though optimal integrated systems for in situ monitoring are still in evolution, great promises lie ahead that will open new paradigm for rapid, comprehensive analysis of cancer development and assist clinicians with powerful tools to guide the diagnosis, prognosis and treatment course in cancer.
Collapse
Affiliation(s)
- Lang Zhou
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, 36849, USA
| | - Lunan Liu
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA; Department of Biomedical Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Muammar Ali Chang
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, 36849, USA
| | - Chao Ma
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA; Department of Biomedical Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA; Department of Biomedical Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Pengyu Chen
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, 36849, USA.
| |
Collapse
|
13
|
Jin G, Zhang Z, Wan J, Wu X, Liu X, Zhang W. G3BP2: Structure and Function. Pharmacol Res 2022; 186:106548. [DOI: 10.1016/j.phrs.2022.106548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/20/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
|
14
|
CRISPR screening in cancer stem cells. Essays Biochem 2022; 66:305-318. [PMID: 35713228 DOI: 10.1042/ebc20220009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/04/2022] [Accepted: 06/07/2022] [Indexed: 12/14/2022]
Abstract
Cancer stem cells (CSCs) are a subpopulation of tumor cells with self-renewal ability. Increasing evidence points to the critical roles of CSCs in tumorigenesis, metastasis, therapy resistance, and cancer relapse. As such, the elimination of CSCs improves cancer treatment outcomes. However, challenges remain due to limited understanding of the molecular mechanisms governing self-renewal and survival of CSCs. Clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 screening has been increasingly used to identify genetic determinants in cancers. In this primer, we discuss the progress made and emerging opportunities of coupling advanced CRISPR screening systems with CSC models to reveal the understudied vulnerabilities of CSCs.
Collapse
|
15
|
Czubak-Prowizor K, Babinska A, Swiatkowska M. The F11 Receptor (F11R)/Junctional Adhesion Molecule-A (JAM-A) (F11R/JAM-A) in cancer progression. Mol Cell Biochem 2021; 477:79-98. [PMID: 34533648 PMCID: PMC8755661 DOI: 10.1007/s11010-021-04259-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 09/08/2021] [Indexed: 12/27/2022]
Abstract
The F11 Receptor (F11R), also called Junctional Adhesion Molecule-A (JAM-A) (F11R/JAM-A), is a transmembrane glycoprotein of the immunoglobulin superfamily, which is mainly located in epithelial and endothelial cell tight junctions and also expressed on circulating platelets and leukocytes. It participates in the regulation of various biological processes, as diverse as paracellular permeability, tight junction formation and maintenance, leukocyte transendothelial migration, epithelial-to-mesenchymal transition, angiogenesis, reovirus binding, and platelet activation. Dysregulation of F11R/JAM-A may result in pathological consequences and disorders in normal cell function. A growing body of evidence points to its role in carcinogenesis and invasiveness, but its tissue-specific pro- or anti-tumorigenic role remains a debated issue. The following review focuses on the F11R/JAM-A tissue-dependent manner in tumorigenesis and metastasis and also discusses the correlation between poor patient clinical outcomes and its aberrant expression. In the future, it will be required to clarify the signaling pathways that are activated or suppressed via the F11R/JAM-A protein in various cancer types to understand its multiple roles in cancer progression and further use it as a novel direct target for cancer treatment.
Collapse
Affiliation(s)
- Kamila Czubak-Prowizor
- Department of Cytobiology and Proteomics, Medical University of Lodz, 6/8 Mazowiecka St., 92-215, Lodz, Poland.
| | - Anna Babinska
- Department of Medicine, State University of New York Downstate Medical Center, 450 Clarkson Ave, Brooklyn, NY, 11203, USA
| | - Maria Swiatkowska
- Department of Cytobiology and Proteomics, Medical University of Lodz, 6/8 Mazowiecka St., 92-215, Lodz, Poland
| |
Collapse
|
16
|
Salinas-Jazmín N, Rosas-Cruz A, Velasco-Velázquez M. Reporter gene systems for the identification and characterization of cancer stem cells. World J Stem Cells 2021; 13:861-876. [PMID: 34367481 PMCID: PMC8316869 DOI: 10.4252/wjsc.v13.i7.861] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/19/2021] [Accepted: 07/05/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs) are tumor cells that share functional characteristics with normal and embryonic stem cells. CSCs have increased tumor-initiating capacity and metastatic potential and lower sensitivity to chemo- and radiotherapy, with important roles in tumor progression and the response to therapy. Thus, a current goal of cancer research is to eliminate CSCs, necessitating an adequate phenotypic and functional characterization of CSCs. Strategies have been developed to identify, enrich, and track CSCs, many of which distinguish CSCs by evaluating the expression of surface markers, the initiation of specific signaling pathways, and the activation of master transcription factors that control stemness in normal cells. We review and discuss the use of reporter gene systems for identifying CSCs. Reporters that are under the control of aldehyde dehydrogenase 1A1, CD133, Notch, Nanog homeobox, Sex-determining region Y-box 2, and POU class 5 homeobox can be used to identify CSCs in many tumor types, track cells in real time, and screen for drugs. Thus, reporter gene systems, in combination with in vitro and in vivo functional assays, can assess changes in the CSCs pool. We present relevant examples of these systems in the evaluation of experimental CSCs-targeting therapeutics, demonstrating their value in CSCs research.
Collapse
Affiliation(s)
- Nohemí Salinas-Jazmín
- Department of Pharmacology, School of Medicine, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Arely Rosas-Cruz
- Department of Pharmacology, School of Medicine, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Marco Velasco-Velázquez
- Department of Pharmacology, School of Medicine, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico.
| |
Collapse
|
17
|
Mohan A, Raj Rajan R, Mohan G, Kollenchery Puthenveettil P, Maliekal TT. Markers and Reporters to Reveal the Hierarchy in Heterogeneous Cancer Stem Cells. Front Cell Dev Biol 2021; 9:668851. [PMID: 34150761 PMCID: PMC8209516 DOI: 10.3389/fcell.2021.668851] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022] Open
Abstract
A subpopulation within cancer, known as cancer stem cells (CSCs), regulates tumor initiation, chemoresistance, and metastasis. At a closer look, CSCs show functional heterogeneity and hierarchical organization. The present review is an attempt to assign marker profiles to define the functional heterogeneity and hierarchical organization of CSCs, based on a series of single-cell analyses. The evidences show that analogous to stem cell hierarchy, self-renewing Quiescent CSCs give rise to the Progenitor CSCs with limited proliferative capacity, and later to a Progenitor-like CSCs, which differentiates to Proliferating non-CSCs. Functionally, the CSCs can be tumor-initiating cells (TICs), drug-resistant CSCs, or metastasis initiating cells (MICs). Although there are certain marker profiles used to identify CSCs of different cancers, molecules like CD44, CD133, ALDH1A1, ABCG2, and pluripotency markers [Octamer binding transcriptional factor 4 (OCT4), SOX2, and NANOG] are used to mark CSCs of a wide range of cancers, ranging from hematological malignancies to solid tumors. Our analysis of the recent reports showed that a combination of these markers can demarcate the heterogeneous CSCs in solid tumors. Reporter constructs are widely used for easy identification and quantification of marker molecules. In this review, we discuss the suitability of reporters for the widely used CSC markers that can define the heterogeneous CSCs. Since the CSC-specific functions of CD44 and CD133 are regulated at the post-translational level, we do not recommend the reporters for these molecules for the detection of CSCs. A promoter-based reporter for ABCG2 may also be not relevant in CSCs, as the expression of the molecule in cancer is mainly regulated by promoter demethylation. In this context, a dual reporter consisting of one of the pluripotency markers and ALDH1A1 will be useful in marking the heterogeneous CSCs. This system can be easily adapted to high-throughput platforms to screen drugs for eliminating CSCs.
Collapse
Affiliation(s)
- Amrutha Mohan
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India.,Manipal Academy of Higher Education, Manipal, India
| | - Reshma Raj Rajan
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Gayathri Mohan
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | | | | |
Collapse
|
18
|
Mohan A, Raj R R, Mohan G, K P P, Thomas Maliekal T. Reporters of Cancer Stem Cells as a Tool for Drug Discovery. Front Oncol 2021; 11:669250. [PMID: 33968778 PMCID: PMC8100607 DOI: 10.3389/fonc.2021.669250] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 03/29/2021] [Indexed: 01/04/2023] Open
Abstract
In view of the importance of cancer stem cells (CSCs) in chemoresistance, metastasis and recurrence, the biology of CSCs were explored in detail. Based on that, several modalities were proposed to target them. In spite of the several clinical trials, a successful CSC-targeting drug is yet to be identified. The number of molecules screened and entered for clinical trial for CSC-targeting is comparatively low, compared to other drugs. The bottle neck is the lack of a high-throughput adaptable screening strategy for CSCs. This review is aimed to identify suitable reporters for CSCs that can be used to identify the heterogeneous CSC populations, including quiescent CSCs, proliferative CSCs, drug resistant CSCs and metastatic CSCs. Analysis of the tumor microenvironment regulating CSCs revealed that the factors in CSC-niche activates effector molecules that function as CSC markers, including pluripotency markers, CD133, ABCG2 and ALDH1A1. Among these factors OCT4, SOX2, NANOG, ABCG2 and ALDH1A1 are ideal for making reporters for CSCs. The pluripotency molecules, like OCT4, SOX2 and NANOG, regulate self-renewal, chemoresistance and metastasis. ABCG2 is a known regulator of drug resistance while ALDH1A1 modulates self-renewal, chemoresistance and metastasis. Considering the heterogeneity of CSCs, including a quiescent population and a proliferative population with metastatic ability, we propose the use of a combination of reporters. A dual reporter consisting of a pluripotency marker and a marker like ALDH1A1 will be useful in screening drugs that target CSCs.
Collapse
Affiliation(s)
- Amrutha Mohan
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India.,Centre for Doctoral Studies, Manipal Academy of Higher Education, Manipal, India
| | - Reshma Raj R
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Gayathri Mohan
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Padmaja K P
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | | |
Collapse
|
19
|
Bao B, Teslow EA, Mitrea C, Boerner JL, Dyson G, Bollig-Fischer A. Role of TET1 and 5hmC in an Obesity-Linked Pathway Driving Cancer Stem Cells in Triple-Negative Breast Cancer. Mol Cancer Res 2020; 18:1803-1814. [PMID: 32913111 PMCID: PMC7718329 DOI: 10.1158/1541-7786.mcr-20-0359] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/05/2020] [Accepted: 09/03/2020] [Indexed: 01/03/2023]
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer that lacks expression of estrogen receptor, progesterone receptor, and the HER2 but is enriched with cancer stem cell-like cells (CSC). CSCs are the fraction of cancer cells recognized as the source of primary malignant tumors that also give rise to metastatic recurrence. 5-Hydroxymethylcytosine (5hmC) is a DNA epigenetic feature derived from 5-methylcytosine by action of tet methylcytosine dioxygenase enzymes (e.g., TET1); and although TET1 and 5hmC are required to maintain embryonic stem cells, the mechanism and role in CSCs remain unknown. Data presented in this report support the conclusion that TET1 and TET1-dependent 5hmC mediate hydrogen peroxide (H2O2)-dependent activation of a novel gene expression cascade driving self-renewal and expansion of CSCs in TNBC. Evidence presented also supports that the H2O2 affecting this pathway arises due to endogenous mechanisms-including downregulation of antioxidant enzyme catalase in TNBC cells-and by exogenous routes, such as systemic inflammation and oxidative stress coupled with obesity, a known risk factor for TNBC incidence and recurrence. IMPLICATIONS: This study elucidates a pathway dependent on H2O2 and linked to obesity-driven TNBC tumor-initiating CSCs; thus, it provides new understanding that may advance TNBC prevention and treatment strategies.
