1
|
Wu B, Shi X, Jiang M, Liu H. Cross-talk between cancer stem cells and immune cells: potential therapeutic targets in the tumor immune microenvironment. Mol Cancer 2023; 22:38. [PMID: 36810098 PMCID: PMC9942413 DOI: 10.1186/s12943-023-01748-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 02/15/2023] [Indexed: 02/24/2023] Open
Abstract
Ongoing research has revealed that the existence of cancer stem cells (CSCs) is one of the biggest obstacles in the current cancer therapy. CSCs make an influential function in tumor progression, recurrence and chemoresistance due to their typical stemness characteristics. CSCs are preferentially distributed in niches, and those niche sites exhibit characteristics typical of the tumor microenvironment (TME). The complex interactions between CSCs and TME illustrate these synergistic effects. The phenotypic heterogeneity within CSCs and the spatial interactions with the surrounding tumor microenvironment led to increased therapeutic challenges. CSCs interact with immune cells to protect themselves against immune clearance by exploiting the immunosuppressive function of multiple immune checkpoint molecules. CSCs also can protect themselves against immune surveillance by excreting extracellular vesicles (EVs), growth factors, metabolites and cytokines into the TME, thereby modulating the composition of the TME. Therefore, these interactions are also being considered for the therapeutic development of anti-tumor agents. We discuss here the immune molecular mechanisms of CSCs and comprehensively review the interplay between CSCs and the immune system. Thus, studies on this topic seem to provide novel ideas for reinvigorating therapeutic approaches to cancer.
Collapse
Affiliation(s)
- Bo Wu
- grid.459742.90000 0004 1798 5889Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042 China
| | - Xiang Shi
- grid.459742.90000 0004 1798 5889Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042 China
| | - Meixi Jiang
- grid.412644.10000 0004 5909 0696Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032 China
| | - Hongxu Liu
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China.
| |
Collapse
|
2
|
Ngo MT, Sarkaria JN, Harley BA. Perivascular Stromal Cells Instruct Glioblastoma Invasion, Proliferation, and Therapeutic Response within an Engineered Brain Perivascular Niche Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201888. [PMID: 36109186 PMCID: PMC9631060 DOI: 10.1002/advs.202201888] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 08/12/2022] [Indexed: 06/15/2023]
Abstract
Glioblastoma (GBM) tumor cells are found in the perivascular niche microenvironment and are believed to associate closely with the brain microvasculature. However, it is largely unknown how the resident cells of the perivascular niche, such as endothelial cells, pericytes, and astrocytes, influence GBM tumor cell behavior and disease progression. A 3D in vitro model of the brain perivascular niche developed by encapsulating brain-derived endothelial cells, pericytes, and astrocytes in a gelatin hydrogel is described. It is shown that brain perivascular stromal cells, namely pericytes and astrocytes, contribute to vascular architecture and maturation. Cocultures of patient-derived GBM tumor cells with brain microvascular cells are used to identify a role for pericytes and astrocytes in establishing a perivascular niche environment that modulates GBM cell invasion, proliferation, and therapeutic response. Engineered models provide unique insight regarding the spatial patterning of GBM cell phenotypes in response to a multicellular model of the perivascular niche. Critically, it is shown that engineered perivascular models provide an important resource to evaluate mechanisms by which intercellular interactions modulate GBM tumor cell behavior, drug response, and provide a framework to consider patient-specific disease phenotypes.