Collapse
Affiliation(s)
- Bin Bao
- Barbara Ann Karmanos Cancer Institute and Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
| | - Emily A Teslow
- Barbara Ann Karmanos Cancer Institute and Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
| | - Cristina Mitrea
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Julie L Boerner
- Barbara Ann Karmanos Cancer Institute and Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
| | - Greg Dyson
- Barbara Ann Karmanos Cancer Institute and Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
| | - Aliccia Bollig-Fischer
- Barbara Ann Karmanos Cancer Institute and Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.
| |
Collapse
|
20
|
Lauko A, Mu Z, Gutmann DH, Naik UP, Lathia JD. Junctional Adhesion Molecules in Cancer: A Paradigm for the Diverse Functions of Cell-Cell Interactions in Tumor Progression. Cancer Res 2020; 80:4878-4885. [PMID: 32816855 DOI: 10.1158/0008-5472.can-20-1829] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/15/2020] [Accepted: 08/07/2020] [Indexed: 01/22/2023]
Abstract
Tight junction (TJ) proteins are essential for mediating interactions between adjacent cells and coordinating cellular and organ responses. Initial investigations into TJ proteins and junctional adhesion molecules (JAM) in cancer suggested a tumor-suppressive role where decreased expression led to increased metastasis. However, recent studies of the JAM family members JAM-A and JAM-C have expanded the roles of these proteins to include protumorigenic functions, including inhibition of apoptosis and promotion of proliferation, cancer stem cell biology, and epithelial-to-mesenchymal transition. JAM function by interacting with other proteins through three distinct molecular mechanisms: direct cell-cell interaction on adjacent cells, stabilization of adjacent cell surface receptors on the same cell, and interactions between JAM and cell surface receptors expressed on adjacent cells. Collectively, these diverse interactions contribute to both the pro- and antitumorigenic functions of JAM. In this review, we discuss these context-dependent functions of JAM in a variety of cancers and highlight key areas that remain poorly understood, including their potentially diverse intracellular signaling networks, their roles in the tumor microenvironment, and the consequences of posttranslational modifications on their function. These studies have implications in furthering our understanding of JAM in cancer and provide a paradigm for exploring additional roles of TJ proteins.
Collapse
Affiliation(s)
- Adam Lauko
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio.,Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Zhaomei Mu
- Cardeza Center for Vascular Biology, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - David H Gutmann
- Washington University School of Medicine, St. Louis, Missouri
| | - Ulhas P Naik
- Cardeza Center for Vascular Biology, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania.
| | - Justin D Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio. .,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio.,Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
21
|
Nagare RP, Sneha S, Krishnapriya S, Ramachandran B, Murhekar K, Vasudevan S, Shabna A, Ganesan TS. ALDH1A1+ ovarian cancer stem cells co-expressing surface markers CD24, EPHA1 and CD9 form tumours in vivo. Exp Cell Res 2020; 392:112009. [PMID: 32305326 DOI: 10.1016/j.yexcr.2020.112009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 04/12/2020] [Indexed: 02/09/2023]
Abstract
One of the reasons for recurrence following treatment of high grade serous ovarian carcinoma (HGSOC) is the persistence of residual cancer stem cells (CSCs). There has been variability between laboratories in the identification of CSC markers for HGSOC. We have identified new surface markers (CD24, CD9 and EPHA1) in addition to those previously known (CD44, CD117 and CD133) using a bioinformatics approach. The expression of these surface markers was evaluated in ovarian cancer cell lines, primary malignant cells (PMCs), normal ovary and HGSOC. There was no preferential expression of any of the markers or a combination. All the markers were expressed at variable levels in ovarian cancer cell lines and PMCs. Only CD117 and CD9 were expressed in the normal ovarian surface epithelium and fallopian tube. Both ALDEFLUOR (ALDH1A1) and side population assays identified a small proportion of cells (<3%) separately that did not overlap with little variability in cell lines and PMCs. All surface markers were co-expressed in ALDH1A1+ cells without preference for one combination. The cell cycle analysis of ALDH1A1+ cells alone revealed that majority of them reside in G0/G1 phase of cell cycle. Further separation of G0 and G1 phases showed that ALDH1A1+ cells reside in G1 phase of the cell cycle. Xenograft assays showed that the combinations of ALDH1A1 + cells co-expressing CD9, CD24 or EPHA1 were more tumorigenic and aggressive with respect to ALDH1A1-cells. These data suggest that a combined approach could be more useful in identifying CSCs in HGSOC.
Collapse
Affiliation(s)
- Rohit P Nagare
- Laboratory for Cancer Biology, Cancer Institute (WIA), Chennai, Tamilnadu, India
| | - Smarakan Sneha
- Laboratory for Cancer Biology, Cancer Institute (WIA), Chennai, Tamilnadu, India
| | - Syama Krishnapriya
- Laboratory for Cancer Biology, Cancer Institute (WIA), Chennai, Tamilnadu, India
| | | | - Kanchan Murhekar
- Department of Pathology, Cancer Institute (WIA), Chennai, Tamilnadu, India
| | - Sekar Vasudevan
- Laboratory for Cancer Biology, Cancer Institute (WIA), Chennai, Tamilnadu, India
| | - Aboo Shabna
- Laboratory for Cancer Biology, Cancer Institute (WIA), Chennai, Tamilnadu, India
| | - Trivadi S Ganesan
- Laboratory for Cancer Biology, Cancer Institute (WIA), Chennai, Tamilnadu, India; Department of Medical Oncology and Clinical Research, Cancer Institute (WIA), Chennai, Tamilnadu, India.
| |
Collapse
|
22
|
Akella NM, Le Minh G, Ciraku L, Mukherjee A, Bacigalupa ZA, Mukhopadhyay D, Sodi VL, Reginato MJ. O-GlcNAc Transferase Regulates Cancer Stem-like Potential of Breast Cancer Cells. Mol Cancer Res 2020; 18:585-598. [PMID: 31974291 PMCID: PMC7127962 DOI: 10.1158/1541-7786.mcr-19-0732] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 12/06/2019] [Accepted: 01/16/2020] [Indexed: 01/01/2023]
Abstract
Breast tumors are heterogeneous and composed of different subpopulation of cells, each with dynamic roles that can change with stage, site, and microenvironment. Cellular heterogeneity is, in part, due to cancer stem-like cells (CSC) that share properties with stem cells and are associated with treatment resistance. CSCs rewire metabolism to meet energy demands of increased growth and biosynthesis. O-GlcNAc transferase enzyme (OGT) uses UDP-GlcNAc as a substrate for adding O-GlcNAc moieties to nuclear and cytoplasmic proteins. OGT/O-GlcNAc levels are elevated in multiple cancers and reducing OGT in cancer cells blocks tumor growth. Here, we report that breast CSCs enriched in mammosphere cultures contain elevated OGT/O-GlcNAcylation. Inhibition of OGT genetically or pharmacologically reduced mammosphere forming efficiency, the CD44H/CD24L, NANOG+, and ALDH+ CSC population in breast cancer cells. Conversely, breast cancer cells overexpressing OGT increased mammosphere formation, CSC populations in vitro, and also increased tumor initiation and CSC frequency in vivo. Furthermore, OGT regulates expression of a number of epithelial-to-mesenchymal transition and CSC markers including CD44, NANOG, and c-Myc. In addition, we identify Krüppel-like factor 8 (KLF8) as a novel regulator of breast cancer mammosphere formation and a critical target of OGT in regulating CSCs. IMPLICATIONS: These findings demonstrate that OGT plays a key role in the regulation of breast CSCs in vitro and tumor initiation in vivo, in part, via regulation of KLF8, and thus inhibition of OGT may serve as a therapeutic strategy to regulate tumor-initiating activity.
Collapse
Affiliation(s)
- Neha M Akella
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Giang Le Minh
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Lorela Ciraku
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Ayonika Mukherjee
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Zachary A Bacigalupa
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Dimpi Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Valerie L Sodi
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Mauricio J Reginato
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania.
| |
Collapse
|
23
|
Ivanova AE, Kravchenko DS, Chumakov SP. A Modified Lentivirus-Based Reporter for Magnetic Separation of Cancer Stem Cells. Mol Biol 2020. [DOI: 10.1134/s0026893319040046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
24
|
Saeednejad Zanjani L, Madjd Z, Rasti A, Asgari M, Abolhasani M, Tam KJ, Roudi R, Mælandsmo GM, Fodstad Ø, Andersson Y. Spheroid-Derived Cells From Renal Adenocarcinoma Have Low Telomerase Activity and High Stem-Like and Invasive Characteristics. Front Oncol 2019; 9:1302. [PMID: 31921617 PMCID: PMC6915099 DOI: 10.3389/fonc.2019.01302] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 11/11/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells (CSCs) are a theorized small subpopulation of cells within tumors thought to be responsible for metastasis, tumor development, disease progression, treatment-resistance, and recurrence. The identification, isolation, and biological characterization of CSCs may therefore facilitate the development of efficient therapeutic strategies targeting CSCs. This study aims to compare the biology and telomerase activity of CSCs to parental cells (PCs) in renal cancer. Renal CSCs were enriched from the ACHN cell line using a sphere culture system. Spheroid-derived cells (SDCs) and their adherent counterparts were compared with respect to their colony and sphere formation, expression of putative CSC markers, tumorigenicity in non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mice, and invasiveness. The expression of genes associated with CSCs, stemness, EMT, apoptosis, and ABC transporters was also compared between the two populations using quantitative real-time PCR (qRT-PCR). Finally, telomerase activity, hTERT expression, and sensitivity to MST-312, a telomerase inhibitor, was investigated between the two populations. We demonstrated that a subpopulation of ACHN cells was capable of growing as spheroids with many properties similar to CSCs, including higher clonogenicity, superior colony- and sphere-forming ability, and stronger tumorigenicity and invasiveness. In addition, SDCs demonstrated a higher expression of markers for CSCs, stemness, EMT, apoptosis, and ABC transporter genes compared to PCs. The expression of hTERT and telomerase activity in SDCs was significantly lower than PCs; however, the SDC population was more sensitive to MST-312 compared to PCs. These findings indicate that the SDC population exhibits stem-like potential and invasive characteristics. Moreover, the reduced expression of hTERT and telomerase activity in SDCs demonstrated that the expressions of hTERT and telomerase activity are not always higher in CSCs. Our results also showed that MST-312 treatment inhibited SDCs more strongly than PCs and may therefore be useful as a complementary targeted therapy against renal CSCs in the future.