Collapse
Affiliation(s)
- Mai T. Ngo
- Department Chemical and Biomolecular EngineeringUniversity of Illinois Urbana‐ChampaignUrbanaIL61801USA
| | | | - Brendan A.C. Harley
- Department Chemical and Biomolecular EngineeringUniversity of Illinois Urbana‐ChampaignUrbanaIL61801USA
- Carl R. Woese Institute for Genomic BiologyUniversity of Illinois Urbana‐ChampaignUrbanaIL61801USA
- Cancer Center at IllinoisUniversity of Illinois Urbana‐ChampaignUrbanaIL61801USA
| |
Collapse
|
3
|
Rocha JD, Uribe D, Delgado J, Niechi I, Alarcón S, Erices JI, Melo R, Fernández-Gajardo R, Salazar-Onfray F, San Martín R, Quezada Monrás C. A 2B Adenosine Receptor Enhances Chemoresistance of Glioblastoma Stem-Like Cells under Hypoxia: New Insights into MRP3 Transporter Function. Int J Mol Sci 2022; 23:ijms23169022. [PMID: 36012307 PMCID: PMC9409164 DOI: 10.3390/ijms23169022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 11/24/2022] Open
Abstract
Glioblastoma is the most common and aggressive primary brain tumor, characterized by its high chemoresistance and the presence of a cell subpopulation that persists under hypoxic niches, called glioblastoma stem-like cells (GSCs). The chemoresistance of GSCs is mediated in part by adenosine signaling and ABC transporters, which extrude drugs outside the cell, such as the multidrug resistance-associated proteins (MRPs) subfamily. Adenosine promotes MRP1-dependent chemoresistance under normoxia. However, adenosine/MRPs-dependent chemoresistance under hypoxia has not been studied until now. Transcript and protein levels were determined by RT-qPCR and Western blot, respectively. MRP extrusion capacity was determined by intracellular 5 (6)-Carboxyfluorescein diacetate (CFDA) accumulation. Cell viability was measured by MTS assays. Cell cycle and apoptosis were determined by flow cytometry. Here, we show for the first time that MRP3 expression is induced under hypoxia through the A2B adenosine receptor. Hypoxia enhances MRP-dependent extrusion capacity and the chemoresistance of GSCs. Meanwhile, MRP3 knockdown decreases GSC viability under hypoxia. Downregulation of the A2B receptor decreases MRP3 expression and chemosensibilizes GSCs treated with teniposide under hypoxia. These data suggest that hypoxia-dependent activation of A2B adenosine receptor promotes survival of GSCs through MRP3 induction.
Collapse
Affiliation(s)
- José-Dellis Rocha
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Daniel Uribe
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Javiera Delgado
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Ignacio Niechi
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5110566, Chile
- Millennium Institute on Immunology and Immunotherapy, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Sebastián Alarcón
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5110566, Chile
| | - José Ignacio Erices
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5110566, Chile
- Millennium Institute on Immunology and Immunotherapy, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Rómulo Melo
- Servicio de Neurocirugía, Instituto de Neurocirugía Dr. Asenjo, Santiago 7500691, Chile
| | | | - Flavio Salazar-Onfray
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 7500691, Chile
- Millennium Institute on Immunology and Immunotherapy, Facultad de Medicina, Universidad de Chile, Santiago 7500691, Chile
| | - Rody San Martín
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Claudia Quezada Monrás
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5110566, Chile
- Millennium Institute on Immunology and Immunotherapy, Universidad Austral de Chile, Valdivia 5090000, Chile
- Correspondence: ; Tel.: +56-63-2221332
| |
Collapse
|
4
|
Notch signaling in malignant gliomas: supporting tumor growth and the vascular environment. Cancer Metastasis Rev 2022; 41:737-747. [PMID: 35624227 DOI: 10.1007/s10555-022-10041-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 05/18/2022] [Indexed: 11/02/2022]
Abstract
Glioblastoma is the most malignant form of glioma, which is the most commonly occurring tumor of the central nervous system. Notch signaling in glioblastoma is considered to be a marker of an undifferentiated tumor cell state, associated with tumor stem cells. Notch is also known for facilitating tumor dormancy escape, recurrence and progression after treatment. Studies in vitro suggest that reducing, removing or blocking the expression of this gene triggers tumor cell differentiation, which shifts the phenotype away from stemness status and consequently facilitates treatment. In contrast, in the vasculature, Notch appears to also function as an important receptor that defines mature non-leaking vessels, and increasing its expression promotes tumor normalization in models of cancer in vivo. Failures in clinical trials with Notch inhibitors are potentially related to their opposing effects on the tumor versus the tumor vasculature, which points to the need for a greater understanding of this signaling pathway.