Collapse
Affiliation(s)
- Leili Saeednejad Zanjani
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.,Department of Urologic Sciences, Vancouver Prostate Center, University of British Columbia, Vancouver, BC, Canada
| | - Arezoo Rasti
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Basic Sciences/Medical Surgical Nursing, Faculty of Nursing and Midwifery, Tehran University of Medical Sciences, Tehran, Iran
| | - Mojgan Asgari
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Hasheminejad Kidney Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Maryam Abolhasani
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Hasheminejad Kidney Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Kevin J Tam
- Department of Urologic Sciences, Vancouver Prostate Center, University of British Columbia, Vancouver, BC, Canada
| | - Raheleh Roudi
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Gunhild Mari Mælandsmo
- Department of Tumor Biology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Øystein Fodstad
- Department of Tumor Biology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Yvonne Andersson
- Department of Tumor Biology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| |
Collapse
|
25
|
Connor EV, Saygin C, Braley C, Wiechert AC, Karunanithi S, Crean-Tate K, Abdul-Karim FW, Michener CM, Rose PG, Lathia JD, Reizes O. Thy-1 predicts poor prognosis and is associated with self-renewal in ovarian cancer. J Ovarian Res 2019; 12:112. [PMID: 31735168 PMCID: PMC6858973 DOI: 10.1186/s13048-019-0590-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 11/04/2019] [Indexed: 01/06/2023] Open
Abstract
Background Ovarian cancer is the leading cause of gynecologic cancer death in the United States despite effective first-line systemic chemotherapy. Cancer stem cells (CSCs) retain the ability to self-renew and proliferate and may be a means of harboring disease that evades standard treatment strategies. We previously performed a high-throughput screen to assess differential protein expression in ovarian CSCs compared to non-CSCs and observed that Thy-1 was more highly expressed in CSCs. Our primary aim was to validate Thy-1 (CD90) as a cancer stem cell (CSC) marker in epithelial ovarian cancer (EOC), correlate with clinical outcomes, and assess as a potential therapeutic target. Results Kaplan Meier (KM) Plotter data were correlated with survival outcomes. Quantitative real-time PCR, flow cytometry, and immunoblots assessed RNA and protein expression. Limiting dilution assays assessed self-renewal capacity and proliferation assays assessed proliferative capacity. RNA in-situ hybridization was performed on patient specimens to assess feasibility. Thy-1 (CD90) is more highly expressed in ovarian CSCs than non-CSCs, in EOC compared to benign ovarian epithelium (P < 0.001), and is highest in serous EOC (P < 0.05). Serous ovarian cancers with high Thy-1 expression have poorer outcomes (median PFS 15.8 vs. 18.3 months, P = 0 < 0.001; median OS 40.1 v. 45.8 months, P = 0.036). Endometrioid ovarian cancers with high Thy-1 have poorer PFS, but no difference in OS (upper quartile PFS 34 v. 11 months, P = 0.013; quartile OS not reached, P = 0.69). In vitro, Thy-1 expression is higher in CSCs versus non-CSCs. EOC cells with high Thy-1 expression demonstrate increased proliferation and self-renewal. Thy-1 knockdown in EOC cells decreases proliferative capacity and self-renewal capacity, and knockdown is associated with decreased expression of stem cell transcription factors NANOG and SOX2. RNA in situ hybridization is feasible in ovarian cancer tissue specimens. Conclusions Thy-1 is a marker of ovarian CSCs. Increased expression of Thy-1 in EOC predicts poor prognosis and is associated with increased proliferative and self-renewal capacity. Thy-1 knockdown decreases proliferative and self-renewal capacity, and represents a potential therapeutic target.
Collapse
Affiliation(s)
- Elizabeth V Connor
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology and Women's Health Institute, Cleveland Clinic, 801 North 29th Street Billings, MT, Cleveland, OH, 59101, USA. .,Billings Clinic Cancer Center, Division of Gynecologic Oncology, 801 North 29th Street, Billings, MT, 59101, USA.
| | - Caner Saygin
- Cancer Impact Area, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Chad Braley
- Cancer Impact Area, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.,Center of Excellence in Gynecological Cancer Research, Cleveland Clinic, Cleveland, OH, USA
| | - Andrew C Wiechert
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology and Women's Health Institute, Cleveland Clinic, 801 North 29th Street Billings, MT, Cleveland, OH, 59101, USA
| | | | - Katie Crean-Tate
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology and Women's Health Institute, Cleveland Clinic, 801 North 29th Street Billings, MT, Cleveland, OH, 59101, USA
| | | | - Chad M Michener
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology and Women's Health Institute, Cleveland Clinic, 801 North 29th Street Billings, MT, Cleveland, OH, 59101, USA.,Center of Excellence in Gynecological Cancer Research, Cleveland Clinic, Cleveland, OH, USA
| | - Peter G Rose
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology and Women's Health Institute, Cleveland Clinic, 801 North 29th Street Billings, MT, Cleveland, OH, 59101, USA.,Center of Excellence in Gynecological Cancer Research, Cleveland Clinic, Cleveland, OH, USA
| | - Justin D Lathia
- Cancer Impact Area, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.,Center of Excellence in Gynecological Cancer Research, Cleveland Clinic, Cleveland, OH, USA
| | - Ofer Reizes
- Cancer Impact Area, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA. .,Center of Excellence in Gynecological Cancer Research, Cleveland Clinic, Cleveland, OH, USA. .,Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA. .,Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Desk NC10, Cleveland, OH, 44195, USA. .,Case Comprehensive Cancer Center, Cleveland, OH, USA.
| |
Collapse
|
26
|
Vaddi PK, Stamnes MA, Cao H, Chen S. Elimination of SOX2/OCT4-Associated Prostate Cancer Stem Cells Blocks Tumor Development and Enhances Therapeutic Response. Cancers (Basel) 2019; 11:cancers11091331. [PMID: 31500347 PMCID: PMC6769476 DOI: 10.3390/cancers11091331] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/29/2019] [Accepted: 09/04/2019] [Indexed: 12/15/2022] Open
Abstract
SOX2 and OCT4 are key regulators of embryonic stem cell pluripotency. They are overexpressed in prostate cancers and have been associated with cancer stem cell (CSC) properties. However, reliable tools for detecting and targeting SOX2/OCT4-overexpressing cells are lacking, limiting our understanding of their roles in prostate cancer initiation, progression, and therapeutic resistance. Here, we show that a fluorescent reporter called SORE6 can identify SOX2/OCT4-overexpressing prostate cancer cells. Among tumor cells, the SORE6 reporter identified a small fraction with CSC hallmarks: rapid self-renewal, the capability to form tumors and metastasize, and resistance to chemotherapies. Transcriptome and biochemical analyses identified PI3K/AKT signaling as critical for maintaining the SORE6+ population. Moreover, a SORE6-driven herpes simplex virus thymidine kinase (TK) expression construct could selectively ablate SORE6+ cells in tumors, blocking tumor initiation and progression, and sensitizing tumors to chemotherapy. This study demonstrates a key role of SOX2/OCT4-associated prostate cancer stem cells in tumor development and therapeutic resistance, and identifies the SORE6 reporter system as a useful tool for characterizing CSCs functions in a native tumor microenvironment.
Collapse
Affiliation(s)
- Prasanna Kumar Vaddi
- The Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| | - Mark A Stamnes
- The Department of Molecular Physiology and Physics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| | - Huojun Cao
- The Department of Endodontics, College of Dentistry and Dental Clinics, University of Iowa, Iowa City, IA 52242, USA.
| | - Songhai Chen
- The Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
- The Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
- The Holden Comprehensive Cancer Center, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
27
|
Bidan N, Bailleul-Dubois J, Duval J, Winter M, Denoulet M, Hannebicque K, El-Sayed IY, Ginestier C, Forissier V, Charafe-Jauffret E, Macario M, Matsunaga YT, Meignan S, Anquez F, Julien S, Bonnefond A, Derhourhi M, Le Bourhis X, Lagadec C. Transcriptomic Analysis of Breast Cancer Stem Cells and Development of a pALDH1A1:mNeptune Reporter System for Live Tracking. Proteomics 2019; 19:e1800454. [PMID: 31430054 DOI: 10.1002/pmic.201800454] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 08/12/2019] [Indexed: 12/24/2022]
Abstract
Many solid cancers are hierarchically organized with a small number of cancer stem cells (CSCs) able to regrow a tumor, while their progeny lacks this feature. Breast CSC is known to contribute to therapy resistance. The study of those cells is usually based on their cell-surface markers like CD44high /CD24low/neg or their aldehyde dehydrogenase (ALDH) activity. However, these markers cannot be used to track the dynamics of CSC. Here, a transcriptomic analysis is performed to identify segregating gene expression in CSCs and non-CSCs, sorted by Aldefluor assay. It is observed that among ALDH-associated genes, only ALDH1A1 isoform is increased in CSCs. A CSC reporter system is then developed by using a far red-fluorescent protein (mNeptune) under the control of ALDH1A1 promoter. mNeptune-positive cells exhibit higher sphere-forming capacity, tumor formation, and increased resistance to anticancer therapies. These results indicate that the reporter identifies cells with stemness characteristics. Moreover, live tracking of cells in a microfluidic system reveals a higher extravasation potential of CSCs. Live tracking of non-CSCs under irradiation treatment show, for the first time, live reprogramming of non-CSCs into CSCs. Therefore, the reporter will allow for cell tracking to better understand the implication of CSCs in breast cancer development and recurrence.