Collapse
|
5
|
Ngo MT, Karvelis E, Harley BAC. Multidimensional hydrogel models reveal endothelial network angiocrine signals increase glioblastoma cell number, invasion, and temozolomide resistance. Integr Biol (Camb) 2021; 12:139-149. [PMID: 32507878 DOI: 10.1093/intbio/zyaa010] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 11/13/2022]
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumor. The tissue microenvironment adjacent to vasculature, termed the perivascular niche, has been implicated in promoting biological processes involved in glioblastoma progression such as invasion, proliferation, and therapeutic resistance. However, the exact nature of the cues that support tumor cell aggression in this niche is largely unknown. Soluble angiocrine factors secreted by tumor-associated vasculature have been shown to support such behaviors in other cancer types. Here, we exploit macroscopic and microfluidic gelatin hydrogel platforms to profile angiocrine factors secreted by self-assembled endothelial networks and evaluate their relevance to glioblastoma biology. Aggregate angiocrine factors support increases in U87-MG cell number, migration, and therapeutic resistance to temozolomide. We also identify a novel role for TIMP1 in facilitating glioblastoma tumor cell migration. Overall, this work highlights the use of multidimensional hydrogel models to evaluate the role of angiocrine signals in glioblastoma progression.
Collapse
Affiliation(s)
- Mai T Ngo
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Elijah Karvelis
- Dept. Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Brendan A C Harley
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
6
|
Monensin inhibits glioblastoma angiogenesis via targeting multiple growth factor receptor signaling. Biochem Biophys Res Commun 2020; 530:479-484. [PMID: 32595038 DOI: 10.1016/j.bbrc.2020.05.057] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 05/10/2020] [Indexed: 02/07/2023]
Abstract
Glioblastoma is characterized by the extensive vascularization with poor prognosis. Targeting both tumor cell and angiogenesis may present an effective therapeutic strategy for glioblastoma. Monensin, a polyether ionophore antibiotic, has been recently recognized as promising anticancer drug candidate due to its potent and selective anti-tumor activities. However, little is known on the effects of monensin on tumor angiogenesis. In this work, we investigated the effects and underlying mechanisms of monensin on glioblastoma angiogenesis and growth. We show that monensin at nanomolar concentrations inhibits early stages of capillary network formation of glioblastoma endothelial cell. Monensin inhibited multiple endothelial cellular events, including migration, growth and survival, without affecting adhesion to Matrigel. We further demonstrate that monensin acts on endothelial cells via suppressing VEGFR- and EGFR-mediated signaling pathways. Monensin also inhibits proliferation and induces apoptosis in a panel of glioblastoma cells. However, monensin is more effective in targeting endothelial cells than tumor cells. Using glioblastoma growth xenograft mice model, we show that monensin at tolerable dose effectively inhibits glioblastoma growth. Of note, there is a significant decreased tumor vascularization from monensin-treated mice. Our work clearly demonstrates the anti-angiogenic activity of monensin and its ability in suppressing multiple tyrosine kinase receptor-mediated pathways. Our findings suggest that is a useful addition to the treatment armamentarium for glioblastoma.
Collapse
|
7
|
Brooks EA, Galarza S, Gencoglu MF, Cornelison RC, Munson JM, Peyton SR. Applicability of drug response metrics for cancer studies using biomaterials. Philos Trans R Soc Lond B Biol Sci 2019; 374:20180226. [PMID: 31431182 PMCID: PMC6627013 DOI: 10.1098/rstb.2018.0226] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2019] [Indexed: 12/31/2022] Open
Abstract
Bioengineers have built models of the tumour microenvironment (TME) in which to study cell-cell interactions, mechanisms of cancer growth and metastasis, and to test new therapies. These models allow researchers to culture cells in conditions that include features of the in vivo TME implicated in regulating cancer progression, such as extracellular matrix (ECM) stiffness, integrin binding to the ECM, immune and stromal cells, growth factor and cytokine depots, and a three-dimensional geometry more representative of the in vivo TME than tissue culture polystyrene (TCPS). These biomaterials could be particularly useful for drug screening applications to make better predictions of efficacy, offering better translation to preclinical models and clinical trials. However, it can be challenging to compare drug response reports across different biomaterial platforms in the current literature. This is, in part, a result of inconsistent reporting and improper use of drug response metrics, and vast differences in cell growth rates across a large variety of biomaterial designs. This study attempts to clarify the definitions of drug response measurements used in the field, and presents examples in which these measurements can and cannot be applied. We suggest as best practice to measure the growth rate of cells in the absence of drug, and follow our 'decision tree' when reporting drug response metrics. This article is part of a discussion meeting issue 'Forces in cancer: interdisciplinary approaches in tumour mechanobiology'.