Collapse
Affiliation(s)
- Nadège Bidan
- University of Lille, U908-CPAC, Cell Plasticity and Cancer, F-59000, Lille, France.,Institut National de la Santé et de la recherche Médicale (INSERM), U908, F-59000, Lille, France
| | - Justine Bailleul-Dubois
- University of Lille, U908-CPAC, Cell Plasticity and Cancer, F-59000, Lille, France.,Institut National de la Santé et de la recherche Médicale (INSERM), U908, F-59000, Lille, France.,Institut pour la Recherche sur le Cancer de Lille (IRCL), 59000, Lille, France
| | - Jérémy Duval
- Institut National de la Santé et de la recherche Médicale (INSERM), U908, F-59000, Lille, France
| | - Marie Winter
- University of Lille, U908-CPAC, Cell Plasticity and Cancer, F-59000, Lille, France.,Institut National de la Santé et de la recherche Médicale (INSERM), U908, F-59000, Lille, France
| | - Marie Denoulet
- University of Lille, U908-CPAC, Cell Plasticity and Cancer, F-59000, Lille, France.,Institut National de la Santé et de la recherche Médicale (INSERM), U908, F-59000, Lille, France
| | - Karine Hannebicque
- Institut National de la Santé et de la recherche Médicale (INSERM), U908, F-59000, Lille, France.,Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, 3 rue Frédéric Combemale, 59000, Lille, France
| | - Ihsan Y El-Sayed
- University of Lille, U908-CPAC, Cell Plasticity and Cancer, F-59000, Lille, France
| | - Christophe Ginestier
- Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la recherche Médicale (INSERM) Paoli-Calmettes Institute, Centre de Recherche en Cancérologie de Marseille (CRCM), Epithelial Stem Cells and Cancer Team, University of Aix-Marseille, 13009, Marseille, France
| | - Violaine Forissier
- Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la recherche Médicale (INSERM) Paoli-Calmettes Institute, Centre de Recherche en Cancérologie de Marseille (CRCM), Epithelial Stem Cells and Cancer Team, University of Aix-Marseille, 13009, Marseille, France
| | - Emmanuelle Charafe-Jauffret
- Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la recherche Médicale (INSERM) Paoli-Calmettes Institute, Centre de Recherche en Cancérologie de Marseille (CRCM), Epithelial Stem Cells and Cancer Team, University of Aix-Marseille, 13009, Marseille, France
| | - Manon Macario
- Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la recherche Médicale (INSERM) Paoli-Calmettes Institute, Centre de Recherche en Cancérologie de Marseille (CRCM), Epithelial Stem Cells and Cancer Team, University of Aix-Marseille, 13009, Marseille, France
| | - Yukiko T Matsunaga
- Center for International Research on Integrative Biomedical Systems (CIBiS), Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan.,LIMMS/CNRS-IIS (UMI 2820), Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan.,Centre National de la Recherche Scientifique (CNRS)/IIS/COL/Lille University SMMiL-E Project, CNRS Délégation Nord-Pas de Calais et Picardie, 2 rue de Canonniers, Lille, Cedex, 59046, France
| | - Samuel Meignan
- Institut National de la Santé et de la recherche Médicale (INSERM), U908, F-59000, Lille, France.,Institut pour la Recherche sur le Cancer de Lille (IRCL), 59000, Lille, France.,Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, 3 rue Frédéric Combemale, 59000, Lille, France
| | - François Anquez
- Laboratory of Physics of Lasers, Atoms and Molecules, UMR CNRS 8523, University of Lille, Villeneuve-d'Ascq, 59655, France
| | - Sylvain Julien
- University of Lille, U908-CPAC, Cell Plasticity and Cancer, F-59000, Lille, France.,Institut National de la Santé et de la recherche Médicale (INSERM), U908, F-59000, Lille, France
| | - Amélie Bonnefond
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, 59000, Lille, France
| | - Mehdi Derhourhi
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, 59000, Lille, France
| | - Xuefen Le Bourhis
- University of Lille, U908-CPAC, Cell Plasticity and Cancer, F-59000, Lille, France.,Institut National de la Santé et de la recherche Médicale (INSERM), U908, F-59000, Lille, France
| | - Chann Lagadec
- University of Lille, U908-CPAC, Cell Plasticity and Cancer, F-59000, Lille, France.,Institut National de la Santé et de la recherche Médicale (INSERM), U908, F-59000, Lille, France.,Institut pour la Recherche sur le Cancer de Lille (IRCL), 59000, Lille, France
| |
Collapse
|
28
|
A novel reporter construct for screening small molecule inhibitors that specifically target self-renewing cancer cells. Exp Cell Res 2019; 383:111551. [PMID: 31401066 DOI: 10.1016/j.yexcr.2019.111551] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 12/11/2022]
Abstract
Cancer stem cells (CSCs) are a subset of cancer cells, which possess self-renewal ability, and lead to tumor progression, metastasis, and resistance to therapy. Live detection and isolation of CSCs are important to understand the biology of CSCs as well as to screen drugs that target them. Even though CSCs are detected using surface markers, there is a lot of inconsistencies for that in a given cancer type. At the same time, self-renewal markers like ALDH1A1, OCT4A and SOX2, which are intracellular molecules, are reliable markers for CSCs in different cancers. In the present study, we generated a reporter construct for self-renewing CSCs, based on ALDH1A1 expression. Oral cancer cells harboring ALDH1A1-DsRed2 were used to screen inhibitors that target CSCs. Our results showed that Comb1, a cocktail of inhibitors for EGF and TGF-β pathways and their intermediates, effectively reduced the DsRed2 population to 34%. Our immunohistochemical analysis on primary oral cancer corroborated the importance of EGF and TGF-β pathways in sustaining CSCs. Since these two pathways are also critical for the self-renewal and differentiation of normal stem cells, Comb1 might abolish them as well. On analysis of the effect of Comb1 on normal murine bone marrow cells, there was no significant change in the stem cell self-renewal and differentiation potential in the treated group compared to untreated cells. To conclude, we claim that ALDH1A1-DsRed2 is a useful tool to detect CSCs, and Comb1 is effective in targeting CSCs without affecting normal stem cells.
Collapse
|
29
|
Beals N, Kasibhatla N, Basu S. Efficient Delivery of Plasmid DNA Using Incorporated Nucleotides for Precise Conjugation of Targeted Nanoparticles. ACS APPLIED BIO MATERIALS 2019; 2:717-727. [DOI: 10.1021/acsabm.8b00596] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Nathan Beals
- Department of Chemistry and Biochemistry, Kent State University, Kent, Ohio 44242, United States
| | - Nithya Kasibhatla
- Department of Chemistry and Biochemistry, Kent State University, Kent, Ohio 44242, United States
| | - Soumitra Basu
- Department of Chemistry and Biochemistry, Kent State University, Kent, Ohio 44242, United States
| |
Collapse
|
30
|
Velasco-Velázquez MA, Velázquez-Quesada I, Vásquez-Bochm LX, Pérez-Tapia SM. Targeting Breast Cancer Stem Cells: A Methodological Perspective. Curr Stem Cell Res Ther 2019; 14:389-397. [PMID: 30147014 DOI: 10.2174/1574888x13666180821155701] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 08/01/2018] [Accepted: 08/09/2018] [Indexed: 02/07/2023]
Abstract
Cancer Stem Cells (CSCs) constitute a subpopulation at the top of the tumor cell hierarchy that contributes to tumor heterogeneity and is uniquely capable of seeding new tumors. Because of their biological properties, CSCs have been pointed out as therapeutic targets for the development of new therapies against breast cancer. The identification of drugs that selectively target breast CSCs requires a clear understanding of their biological functions and the experimental methods to evaluate such hallmarks. Herein, we review the methods to study breast CSCs properties and discuss their value in the preclinical evaluation of CSC-targeting drugs.
Collapse
Affiliation(s)
- Marco A Velasco-Velázquez
- Departamento de Farmacologia, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Ciudad de Mexico, Mexico
- Unidad Periférica de Investigación en Biomedicina Traslacional, Facultad de Medicina, UNAM, Ciudad de México, México
| | - Inés Velázquez-Quesada
- Departamento de Farmacologia, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Ciudad de Mexico, Mexico
- Unidad de Desarrollo e Investigacion en Bioprocesos, ENCB, Instituto Politecnico Nacional, Ciudad de Mexico, Mexico
| | - Luz X Vásquez-Bochm
- Departamento de Farmacologia, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Ciudad de Mexico, Mexico
- Posgrado en Ciencias Químicas, UNAM, Ciudad de México, México
| | - Sonia M Pérez-Tapia
- Unidad de Desarrollo e Investigacion en Bioprocesos, ENCB, Instituto Politecnico Nacional, Ciudad de Mexico, Mexico
| |
Collapse
|
31
|
Vellanki SH, Cruz RGB, Jahns H, Hudson L, Sette G, Eramo A, Hopkins AM. Natural compound Tetrocarcin-A downregulates Junctional Adhesion Molecule-A in conjunction with HER2 and inhibitor of apoptosis proteins and inhibits tumor cell growth. Cancer Lett 2018; 440-441:23-34. [PMID: 30312728 DOI: 10.1016/j.canlet.2018.09.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 09/14/2018] [Accepted: 09/28/2018] [Indexed: 01/12/2023]
Abstract
Overexpression of the tight junction protein Junctional Adhesion Molecule-A (JAM-A) has been linked to aggressive disease in breast and other cancers, but JAM-targeting drugs remain elusive. Screening of a natural compound library identified the antibiotic Tetrocarcin-A as a novel downregulator of JAM-A and human epidermal growth factor receptor-2 (HER2) protein expression in breast cancer cells. Lysosomal inhibition partially rescued the downregulation of JAM-A and HER2 caused by Tetrocarcin-A, and attenuated its cytotoxic activity. Tetrocarcin-A treatment or JAM-A silencing reduced AKT and ERK phosphorylation, inhibited c-FOS phosphorylation at Threonine-232 (its transcriptional regulation site), inhibited nuclear localization of c-FOS, and downregulated expression of the inhibitor of apoptosis proteins (IAP). This was accompanied by Tetrocarcin-A-induced caspase-dependent apoptosis. To begin evaluating the potential clinical relevance of our findings, we extended our studies to other models. Encouragingly, Tetrocarcin-A downregulated JAM-A expression and caused cytotoxicity in primary breast cells and lung cancer stem cells, and inhibited the growth of xenografts in a semi-in vivo model involving invasion across the chicken egg chorioallantoic membrane. Taken together, our data suggest that Tetrocarcin-A warrants future evaluation as a novel cancer therapeutic by virtue of its ability to downregulate JAM-A expression, reduce tumorigenic signaling and induce apoptosis.
Collapse
Affiliation(s)
| | - Rodrigo G B Cruz
- Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Hanne Jahns
- Pathobiology Section, School of Veterinary Medicine, University College Dublin, Ireland
| | - Lance Hudson
- Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Giovanni Sette
- Department of Oncology and Molecular Medicine - Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161, Rome, Italy
| | - Adriana Eramo
- Department of Oncology and Molecular Medicine - Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161, Rome, Italy
| | - Ann M Hopkins
- Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland.
| |
Collapse
|
32
|
Kumar S, Wilkes DW, Samuel N, Blanco MA, Nayak A, Alicea-Torres K, Gluck C, Sinha S, Gabrilovich D, Chakrabarti R. ΔNp63-driven recruitment of myeloid-derived suppressor cells promotes metastasis in triple-negative breast cancer. J Clin Invest 2018; 128:5095-5109. [PMID: 30295647 DOI: 10.1172/jci99673] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 08/28/2018] [Indexed: 12/30/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is particularly aggressive, with enhanced incidence of tumor relapse, resistance to chemotherapy, and metastases. As the mechanistic basis for this aggressive phenotype is unclear, treatment options are limited. Here, we showed an increased population of myeloid-derived immunosuppressor cells (MDSCs) in TNBC patients compared with non-TNBC patients. We found that high levels of the transcription factor ΔNp63 correlate with an increased number of MDSCs in basal TNBC patients, and that ΔNp63 promotes tumor growth, progression, and metastasis in human and mouse TNBC cells. Furthermore, we showed that MDSC recruitment to the primary tumor and metastatic sites occurs via direct ΔNp63-dependent activation of the chemokines CXCL2 and CCL22. CXCR2/CCR4 inhibitors reduced MDSC recruitment, angiogenesis, and metastasis, highlighting a novel treatment option for this subset of TNBC patients. Finally, we found that MDSCs secrete prometastatic factors such as MMP9 and chitinase 3-like 1 to promote TNBC cancer stem cell function, thereby identifying a nonimmunologic role for MDSCs in promoting TNBC progression. These findings identify a unique crosstalk between ΔNp63+ TNBC cells and MDSCs that promotes tumor progression and metastasis, which could be exploited in future combined immunotherapy/chemotherapy strategies for TNBC patients.
Collapse
Affiliation(s)
- Sushil Kumar
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David W Wilkes
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nina Samuel
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mario Andres Blanco
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anupma Nayak
- Department of Pathology and Laboratory Medicine at the Hospital of the University of Pennsylvania, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Christian Gluck
- Department of Biochemistry, State University of New York, Buffalo, New York, USA
| | - Satrajit Sinha
- Department of Biochemistry, State University of New York, Buffalo, New York, USA
| | | | - Rumela Chakrabarti
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
33
|
Skvortsov S, Skvortsova II, Tang DG, Dubrovska A. Concise Review: Prostate Cancer Stem Cells: Current Understanding. Stem Cells 2018; 36:1457-1474. [PMID: 29845679 DOI: 10.1002/stem.2859] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 04/05/2018] [Accepted: 05/01/2018] [Indexed: 12/29/2022]
Abstract
Prostate cancer (PCa) is heterogeneous, harboring phenotypically diverse cancer cell types. PCa cell heterogeneity is caused by genomic instability that leads to the clonal competition and evolution of the cancer genome and by epigenetic mechanisms that result in subclonal cellular differentiation. The process of tumor cell differentiation is initiated from a population of prostate cancer stem cells (PCSCs) that possess many phenotypic and functional properties of normal stem cells. Since the initial reports on PCSCs in 2005, there has been much effort to elucidate their biological properties, including unique metabolic characteristics. In this Review, we discuss the current methods for PCSC enrichment and analysis, the hallmarks of PCSC metabolism, and the role of PCSCs in tumor progression. Stem Cells 2018;36:1457-1474.