Collapse
Affiliation(s)
- Elizabeth A. Brooks
- Department of Chemical Engineering, University of Massachusetts Amherst, 240 Thatcher Road, Amherst, MA 01003-9364, USA
| | - Sualyneth Galarza
- Department of Chemical Engineering, University of Massachusetts Amherst, 240 Thatcher Road, Amherst, MA 01003-9364, USA
| | - Maria F. Gencoglu
- Department of Chemical Engineering, University of Massachusetts Amherst, 240 Thatcher Road, Amherst, MA 01003-9364, USA
| | - R. Chase Cornelison
- Department of Biomedical Engineering and Mechanics, Virginia Tech, 325 Stanger Street, Blacksburg, VA 24061, USA
| | - Jennifer M. Munson
- Department of Biomedical Engineering and Mechanics, Virginia Tech, 325 Stanger Street, Blacksburg, VA 24061, USA
| | - Shelly R. Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst, 240 Thatcher Road, Amherst, MA 01003-9364, USA
| |
Collapse
|
8
|
Zhang Y, Zhang C, Zhang H, Zeng W, Li S, Chen C, Song X, Sun J, Sun Z, Cui C, Cao X, Zheng L, Wang P, Zhao W, Zhang Z, Xu Y, Zhu M, Chen H. ZIPK mediates endothelial cell contraction through myosin light chain phosphorylation and is required for ischemic-reperfusion injury. FASEB J 2019; 33:9062-9074. [PMID: 31180722 DOI: 10.1096/fj.201802052rrr] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The paracellular gap formed by endothelial cell (EC) contraction is fundamental for endothelium permeability, but the mechanism underlying EC contraction has yet to be determined. Here, we identified the zipper-interacting protein kinase (ZIPK) as the kinase for EC contraction and myosin light chain (MLC) phosphorylation. Inhibition of ZIPK activity by pharmacological inhibitors and small interfering RNAs led to a significant decrease in the mono- and diphosphorylation of MLCs along with a contractile response to thrombin, suggesting an essential role of ZIPK in EC paracellular permeability. To assess the role of ZIPK in vivo, we established mouse lines with conditional deletion of Zipk gene. The endothelium-specific deletion of Zipk led to embryonic lethality, whereas the UBC-CreERT2-mediated deletion of Zipk by tamoxifen induction at adulthood caused no apparent phenotype. The induced deletion of Zipk significantly inhibited ischemia-reperfusion-induced blood-brain barrier dysfunction and neuronal injuries from middle cerebral artery occlusion and reperfusion, as evidenced by reduced infarct and edema volume, attenuated Evans blue dye leakage, and improved neuronal behavior. We thus concluded that ZIPK and its phosphorylation of MLC were required for EC contraction and ischemic neuronal injuries. ZIPK may be a prospective therapeutic target for stroke.-Zhang, Y., Zhang, C., Zhang, H., Zeng, W., Li, S., Chen, C., Song, X., Sun, J., Sun, Z., Cui, C., Cao, X., Zheng, L., Wang, P., Zhao, W., Zhang, Z., Xu, Y., Zhu, M., Chen, H. ZIPK mediates endothelial cell contraction through myosin light chain phosphorylation and is required for ischemic-reperfusion injury.