Collapse
Affiliation(s)
- Sergej Skvortsov
- Laboratory for Experimental and Translational Research on Radiation Oncology (EXTRO-Lab), Department of Therapeutic Radiology and Oncology, Innsbruck Medical University, Innsbruck, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - Ira-Ida Skvortsova
- Laboratory for Experimental and Translational Research on Radiation Oncology (EXTRO-Lab), Department of Therapeutic Radiology and Oncology, Innsbruck Medical University, Innsbruck, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York, USA.,Cancer Stem Cell Institute, Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Anna Dubrovska
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
34
|
O'Conor CJ, Chen T, González I, Cao D, Peng Y. Cancer stem cells in triple-negative breast cancer: a potential target and prognostic marker. Biomark Med 2018; 12:813-820. [PMID: 29902924 DOI: 10.2217/bmm-2017-0398] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 02/27/2018] [Indexed: 12/18/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive disease with poor outcome, and lacks targeted therapy. Recent studies suggest that cancer stem cells play an important role in tumorigenesis and tumor biology of TNBC. Both CD44+/CD24- and ALDH1+ breast cancer stem cells are enriched in TNBC and may contribute to the propensity of TNBC for chemotherapy resistance and tumor metastasis. There is new evidence to support the evaluation of cancer stem cells in TNBC for diagnostic purposes. Targeting cancer stem cells may also be a promising, novel strategy for the treatment of TNBC. This review highlights the current understanding of breast cancer stem cells in TNBC, with focus on CD44+/CD24- and ALDH1+ breast cancer stem cells. It is our hope that this work provides insight into the potential role of cancer stem cells in the prognostication and therapeutic targeting of TNBC.
Collapse
Affiliation(s)
- Christopher J O'Conor
- Department of Pathology & Immunology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Tiffany Chen
- Department of Pathology & Immunology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Iván González
- Department of Pathology & Immunology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Dengfeng Cao
- Department of Pathology & Immunology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Yan Peng
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
35
|
Cheng CC, Shi LH, Wang XJ, Wang SX, Wan XQ, Liu SR, Wang YF, Lu Z, Wang LH, Ding Y. Stat3/Oct-4/c-Myc signal circuit for regulating stemness-mediated doxorubicin resistance of triple-negative breast cancer cells and inhibitory effects of WP1066. Int J Oncol 2018; 53:339-348. [PMID: 29750424 DOI: 10.3892/ijo.2018.4399] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 04/11/2018] [Indexed: 01/06/2023] Open
Abstract
Doxorubicin (Dox) is widely used in the treatment of triple-negative breast cancer cells (TNBCs), however resistance limits its effectiveness. Cancer stem cells (CSCs) are associated with Dox resistance in MCF-7 estrogen receptor positive breast cancer cells. Signal transducer and activator of transcription 3 (Stat3) may functionally shift non-CSCs towards CSCs. However, whether Stat3 drives the formation of CSCs during the development of resistance in TNBC, and whether a Stat3 inhibitor reverses CSC-mediated Dox resistance, remains to be elucidated. In the present study, human MDA-MB-468 and murine 4T1 mammary carcinoma cell lines with the typical characteristics of TNBCs, were compared with estrogen receptor-positive MCF-7 cells as a model system. The MTT assay was used to detect cytotoxicity of Dox. In addition, the expression levels of CSC-specific markers and transcriptional factors were measured by western blotting, immunofluorescence staining and flow cytometry. The mammosphere formation assay was used to detect stem cell activity. Under long-term continuous treatment with Dox at a low concentration, TNBC cultures not only exhibited a drug-resistant phenotype, but also showed CSC properties. These Dox-resistant TNBC cells showed activation of Stat3 and high expression levels of pluripotency transcription factors octamer-binding transcription factor-4 (Oct-4) and c-Myc, which was different from the high expression of superoxide dismutase 2 (Sox2) in Dox-resistant MCF-7 cells. WP1066 inhibited the phosphorylation of Stat3, and decreased the expression of Oct-4 and c-Myc, leading to a reduction in the CD44-positive cell population, and restoring the sensitivity of the cells to Dox. Taken together, a novel signal circuit of Stat3/Oct-4/c-Myc was identified for regulating stemness-mediated Dox resistance in TNBC. The Stat3 inhibitor WP1066 was able to overcome the resistance to Dox through decreasing the enrichment of CSCs, highlighting the therapeutic potential of WP1066 as a novel sensitizer of Dox-resistant TNBC.
Collapse
Affiliation(s)
- Cong-Cong Cheng
- Laboratory of Molecular Oncology, Weifang Medical College, Weifang, Shandong 261053, P.R. China
| | - Li-Hong Shi
- Laboratory of Molecular Oncology, Weifang Medical College, Weifang, Shandong 261053, P.R. China
| | - Xue-Jian Wang
- Laboratory of Molecular Oncology, Weifang Medical College, Weifang, Shandong 261053, P.R. China
| | - Shu-Xiao Wang
- Department of Pharmacology, Weifang Medical College, Weifang, Shandong 261053, P.R. China
| | - Xiao-Qing Wan
- Laboratory of Molecular Oncology, Weifang Medical College, Weifang, Shandong 261053, P.R. China
| | - Shu-Rong Liu
- Laboratory of Molecular Oncology, Weifang Medical College, Weifang, Shandong 261053, P.R. China
| | - Yi-Fei Wang
- Laboratory of Molecular Oncology, Weifang Medical College, Weifang, Shandong 261053, P.R. China
| | - Zhong Lu
- Laboratory of Molecular Oncology, Weifang Medical College, Weifang, Shandong 261053, P.R. China
| | - Li-Hua Wang
- Laboratory of Molecular Oncology, Weifang Medical College, Weifang, Shandong 261053, P.R. China
| | - Yi Ding
- Laboratory of Molecular Oncology, Weifang Medical College, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
36
|
Beals N, Model MA, Worden M, Hegmann T, Basu S. Intermolecular G-Quadruplex Induces Hyaluronic Acid-DNA Superpolymers Causing Cancer Cell Swelling, Blebbing, and Death. ACS APPLIED MATERIALS & INTERFACES 2018; 10:6869-6878. [PMID: 29400433 DOI: 10.1021/acsami.7b16983] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Over the past decade, nanomedicine has gained considerable attraction through its relevance, for example, in "smart" delivery, thus creating platforms for novel treatments. Here, we report a natural polymer-DNA conjugate that undergoes self-assembly in a K+-dependent fashion to form a G-quadruplex (GQ) and generate superpolymeric structures. We derivatized a thiolated conjugate of the naturally occurring glycosaminoglycan polymer hyaluronic acid (HASH) with short G-rich DNA (HASH-DNA) that can form an intermolecular noncanonical GQ structure. Gel mobility shift assay and circular dichroism measurements confirmed HASH conjugation to DNA and K+-dependent GQ formation, respectively. Transmission electron microscopy and scanning electron microscopy results indicated that the addition of K+ to the HASH-DNA conjugate led to the formation of micron-range structures, whereas control samples remained unordered and as a nebulous globular form. Confocal microscopy of a fluorescently labeled form of the superpolymer verified increased cellular uptake. The HASH-DNA conjugates showed toxicity in HeLa cells, whereas a scrambled DNA (Mut) conjugate HASH-Mut showed no cytotoxicity, presumably because of nonformation of the superpolymeric structure. To understand the mechanism of cell death and if the superpolymeric structure is responsible for it, we monitored the cell size and observed an average of 23% increase in size compared to 4.5% in control cells at 4.5 h. We believe that cellular stress is generated presumably by the intracellular assembly of this large superpolymeric nanostructure causing cell blebbing with no exit option. This approach provides a new strategy of cellular delivery of a targeted naturally occurring polymer and a novel way to induce superpolymeric structure formation that acts as a therapeutic.
Collapse
Affiliation(s)
- Nathan Beals
- Department of Chemistry and Biochemistry, ‡Department of Biological Sciences, and §Liquid Crystals Institute, Kent State University , Kent, Ohio 44242, United States
| | - Michael A Model
- Department of Chemistry and Biochemistry, ‡Department of Biological Sciences, and §Liquid Crystals Institute, Kent State University , Kent, Ohio 44242, United States
| | - Matt Worden
- Department of Chemistry and Biochemistry, ‡Department of Biological Sciences, and §Liquid Crystals Institute, Kent State University , Kent, Ohio 44242, United States
| | - Torsten Hegmann
- Department of Chemistry and Biochemistry, ‡Department of Biological Sciences, and §Liquid Crystals Institute, Kent State University , Kent, Ohio 44242, United States
| | - Soumitra Basu
- Department of Chemistry and Biochemistry, ‡Department of Biological Sciences, and §Liquid Crystals Institute, Kent State University , Kent, Ohio 44242, United States
| |
Collapse
|
37
|
Blum W, Henzi T, Schwaller B, Pecze L. Biological noise and positional effects influence cell stemness. J Biol Chem 2018; 293:5247-5258. [PMID: 29440274 DOI: 10.1074/jbc.ra117.001643] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 02/12/2018] [Indexed: 01/01/2023] Open
Abstract
Biological (or cellular) noise is the random quantitative variability of proteins and other molecules in individual, genetically identical cells. As the result of biological noise in the levels of some transcription factors that determine a cell's differentiation status, differentiated cells may dedifferentiate to a stem cell state given a sufficiently long time period. Here, to provide direct evidence supporting this hypothesis, we used a live-cell monitoring system based on enhanced green fluorescent protein (eGFP) expression to continuously assess the "stemness" of individual human and murine malignant mesothelioma cells over a period of up to 3 months. Re-expression of the transcription factors, the top hierarchical stemness markers Sox2 (SRY-box 2) and Oct4 (octamer-binding transcription factor), monitored as cell eGFP expression was observed in a subpopulation of differentiated eGFP(-) malignant mesothelioma cells. However, we found that this transition was extremely rare. Of note, when it did occur, neighboring cells that were not direct descendants of a newly emerged eGFP(+) stem cell were more likely than non-neighboring cells to also become an eGFP(+) stem cell. This observation suggested a positional effect and led to a clustered "mosaic" reappearance of eGFP(+) stem cells. Moreover, stem cells reappeared even in cell cultures derived from one single differentiated eGFP(-) cell. On the basis of our experimental in vitro and in vivo findings, we developed a tumor growth model to predict the clustered localization of cancer stem cells within a tumor mass.