Collapse
Affiliation(s)
- Yiteng Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Chenghai Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, and Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China
| | - He Zhang
- Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Weiwei Zeng
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Shuai Li
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Caiping Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, and Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China
| | - Xiaobin Song
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Jie Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, and Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China
| | - Zhiyuan Sun
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Congcong Cui
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Xiang Cao
- Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Lirong Zheng
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Pei Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, and Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China
| | - Wei Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, and Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China
| | - Zhao Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yun Xu
- Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Minsheng Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, and Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China
| | - Huaqun Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
9
|
Ngo MT, Harley BAC. Perivascular signals alter global gene expression profile of glioblastoma and response to temozolomide in a gelatin hydrogel. Biomaterials 2019; 198:122-134. [PMID: 29941152 DOI: 10.1101/273763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/30/2018] [Accepted: 06/10/2018] [Indexed: 05/25/2023]
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumor, with patients exhibiting poor survival (median survival time: 15 months). Difficulties in treating GBM include not only the inability to resect the diffusively-invading tumor cells, but also therapeutic resistance. The perivascular niche (PVN) within the GBM tumor microenvironment contributes significantly to tumor cell invasion, cancer stem cell maintenance, and has been shown to protect tumor cells from radiation and chemotherapy. In this study, we examine how the inclusion of non-tumor cells in culture with tumor cells within a hydrogel impacts the overall gene expression profile of an in vitro artificial perivascular niche (PVN) comprised of endothelial and stromal cells directly cultured with GBM tumor cells within a methacrylamide-functionalized gelatin hydrogel. Using RNA-seq, we demonstrate that genes related to angiogenesis and extracellular matrix remodeling are upregulated in the PVN model compared to hydrogels containing only tumor or perivascular niche cells, while downregulated genes are related to cell cycle and DNA damage repair. Signaling pathways and genes commonly implicated in GBM malignancy, such as MGMT, EGFR, PI3K-Akt signaling, and Ras/MAPK signaling are also upregulated in the PVN model. We describe the kinetics of gene expression within the PVN hydrogels over a course of 14 days, observing the patterns associated with tumor cell-mediated endothelial network co-option and regression. We finally examine the effect of temozolomide, a frontline chemotherapy used clinically against GBM, on the PVN culture. Notably, the PVN model is less responsive to TMZ compared to hydrogels containing only tumor cells. Overall, these results demonstrate that inclusion of cellular and matrix-associated elements of the PVN within an in vitro model of GBM allows for the development of gene expression patterns and therapeutic response relevant to GBM.
Collapse
Affiliation(s)
- Mai T Ngo
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Brendan A C Harley
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
10
|
Zhong M, Zhong C, Hu P, Cui W, Wang G, Gao H, Liu C, Liu Z, Li Z, Li C, Gohda E. Restoration of stemness-high tumor cell-mediated suppression of murine dendritic cell activity and inhibition of tumor growth by low molecular weight oyster polysaccharide. Int Immunopharmacol 2018; 65:221-232. [PMID: 30321818 DOI: 10.1016/j.intimp.2018.10.003] [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: 05/05/2018] [Revised: 09/24/2018] [Accepted: 10/03/2018] [Indexed: 12/11/2022]
Abstract
Dendritic cells (DCs) play key regulatory roles in tumor immunity: increased activity of DCs infiltrating tumor tissues leads to enhancement of tumor immunity. Functions of DCs are also modulated by tumor cell-derived factors. Here, we investigated the effects of low molecular weight oyster polysaccharide (LMW-OPS) on differentiation and function of bone marrow-derived DCs (BMDCs) exposed to a conditioned medium (CM) obtained from spheres of stemness-high colorectal cancer cell lines CMT93 and CT26. The CM containing a detectable level of TGF-β1 was found to down-regulate the surface expression of major histocompatibility complex class II of BMDCs and to inhibit the potency of BMDCs to stimulate T cells. Those suppressions were partly restored and completely restored by addition of anti-TGF-β1 and LMW-OPS, respectively. Production of IFN-γ during allogeneic T cell responses was inhibited by the CM, whereas production of TGF-β1 was augmented by the CM. The IFN-γ profile was also reversed by addition of LMW-OPS. Nuclear translocation of β-catenin, but not that of NF-κB p65, was induced by TGF-β1. NF-κB p65 nuclear translocation, but not β-catenin nuclear translocation, was induced by LMW-OPS. Intraperitoneal injection of LMW-OPS significantly suppressed tumor growth in syngeneic tumor models using CMT93 and CT26 sphere cells, whereas it had no inhibitory effect on the proliferation of either cell line. The results demonstrated that LMW-OPS relieved stemness-high tumor cell-mediated suppression of BMDC function and indicated the in vivo anti-tumor activity of LMW-OPS in which re-stimulation of the activity of DCs infiltrating tumor tissues is presumed to be involved.