Collapse
Affiliation(s)
- Walter Blum
- From the Unit of Anatomy, Section of Medicine, University of Fribourg, 1700 Fribourg, Switzerland
| | - Thomas Henzi
- From the Unit of Anatomy, Section of Medicine, University of Fribourg, 1700 Fribourg, Switzerland
| | - Beat Schwaller
- From the Unit of Anatomy, Section of Medicine, University of Fribourg, 1700 Fribourg, Switzerland
| | - László Pecze
- From the Unit of Anatomy, Section of Medicine, University of Fribourg, 1700 Fribourg, Switzerland
| |
Collapse
|
38
|
Thiagarajan PS, Sinyuk M, Turaga SM, Mulkearns-Hubert EE, Hale JS, Rao V, Demelash A, Saygin C, China A, Alban TJ, Hitomi M, Torre-Healy LA, Alvarado AG, Jarrar A, Wiechert A, Adorno-Cruz V, Fox PL, Calhoun BC, Guan JL, Liu H, Reizes O, Lathia JD. Cx26 drives self-renewal in triple-negative breast cancer via interaction with NANOG and focal adhesion kinase. Nat Commun 2018; 9:578. [PMID: 29422613 PMCID: PMC5805730 DOI: 10.1038/s41467-018-02938-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 01/09/2018] [Indexed: 12/18/2022] Open
Abstract
Tumors adapt their phenotypes during growth and in response to therapies through dynamic changes in cellular processes. Connexin proteins enable such dynamic changes during development, and their dysregulation leads to disease states. The gap junction communication channels formed by connexins have been reported to exhibit tumor-suppressive functions, including in triple-negative breast cancer (TNBC). However, we find that connexin 26 (Cx26) is elevated in self-renewing cancer stem cells (CSCs) and is necessary and sufficient for their maintenance. Cx26 promotes CSC self-renewal by forming a signaling complex with the pluripotency transcription factor NANOG and focal adhesion kinase (FAK), resulting in NANOG stabilization and FAK activation. This FAK/NANOG-containing complex is not formed in mammary epithelial or luminal breast cancer cells. These findings challenge the paradigm that connexins are tumor suppressors in TNBC and reveal a unique function for Cx26 in regulating the core self-renewal signaling that controls CSC maintenance. Connexin proteins are usually considered as tumor suppressors. Here, the authors show that connexin 26 (Cx26) regulates the self-renewal of breast cancer stem cells via a ternary complex with FAK and NANOG.
Collapse
Affiliation(s)
- Praveena S Thiagarajan
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44915, USA.,Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Maksim Sinyuk
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44915, USA
| | - Soumya M Turaga
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44915, USA
| | - Erin E Mulkearns-Hubert
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44915, USA
| | - James S Hale
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44915, USA
| | - Vinay Rao
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44915, USA
| | - Abeba Demelash
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44915, USA
| | - Caner Saygin
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44915, USA
| | - Arnab China
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44915, USA
| | - Tyler J Alban
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44915, USA.,Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Masahiro Hitomi
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44915, USA.,Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Luke A Torre-Healy
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44915, USA
| | - Alvaro G Alvarado
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44915, USA
| | - Awad Jarrar
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44915, USA
| | - Andrew Wiechert
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44915, USA
| | - Valery Adorno-Cruz
- Case Comprehensive Cancer Center, Cleveland, OH, 44195, USA.,Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.,Departments of Pharmacology and Medicine, Northwestern University School of Medicine, Chicago, IL, 60611, USA
| | - Paul L Fox
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44915, USA.,Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA.,Case Comprehensive Cancer Center, Cleveland, OH, 44195, USA
| | | | - Jun-Lin Guan
- Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Huiping Liu
- Case Comprehensive Cancer Center, Cleveland, OH, 44195, USA.,Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.,Departments of Pharmacology and Medicine, Northwestern University School of Medicine, Chicago, IL, 60611, USA
| | - Ofer Reizes
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44915, USA. .,Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA. .,Case Comprehensive Cancer Center, Cleveland, OH, 44195, USA.
| | - Justin D Lathia
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44915, USA. .,Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA. .,Case Comprehensive Cancer Center, Cleveland, OH, 44195, USA.
| |
Collapse
|
39
|
Increased cancer stem cell invasion is mediated by myosin IIB and nuclear translocation. Oncotarget 2018; 7:47586-47592. [PMID: 27285763 PMCID: PMC5216963 DOI: 10.18632/oncotarget.9896] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 05/01/2016] [Indexed: 01/21/2023] Open
Abstract
Despite many advances in the treatment of breast cancer, it remains one of the leading causes of death among women. One hurdle for effective therapy is the treatment of the highly invasive and tumorigenic subpopulation of tumors called cancer stem cells (CSCs). CSCs, when stimulated with EGF, migrate through a physiological 3D collagen matrix at a higher velocity than non-stem cancer cells (non-SCCs). This increased invasion is due, in part, by an enhanced nuclear translocation ability of CSCs. We observed no difference between CSC and non-SCC in cellular migration rates on a 2D surface. Furthermore, during transwell migration using large diameter transwell pores, both CSC and non-SCC populations migrated with similar efficiency. However, when challenged with more restrictive transwells, CSCs were dramatically more capable of transwell migration. These results implicate nuclear translocation as a major rate limiting factor for CSC dissemination. We further show that non-muscle myosin IIB is critical for this enhanced nuclear translocation and the ability for cancer stem cells to efficiently migrate through restrictive 3D environments. These studies suggest that cytoskeletal elements upregulated in CSCs, such as myosin IIB, may be valuable targets for intervention in cancer stem cell dispersal from tumors.
Collapse
|
40
|
Saygin C, Samour M, Chumakova A, Jarrar A, Lathia JD, Reizes O. Reporter Systems to Study Cancer Stem Cells. Methods Mol Biol 2018; 1516:319-333. [PMID: 27221339 DOI: 10.1007/7651_2016_360] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cancer stem cells have been identified in primary tumors, patient derived xenografts, and established cancer cell lines. The development of reporters has enabled investigators to rapidly enrich for these cells and more importantly track these cells in real time. Here we describe the current state of the reporter field and their use and limitations in multiple cancers.
Collapse
Affiliation(s)
- Caner Saygin
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NC 10, Cleveland, OH, 44195, USA
| | - Mohamed Samour
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NC 10, Cleveland, OH, 44195, USA.,Medicine Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Anastasia Chumakova
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NC 10, Cleveland, OH, 44195, USA
| | - Awad Jarrar
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NC 10, Cleveland, OH, 44195, USA
| | - Justin D Lathia
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NC 10, Cleveland, OH, 44195, USA.,Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH, 44195, USA.,Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA
| | - Ofer Reizes
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NC 10, Cleveland, OH, 44195, USA. .,Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA. .,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH, 44195, USA. .,Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA.
| |
Collapse
|
41
|
Wiechert A, Saygin C, Thiagarajan PS, Rao VS, Hale JS, Gupta N, Hitomi M, Nagaraj AB, DiFeo A, Lathia JD, Reizes O. Cisplatin induces stemness in ovarian cancer. Oncotarget 2017; 7:30511-22. [PMID: 27105520 PMCID: PMC5058697 DOI: 10.18632/oncotarget.8852] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/31/2016] [Indexed: 12/11/2022] Open
Abstract
The mainstay of treatment for ovarian cancer is platinum-based cytotoxic chemotherapy. However, therapeutic resistance and recurrence is a common eventuality for nearly all ovarian cancer patients, resulting in poor median survival. Recurrence is postulated to be driven by a population of self-renewing, therapeutically resistant cancer stem cells (CSCs). A current limitation in CSC studies is the inability to interrogate their dynamic changes in real time. Here we utilized a GFP reporter driven by the NANOG-promoter to enrich and track ovarian CSCs. Using this approach, we identified a population of cells with CSC properties including enhanced expression of stem cell transcription factors, self-renewal, and tumor initiation. We also observed elevations in CSC properties in cisplatin-resistant ovarian cancer cells as compared to cisplatin-naïve ovarian cancer cells. CD49f, a marker for CSCs in other solid tumors, enriched CSCs in cisplatin-resistant and -naïve cells. NANOG-GFP enriched CSCs (GFP+ cells) were more resistant to cisplatin as compared to GFP-negative cells. Moreover, upon cisplatin treatment, the GFP signal intensity and NANOG expression increased in GFP-negative cells, indicating that cisplatin was able to induce the CSC state. Taken together, we describe a reporter-based strategy that allows for determination of the CSC state in real time and can be used to detect the induction of the CSC state upon cisplatin treatment. As cisplatin may provide an inductive stress for the stem cell state, future efforts should focus on combining cytotoxic chemotherapy with a CSC targeted therapy for greater clinical utility.
Collapse
Affiliation(s)
- Andrew Wiechert
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Division of Gynecological Oncology, Women's Health Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Caner Saygin
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Praveena S Thiagarajan
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Vinay S Rao
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - James S Hale
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Nikhil Gupta
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
| | - Masahiro Hitomi
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH 44195, USA
| | - Anil Belur Nagaraj
- Department of Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Analisa DiFeo
- Department of Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Justin D Lathia
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH 44195, USA.,Department of Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Ofer Reizes
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH 44195, USA.,Department of Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| |
Collapse
|
42
|
Beals N, Thiagarajan PS, Soehnlen E, Das A, Reizes O, Lathia JD, Basu S. Five-Part Pentameric Nanocomplex Shows Improved Efficacy of Doxorubicin in CD44+ Cancer Cells. ACS OMEGA 2017; 2:7702-7713. [PMID: 30023561 PMCID: PMC6044869 DOI: 10.1021/acsomega.7b01168] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 10/27/2017] [Indexed: 06/08/2023]
Abstract
The CD44 receptor is common among many cancer types where overexpression is synonymous with poor prognosis in prostate, glioma, and breast cancer. More notably CD44 overexpression has been shown in a number of different cancer stem cells (CSC) which are present in many solid tumors and drive growth, recurrence, and resistance to conventional therapies. Triple negative breast cancer CSCs correlate to worse prognosis and early relapse due to higher drug resistance and increased tumor heterogeneity and thus are prime targets for anticancer therapy. To specifically target cells overexpressing CD44 receptors, including CSCs, we synthesized a pentameric nanocomplex (PNC) containing gold nanoparticles, doxorubicin (Dox) conjugated to thiolated hyaluronic acid via an acid-labile hydrazone bond, and thiolated poly(ethylene glycol) DNA CD44 aptamer. In vitro drug release was highest at 8 h time point at acidic pH (pH 4.7) and in 10 mM glutathione. The PNC is almost an order of magnitude more effective than Dox alone in CD44+ cells versus CD44 low cells. Functionally, the PNC reduced CSC self-renewal. The PNC provides a therapeutic strategy that can improve the efficiency of Dox and decrease nontargeted toxicity thereby prolonging its use to individual patients.
Collapse
Affiliation(s)
- Nathan Beals
- Department
of Chemistry and Biochemistry, Kent State
University, Kent, Ohio 44242, United
States
| | - Praveena S. Thiagarajan
- Department
of Cellular and Molecular Medicine, Lerner
Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| | - Eric Soehnlen
- Department
of Chemistry and Biochemistry, Kent State
University, Kent, Ohio 44242, United
States
| | - Arijit Das
- Department
of Chemistry and Biochemistry, Kent State
University, Kent, Ohio 44242, United
States
| | - Ofer Reizes
- Department
of Cellular and Molecular Medicine, Lerner
Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
- Department
of Molecular Medicine, Cleveland Clinic
Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio 44195, United States
- Case
Comprehensive Cancer Center, Cleveland, Ohio 44106-5065, United States
| | - Justin D. Lathia
- Department
of Cellular and Molecular Medicine, Lerner
Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
- Department
of Molecular Medicine, Cleveland Clinic
Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio 44195, United States
- Case
Comprehensive Cancer Center, Cleveland, Ohio 44106-5065, United States
| | - Soumitra Basu
- Department
of Chemistry and Biochemistry, Kent State
University, Kent, Ohio 44242, United
States
| |
Collapse
|
43
|
Sneha S, Nagare RP, Priya SK, Sidhanth C, Pors K, Ganesan TS. Therapeutic antibodies against cancer stem cells: a promising approach. Cancer Immunol Immunother 2017; 66:1383-1398. [PMID: 28840297 PMCID: PMC11028654 DOI: 10.1007/s00262-017-2049-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 08/03/2017] [Indexed: 12/18/2022]
Abstract
Monoclonal antibodies have been extensively used to treat malignancy along with routine chemotherapeutic drugs. Chemotherapy for metastatic cancer has not been successful in securing long-term remission of disease. This is in part due to the resistance of cancer cells to drugs. One aspect of the drug resistance is the inability of conventional drugs to eliminate cancer stem cells (CSCs) which often constitute less than 1-2% of the whole tumor. In some tumor types, it is possible to identify these cells using surface markers. Monoclonal antibodies targeting these CSCs are an attractive option for a new therapeutic approach. Although administering antibodies has not been effective, when combined with chemotherapy they have proved synergistic. This review highlights the potential of improving treatment efficacy using functional antibodies against CSCs, which could be combined with chemotherapy in the future.