Collapse
Affiliation(s)
- Ming Zhong
- Institute of Tumor Pharmacology, Jining Medical College, Rizhao, China.
| | - Cheng Zhong
- Division of Stem Cell Dynamics, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Pei Hu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Wen Cui
- Institute of Tumor Pharmacology, Jining Medical College, Rizhao, China
| | - Guanghui Wang
- Institute of Tumor Pharmacology, Jining Medical College, Rizhao, China
| | - Huijei Gao
- Institute of Tumor Pharmacology, Jining Medical College, Rizhao, China
| | - Chao Liu
- Institute of Tumor Pharmacology, Jining Medical College, Rizhao, China
| | - Zhiqiang Liu
- Institute of Tumor Pharmacology, Jining Medical College, Rizhao, China
| | - Zhihua Li
- Institute of Tumor Pharmacology, Jining Medical College, Rizhao, China
| | - Chunxia Li
- Institute of Tumor Pharmacology, Jining Medical College, Rizhao, China
| | - Eiichi Gohda
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
11
|
Ngo MT, Harley BAC. Perivascular signals alter global gene expression profile of glioblastoma and response to temozolomide in a gelatin hydrogel. Biomaterials 2018; 198:122-134. [PMID: 29941152 DOI: 10.1016/j.biomaterials.2018.06.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/30/2018] [Accepted: 06/10/2018] [Indexed: 12/22/2022]
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumor, with patients exhibiting poor survival (median survival time: 15 months). Difficulties in treating GBM include not only the inability to resect the diffusively-invading tumor cells, but also therapeutic resistance. The perivascular niche (PVN) within the GBM tumor microenvironment contributes significantly to tumor cell invasion, cancer stem cell maintenance, and has been shown to protect tumor cells from radiation and chemotherapy. In this study, we examine how the inclusion of non-tumor cells in culture with tumor cells within a hydrogel impacts the overall gene expression profile of an in vitro artificial perivascular niche (PVN) comprised of endothelial and stromal cells directly cultured with GBM tumor cells within a methacrylamide-functionalized gelatin hydrogel. Using RNA-seq, we demonstrate that genes related to angiogenesis and extracellular matrix remodeling are upregulated in the PVN model compared to hydrogels containing only tumor or perivascular niche cells, while downregulated genes are related to cell cycle and DNA damage repair. Signaling pathways and genes commonly implicated in GBM malignancy, such as MGMT, EGFR, PI3K-Akt signaling, and Ras/MAPK signaling are also upregulated in the PVN model. We describe the kinetics of gene expression within the PVN hydrogels over a course of 14 days, observing the patterns associated with tumor cell-mediated endothelial network co-option and regression. We finally examine the effect of temozolomide, a frontline chemotherapy used clinically against GBM, on the PVN culture. Notably, the PVN model is less responsive to TMZ compared to hydrogels containing only tumor cells. Overall, these results demonstrate that inclusion of cellular and matrix-associated elements of the PVN within an in vitro model of GBM allows for the development of gene expression patterns and therapeutic response relevant to GBM.