Collapse
Affiliation(s)
- Smarakan Sneha
- Laboratory for Cancer Biology, Department of Medical Oncology and Clinical Research, Cancer Institute (WIA), 38, Sardar Patel Road, Chennai, Tamil Nadu, 600 036, India
| | - Rohit Pravin Nagare
- Laboratory for Cancer Biology, Department of Medical Oncology and Clinical Research, Cancer Institute (WIA), 38, Sardar Patel Road, Chennai, Tamil Nadu, 600 036, India
| | - Syama Krishna Priya
- Laboratory for Cancer Biology, Department of Medical Oncology and Clinical Research, Cancer Institute (WIA), 38, Sardar Patel Road, Chennai, Tamil Nadu, 600 036, India
| | - Chirukandath Sidhanth
- Laboratory for Cancer Biology, Department of Medical Oncology and Clinical Research, Cancer Institute (WIA), 38, Sardar Patel Road, Chennai, Tamil Nadu, 600 036, India
| | - Klaus Pors
- Institute of Cancer Therapeutics, University of Bradford, Bradford, BD7 1DP, UK
| | - Trivadi Sundaram Ganesan
- Laboratory for Cancer Biology, Department of Medical Oncology and Clinical Research, Cancer Institute (WIA), 38, Sardar Patel Road, Chennai, Tamil Nadu, 600 036, India.
| |
Collapse
|
44
|
Rosager AM, Sørensen MD, Dahlrot RH, Boldt HB, Hansen S, Lathia JD, Kristensen BW. Expression and prognostic value of JAM-A in gliomas. J Neurooncol 2017; 135:107-117. [PMID: 28677106 PMCID: PMC5658466 DOI: 10.1007/s11060-017-2555-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 06/27/2017] [Indexed: 12/26/2022]
Abstract
Gliomas are among the most lethal cancers, being highly resistant to both chemo- and radiotherapy. The expression of junctional adhesion molecule-A (JAM-A) was recently identified on the surface of stem cell-like brain tumor-initiating cells and suggested to function as a unique glioblastoma niche adhesion factor influencing the tumorigenic potential of brain tumor-initiating cells. We have recently identified high JAM-A expression to be associated with poor outcome in glioblastomas, and our aim was to further investigate the expression of JAM-A in gliomas focusing especially on the prognostic value in WHO grade II and III gliomas. JAM-A protein expression was evaluated by immunohistochemistry and advanced quantitative image analysis with continuous estimates of staining intensity. The JAM-A antibody stained tumor cell membranes and cytoplasm to various extent in different glioma subtypes, and the intensity was higher in glioblastomas than low-grade gliomas. We could not detect an association with overall survival in patients with grade II and III tumors. Double-immunofluorescence stainings in glioblastomas revealed co-expression of JAM-A with CD133, SOX2, nestin, and GFAP in tumor cells as well as some co-expression with the microglial/macrophage marker IBA-1. In conclusion, JAM-A expression was higher in glioblastomas compared to low-grade gliomas and co-localized with recognized stem cell markers suggesting an association of JAM-A with glioma aggressiveness. No significant association between JAM-A expression and overall survival was found in grade II and III gliomas. Further research is needed to determine the function and clinical impact of JAM-A in gliomas.
Collapse
Affiliation(s)
- Ann Mari Rosager
- Department of Pathology, Odense University Hospital, Winsløwparken 15, 3rd floor, 5000, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Winsløwparken 19, 3rd floor, 5000, Odense, Denmark
| | - Mia D Sørensen
- Department of Pathology, Odense University Hospital, Winsløwparken 15, 3rd floor, 5000, Odense, Denmark.
- Department of Clinical Research, University of Southern Denmark, Winsløwparken 19, 3rd floor, 5000, Odense, Denmark.
| | - Rikke H Dahlrot
- Department of Oncology, Odense University Hospital, Sdr. Boulevard 29, 5000, Odense, Denmark
| | - Henning B Boldt
- Department of Pathology, Odense University Hospital, Winsløwparken 15, 3rd floor, 5000, Odense, Denmark
| | - Steinbjørn Hansen
- Department of Clinical Research, University of Southern Denmark, Winsløwparken 19, 3rd floor, 5000, Odense, Denmark
- Department of Oncology, Odense University Hospital, Sdr. Boulevard 29, 5000, Odense, Denmark
| | - Justin D Lathia
- Department of Cellular and Molecular Medicine, Lerner Research Institute, 9500 Euclid Avenue, NC10, Cleveland, OH, 44195, USA
| | - Bjarne W Kristensen
- Department of Pathology, Odense University Hospital, Winsløwparken 15, 3rd floor, 5000, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Winsløwparken 19, 3rd floor, 5000, Odense, Denmark
| |
Collapse
|
45
|
Maity S, Sadlowski CM, George Lin JM, Chen CH, Peng LH, Lee ES, Vegesna GK, Lee C, Kim SH, Mochly-Rosen D, Kumar S, Murthy N. Thiophene bridged aldehydes (TBAs) image ALDH activity in cells via modulation of intramolecular charge transfer. Chem Sci 2017; 8:7143-7151. [PMID: 29081945 PMCID: PMC5635522 DOI: 10.1039/c7sc03017g] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 08/01/2017] [Indexed: 12/15/2022] Open
Abstract
Aldehyde dehydrogenases (ALDHs) catalyze the oxidation of an aldehyde to a carboxylic acid and are implicated in the etiology of numerous diseases. However, despite their importance, imaging ALDH activity in cells is challenging due to a lack of fluorescent imaging probes. In this report, we present a new family of fluorescent probes composed of an oligothiophene flanked by an aldehyde and an electron donor, termed thiophene-bridged aldehydes (TBAs), which can image ALDH activity in cells. The TBAs image ALDH activity via a fluorescence sensing mechanism based on the modulation of intramolecular charge transfer (ICT) and this enables the TBAs and their ALDH-mediated oxidized products, thiophene-bridged carboxylates (TBCs), to have distinguishable fluorescence spectra. Herein, we show that the TBAs can image ALDH activity in cells via fluorescence microscopy, flow cytometry, and in a plate reader. Using TBA we were able to develop a cell-based high throughput assay for ALDH inhibitors, for the first time, and screened a large, 1460-entry electrophile library against A549 cells. We identified α,β-substituted acrylamides as potent electrophile fragments that can inhibit ALDH activity in cells. These inhibitors sensitized drug-resistant glioblastoma cells to the FDA approved anti-cancer drug, temozolomide. The TBAs have the potential to make the analysis of ALDH activity in cells routinely possible given their ability to spectrally distinguish between an aldehyde and a carboxylic acid.
Collapse
Affiliation(s)
- Santanu Maity
- Department of Bioengineering , University of California , 140 Hearst Memorial Mining Building , Berkeley , CA 94720 , USA .
| | - Corinne M Sadlowski
- Department of Bioengineering , University of California , 140 Hearst Memorial Mining Building , Berkeley , CA 94720 , USA .
| | - Jung-Ming George Lin
- Department of Bioengineering , University of California , 140 Hearst Memorial Mining Building , Berkeley , CA 94720 , USA .
- The UC Berkeley-UCSF Graduate Program in Bioengineering , UC Berkeley , Berkeley , California , USA
| | - Che-Hong Chen
- Department of Chemical and Systems Biology , Stanford University , School of Medicine , Stanford , CA 94305-5174 , USA
| | - Li-Hua Peng
- Department of Bioengineering , University of California , 140 Hearst Memorial Mining Building , Berkeley , CA 94720 , USA .
| | - Eun-Soo Lee
- Korea Research Institute of Standards and Science , 267 Gajeong-ro, Yuseong-gu , Daejeon , Republic of Korea
| | - Giri K Vegesna
- Department of Bioengineering , University of California , 140 Hearst Memorial Mining Building , Berkeley , CA 94720 , USA .
| | - Charles Lee
- Department of Bioengineering , University of California , 140 Hearst Memorial Mining Building , Berkeley , CA 94720 , USA .
| | - Se-Hwa Kim
- Korea Research Institute of Standards and Science , 267 Gajeong-ro, Yuseong-gu , Daejeon , Republic of Korea
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology , Stanford University , School of Medicine , Stanford , CA 94305-5174 , USA
| | - Sanjay Kumar
- Department of Bioengineering , University of California , 140 Hearst Memorial Mining Building , Berkeley , CA 94720 , USA .
- The UC Berkeley-UCSF Graduate Program in Bioengineering , UC Berkeley , Berkeley , California , USA
| | - Niren Murthy
- Department of Bioengineering , University of California , 140 Hearst Memorial Mining Building , Berkeley , CA 94720 , USA .
- The UC Berkeley-UCSF Graduate Program in Bioengineering , UC Berkeley , Berkeley , California , USA
| |
Collapse
|
46
|
Park JW, Jung KH, Lee JH, Moon SH, Cho YS, Choe YS, Lee KH. Imaging Early Fate of Cancer Stem Cells in Mouse Hindlimbs with Sodium Iodide Symporter Gene and I-124 PET. Mol Imaging Biol 2017; 18:748-57. [PMID: 26914278 DOI: 10.1007/s11307-016-0941-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE We investigated the capacity of sodium/iodide symporter (NIS) positron emission tomography (PET) to image and quantitate early engraftment and survival of cancer stem cells (CSCs) in living mice. PROCEDURES CT26 colon cancer cells and CSCs were infected with an adenovirus expressing both NIS and enhanced green fluorescent protein (EGFP). Cells were implanted into normal and ischemic hindlimbs of mice, and serial optical and I-124 PET imaging was performed. Extracted tissues underwent I-124 measurements and confocal microscopy. RESULTS NIS.EGFP gene transfer increased fluorescence and I-124 uptake of CSCs and CT26 cells without adverse effects. I-124 PET clearly visualized implanted tumor cells in vivo, whereas optical imaging was suboptimal. PET revealed 1.95, 2.22, and 1.93-fold greater I-124 uptake by CSC inoculation into ischemic compared to non-ischemic limbs at 2, 15, and 24 h, respectively. CT26 cells showed similar but smaller differences. PET findings were confirmed by ex vivo measurements and confocal microscopy. CONCLUSIONS NIS PET can help identify microenvironment conditions that influence early survival of implanted CSCs.
Collapse
Affiliation(s)
- Jin Won Park
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Korea
| | - Kyung-Ho Jung
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Korea
| | - Jin Hee Lee
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Korea
| | - Seung Hwan Moon
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Young Seok Cho
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Yearn Seung Choe
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Korea
| | - Kyung-Han Lee
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea. .,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Korea.
| |
Collapse
|
47
|
Saygin C, Wiechert A, Rao VS, Alluri R, Connor E, Thiagarajan PS, Hale JS, Li Y, Chumakova A, Jarrar A, Parker Y, Lindner DJ, Nagaraj AB, Kim JJ, DiFeo A, Abdul-Karim FW, Michener C, Rose PG, DeBernardo R, Mahdi H, McCrae KR, Lin F, Lathia JD, Reizes O. CD55 regulates self-renewal and cisplatin resistance in endometrioid tumors. J Exp Med 2017; 214:2715-2732. [PMID: 28838952 PMCID: PMC5584126 DOI: 10.1084/jem.20170438] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 05/22/2017] [Accepted: 06/27/2017] [Indexed: 12/26/2022] Open
Abstract
CD55 is a membrane complement regulatory protein that attenuates complement-mediated cytotoxicity. Saygin et al. elucidate a new role for CD55 as a signaling hub for cancer stem cell self-renewal and cisplatin resistance pathways in endometrioid tumors and open a new line of research into chemotherapeutic-refractory cancers. Effective targeting of cancer stem cells (CSCs) requires neutralization of self-renewal and chemoresistance, but these phenotypes are often regulated by distinct molecular mechanisms. Here we report the ability to target both of these phenotypes via CD55, an intrinsic cell surface complement inhibitor, which was identified in a comparative analysis between CSCs and non-CSCs in endometrioid cancer models. In this context, CD55 functions in a complement-independent manner and required lipid raft localization for CSC maintenance and cisplatin resistance. CD55 regulated self-renewal and core pluripotency genes via ROR2/JNK signaling and in parallel cisplatin resistance via lymphocyte-specific protein tyrosine kinase (LCK) signaling, which induced DNA repair genes. Targeting LCK signaling via saracatinib, an inhibitor currently undergoing clinical evaluation, sensitized chemoresistant cells to cisplatin. Collectively, our findings identify CD55 as a unique signaling node that drives self-renewal and therapeutic resistance through a bifurcating signaling axis and provides an opportunity to target both signaling pathways in endometrioid tumors.
Collapse
Affiliation(s)
- Caner Saygin
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Andrew Wiechert
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH.,Department of Obstetrics and Gynecology, Women's Health Institute, Cleveland Clinic, Cleveland, OH
| | - Vinay S Rao
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Ravi Alluri
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Elizabeth Connor
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH.,Department of Obstetrics and Gynecology, Women's Health Institute, Cleveland Clinic, Cleveland, OH
| | - Praveena S Thiagarajan
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - James S Hale
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Yan Li
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Anastasia Chumakova
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Awad Jarrar
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Yvonne Parker
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Daniel J Lindner
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Anil Belur Nagaraj
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH
| | - J Julie Kim
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL
| | - Analisa DiFeo
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Fadi W Abdul-Karim
- Pathology and Laboratory Medicine Institute, Department of Anatomical Pathology, Cleveland Clinic, Cleveland, OH
| | - Chad Michener
- Department of Obstetrics and Gynecology, Women's Health Institute, Cleveland Clinic, Cleveland, OH
| | - Peter G Rose
- Department of Obstetrics and Gynecology, Women's Health Institute, Cleveland Clinic, Cleveland, OH
| | - Robert DeBernardo
- Department of Obstetrics and Gynecology, Women's Health Institute, Cleveland Clinic, Cleveland, OH
| | - Haider Mahdi
- Department of Obstetrics and Gynecology, Women's Health Institute, Cleveland Clinic, Cleveland, OH
| | - Keith R McCrae
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Feng Lin
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Justin D Lathia
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH .,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH
| | - Ofer Reizes
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH .,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
48
|
Thiagarajan PS, Zheng Q, Bhagrath M, Mulkearns-Hubert EE, Myers MG, Lathia JD, Reizes O. STAT3 activation by leptin receptor is essential for TNBC stem cell maintenance. Endocr Relat Cancer 2017; 24:415-426. [PMID: 28729467 PMCID: PMC5551450 DOI: 10.1530/erc-16-0349] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 06/09/2017] [Indexed: 12/31/2022]
Abstract
Leptin (LEP) binds to the long form of the leptin receptor (LEPRb), leading to the activation of multiple signaling pathways that are potential targets for disrupting the obesity-breast cancer link. In triple-negative breast cancer (TNBC), LEP is hypothesized to predominantly mediate its tumorigenic effects via a subpopulation of LEPRb-positive tumor cells termed cancer stem cells (CSCs) that can initiate tumors and induce tumor progression. Previously, we showed that LEP promotes CSC survival in vivo Moreover, silencing LEPRb in TNBC cells compromised the CSC state. The mechanisms by which LEPRb regulates TNBC CSC intracellular signaling are not clear. We hypothesized that activation of LEPRb signaling is sufficient to drive CSC maintenance in TNBC. Here, we show that activation of LEPRb in non-CSCs isolated using our CSC reporter system resulted in a transition to the stem cell state. In CSCs, LEP induced STAT3 phosphorylation, whereas LEP did not induce STAT3 phosphorylation in non-CSCs. Introduction of constitutively active STAT3 into LEPRb-transfected non-CSCs significantly induced NANOG, SOX2 and OCT4 expression compared with control non-CSCs. To determine the intracellular phospho-tyrosine residue of LEPRb that is necessary for the induction of the stem cell state in non-CSCs, we transfected the tyrosine residue point mutants L985, F1077 and S1138 into non-CSCs. Non-CSCs transfected with the L985 mutant exhibited increased STAT3 phosphorylation, increased SOCS3 expression and an induction of GFP expression compared with non-CSCs expressing the F1077 and S1138 mutants. Our data demonstrate that LEPRb-induced STAT3 activation is essential for the induction and maintenance of TNBC CSCs.
Collapse
Affiliation(s)
- Praveena S Thiagarajan
- Department of Cellular and Molecular MedicineLerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular MedicineCleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
| | - Qiao Zheng
- Department of Cellular and Molecular MedicineLerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Manvir Bhagrath
- Department of Cellular and Molecular MedicineLerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Erin E Mulkearns-Hubert
- Department of Cellular and Molecular MedicineLerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Martin G Myers
- Departments of Internal Medicine and Molecular and Integrative PhysiologyUniversity of Michigan, Ann Arbor, Michigan, USA
| | - Justin D Lathia
- Department of Cellular and Molecular MedicineLerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular MedicineCleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
- Case Comprehensive Cancer CenterCase Western Reserve University, Cleveland, Ohio, USA
| | - Ofer Reizes
- Department of Cellular and Molecular MedicineLerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular MedicineCleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
- Case Comprehensive Cancer CenterCase Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
49
|
Abstract
Advances in cancer research in the past have led to an evolving understanding of cancer pathogenesis and the development of novel drugs that significantly improve patient outcomes. However, many patients still encounter treatment resistance, recurrence, or metastasis and eventually die from progressing disease. Experimental evidence indicates that a subpopulation of cancer cells, called cancer stem cells (CSCs), possess "stemness" properties similar to normal stem cells, including self-renewal, differentiation, and proliferative potential. These stemness properties are lost during differentiation and are governed by pathways such as STAT3, NANOG, NOTCH, WNT, and HEDGEHOG, which are highly dysregulated in CSCs due to genetic and epigenetic changes. Promising results have been observed in preclinical models targeting these CSCs through the disruption of stemness pathways in combination with current treatment modalities. This has led to anti-CSC-based clinical trials in multiple stages of development. In this review, we discuss the role of CSCs and stemness pathways in cancer treatment and how they relate to clinical observations. Because CSCs and the stemness pathways governing them may explain the negative clinical outcomes observed during treatment, it is important for oncologists to understand how they contribute to cancer progression and how they may be targeted to improve patient outcomes.
Collapse
Affiliation(s)
- Justin D Lathia
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., NC10, Cleveland, OH, 44195, USA.
| | - Huiping Liu
- Departments of Pharmacology and Medicine (Hematology/Oncology), Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
50
|
Strnadel J, Choi S, Fujimura K, Wang H, Zhang W, Wyse M, Wright T, Gross E, Peinado C, Park HW, Bui J, Kelber J, Bouvet M, Guan KL, Klemke RL. eIF5A-PEAK1 Signaling Regulates YAP1/TAZ Protein Expression and Pancreatic Cancer Cell Growth. Cancer Res 2017; 77:1997-2007. [PMID: 28381547 PMCID: PMC5392372 DOI: 10.1158/0008-5472.can-16-2594] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/05/2016] [Accepted: 12/30/2016] [Indexed: 01/16/2023]
Abstract
In pancreatic ductal adenocarcinoma (PDAC), mutant KRAS stimulates the translation initiation factor eIF5A and upregulates the focal adhesion kinase PEAK1, which transmits integrin and growth factor signals mediated by the tumor microenvironment. Although eIF5A-PEAK1 signaling contributes to multiple aggressive cancer cell phenotypes, the downstream signaling processes that mediate these responses are uncharacterized. Through proteomics and informatic analyses of PEAK1-depleted PDAC cells, we defined protein translation, cytoskeleton organization, and cell-cycle regulatory pathways as major pathways controlled by PEAK1. Biochemical and functional studies revealed that the transcription factors YAP1 and TAZ are key targets of eIF5A-PEAK1 signaling. YAP1/TAZ coimmunoprecipitated with PEAK1. Interfering with eIF5A-PEAK1 signaling in PDAC cells inhibited YAP/TAZ protein expression, decreasing expression of stem cell-associated transcription factors (STF) including Oct4, Nanog, c-Myc, and TEAD, thereby decreasing three-dimensional (3D) tumor sphere growth. Conversely, amplified eIF5A-PEAK1 signaling increased YAP1/TAZ expression, increasing expression of STF and enhancing 3D tumor sphere growth. Informatic interrogation of mRNA sequence databases revealed upregulation of the eIF5A-PEAK1-YAP1-TEAD signaling module in PDAC patients. Taken together, our findings indicate that eIF5A-PEAK1-YAP signaling contributes to PDAC development by regulating an STF program associated with increased tumorigenicity. Cancer Res; 77(8); 1997-2007. ©2017 AACR.
Collapse
Affiliation(s)
- Jan Strnadel
- Department of Pathology, University of California, San Diego, La Jolla, California
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Sunkyu Choi
- Department of Pathology, University of California, San Diego, La Jolla, California
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Ken Fujimura
- Department of Pathology, University of California, San Diego, La Jolla, California
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Huawei Wang
- Department of Pathology, University of California, San Diego, La Jolla, California
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Wei Zhang
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Meghan Wyse
- Department of Pathology, University of California, San Diego, La Jolla, California
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Tracy Wright
- Department of Pathology, University of California, San Diego, La Jolla, California
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Emilie Gross
- Department of Pathology, University of California, San Diego, La Jolla, California
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Carlos Peinado
- Department of Pathology, University of California, San Diego, La Jolla, California
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Hyun Woo Park
- Moores Cancer Center, University of California, San Diego, La Jolla, California
- Department of Pharmacology, University of California, San Diego, La Jolla, California
| | - Jack Bui
- Department of Pathology, University of California, San Diego, La Jolla, California
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Jonathan Kelber
- Department of Biology, California State University Northridge, Northridge, California
| | - Michael Bouvet
- Department of Surgery, University of California, San Diego, La Jolla, California
| | - Kun-Liang Guan
- Moores Cancer Center, University of California, San Diego, La Jolla, California
- Department of Pharmacology, University of California, San Diego, La Jolla, California
| | - Richard L Klemke
- Department of Pathology, University of California, San Diego, La Jolla, California.
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| |
Collapse
|