Collapse
Affiliation(s)
- Mai T Ngo
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Brendan A C Harley
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
12
|
Torres A, Vargas Y, Uribe D, Jaramillo C, Gleisner A, Salazar-Onfray F, López MN, Melo R, Oyarzún C, San Martín R, Quezada C. Adenosine A3 receptor elicits chemoresistance mediated by multiple resistance-associated protein-1 in human glioblastoma stem-like cells. Oncotarget 2018; 7:67373-67386. [PMID: 27634913 PMCID: PMC5341882 DOI: 10.18632/oncotarget.12033] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 08/29/2016] [Indexed: 12/31/2022] Open
Abstract
MRP1 transporter correlates positively with glioma malignancy and the Multiple Drug Resistance (MDR) phenotype in Glioblastoma Multiforme (GBM). Evidence shows that the MRP1 transporter is controlled by the adenosine signalling axis. The aim of this study was to identify the role of adenosine on the MDR phenotype in Glioblastoma Stem-like Cells (GSCs), the cell population responsible for the tumorigenic and chemoresistance capabilities of this tumour. We found that GSCs have increased intrinsic capacity to generate extracellular adenosine, thus controlling MRP1 transporter expression and activity via activation of the adenosine A3 receptor (A3AR). We showed PI3K/Akt and MEK/ERK1/2 signaling pathways downstream A3AR to control MRP1 in GSCs. In vitro pharmacological blockade of A3AR had a chemosensitizing effect, enhancing the actions of antitumour drugs and decreasing cell viability and proliferation of GSCs. In addition, we produced an in vivo xenograft model by subcutaneous inoculation of human GSCs in NOD/SCID-IL2Rg null mice. Pharmacological blockade of A3AR generated a chemosensitizing effect, enhancing the effectiveness of the MRP1 transporter substrate, vincristine, reducing tumour size and the levels of CD44 and Nestin stem cell markers as well as the Ki-67 proliferation indicator. In conclusion, we demonstrated the chemosensitizing effect of A3AR blockade on GSCs.
Collapse
Affiliation(s)
- Angelo Torres
- Laboratorio de Patología Molecular, Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Yosselyn Vargas
- Laboratorio de Patología Molecular, Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Daniel Uribe
- Laboratorio de Patología Molecular, Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Catherine Jaramillo
- Laboratorio de Patología Molecular, Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Alejandra Gleisner
- Instituto Milenio de Inmunología e Inmunoterapia, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Flavio Salazar-Onfray
- Instituto Milenio de Inmunología e Inmunoterapia, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Mercedes N López
- Instituto Milenio de Inmunología e Inmunoterapia, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Rómulo Melo
- Servicio de Neurocirugía, Instituto de Neurocirugía Dr. Asenjo, Santiago, Chile
| | - Carlos Oyarzún
- Laboratorio de Patología Molecular, Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Rody San Martín
- Laboratorio de Patología Molecular, Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Claudia Quezada
- Laboratorio de Patología Molecular, Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
13
|
Lindoso RS, Collino F, Vieyra A. Extracellular vesicles as regulators of tumor fate: crosstalk among cancer stem cells, tumor cells and mesenchymal stem cells. Stem Cell Investig 2017; 4:75. [PMID: 29057247 DOI: 10.21037/sci.2017.08.08] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 08/27/2017] [Indexed: 12/16/2022]
Abstract
The tumor microenvironment comprises a heterogeneous population of tumorigenic and non-tumorigenic cells. Cancer stem cells (CSCs) and mesenchymal stem cells (MSCs) are components of this microenvironment and have been described as key regulators of different aspects of tumor physiology. They act differently on the tumor: CSCs are described as tumor initiators and are associated with tumor growth, drug resistance and metastasis; MSCs can integrate the tumor microenvironment after recruitment and interact with cancer cells to promote tumor modifications. Extracellular vesicles (EVs) have emerged as an important mechanism of cell communication under the physiological and pathological conditions. In cancer, secretion of EVs seems to be one of the main mechanisms by which stem cells interact with other tumor and non-tumor cells. The transfer of bioactive molecules (lipids, proteins and RNAs) compartmentalized into EVs triggers different responses in the target cells, regulating several processes in the tumor as angiogenesis, tumor invasiveness and immune escape. This review focuses on the role of CSCs and MSCs in modulating the tumor microenvironment through secretion of EVs, addressing different aspects of the multidirectional interactions among stem cells, tumor and tumor-associated cells.
Collapse
Affiliation(s)
- Rafael Soares Lindoso
- Carlos Chagas Institute of Biophysics, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.,National Center for Structural Biology and Bioimaging-CENABIO, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Federica Collino
- Carlos Chagas Institute of Biophysics, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.,National Center for Structural Biology and Bioimaging-CENABIO, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Adalberto Vieyra
- Carlos Chagas Institute of Biophysics, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.,National Center for Structural Biology and Bioimaging-CENABIO, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.,Graduate Program of Translational Biomedicine, Grande Rio University, 25071-202 Duque de Caxias, Brazil
| |
Collapse
|
14
|
Mohanty S, Chen Z, Li K, Morais GR, Klockow J, Yerneni K, Pisani L, Chin FT, Mitra S, Cheshier S, Chang E, Gambhir SS, Rao J, Loadman PM, Falconer RA, Daldrup-Link HE. A Novel Theranostic Strategy for MMP-14-Expressing Glioblastomas Impacts Survival. Mol Cancer Ther 2017; 16:1909-1921. [PMID: 28659432 DOI: 10.1158/1535-7163.mct-17-0022] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 05/09/2017] [Accepted: 06/12/2017] [Indexed: 02/06/2023]
Abstract
Glioblastoma (GBM) has a dismal prognosis. Evidence from preclinical tumor models and human trials indicates the role of GBM-initiating cells (GIC) in GBM drug resistance. Here, we propose a new treatment option with tumor enzyme-activatable, combined therapeutic and diagnostic (theranostic) nanoparticles, which caused specific toxicity against GBM tumor cells and GICs. The theranostic cross-linked iron oxide nanoparticles (CLIO) were conjugated to a highly potent vascular disrupting agent (ICT) and secured with a matrix-metalloproteinase (MMP-14) cleavable peptide. Treatment with CLIO-ICT disrupted tumor vasculature of MMP-14-expressing GBM, induced GIC apoptosis, and significantly impaired tumor growth. In addition, the iron core of CLIO-ICT enabled in vivo drug tracking with MR imaging. Treatment with CLIO-ICT plus temozolomide achieved tumor remission and significantly increased survival of human GBM-bearing mice by more than 2-fold compared with treatment with temozolomide alone. Thus, we present a novel therapeutic strategy with significant impact on survival and great potential for clinical translation. Mol Cancer Ther; 16(9); 1909-21. ©2017 AACR.
Collapse
Affiliation(s)
- Suchismita Mohanty
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
| | - Zixin Chen
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
| | - Kai Li
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
| | - Goreti Ribeiro Morais
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford, United Kingdom
| | - Jessica Klockow
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
| | - Ketan Yerneni
- Department of Biology, Skidmore College, Saratoga Springs, New York
| | - Laura Pisani
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
| | - Frederick T Chin
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
| | - Siddharta Mitra
- Department of Neurosurgery, Institute for Stem Cell Biology and Regenerative Medicine and Division of Pediatric Neurosurgery, Lucile Packard Children's Hospital, Stanford University, Stanford, California
| | - Samuel Cheshier
- Department of Neurosurgery, Institute for Stem Cell Biology and Regenerative Medicine and Division of Pediatric Neurosurgery, Lucile Packard Children's Hospital, Stanford University, Stanford, California
| | | | - Sanjiv Sam Gambhir
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
- Department of Bioengineering, Stanford University, Stanford, California
- Department of Materials Science & Engineering, Stanford University, Stanford, California
| | - Jianghong Rao
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
| | - Paul M Loadman
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford, United Kingdom
| | - Robert A Falconer
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford, United Kingdom
| | - Heike E Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California.
| |
Collapse
|
15
|
Prasetyanti PR, Medema JP. Intra-tumor heterogeneity from a cancer stem cell perspective. Mol Cancer 2017; 16:41. [PMID: 28209166 PMCID: PMC5314464 DOI: 10.1186/s12943-017-0600-4] [Citation(s) in RCA: 507] [Impact Index Per Article: 72.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 01/20/2017] [Indexed: 02/08/2023] Open
Abstract
Tumor heterogeneity represents an ongoing challenge in the field of cancer therapy. Heterogeneity is evident between cancers from different patients (inter-tumor heterogeneity) and within a single tumor (intra-tumor heterogeneity). The latter includes phenotypic diversity such as cell surface markers, (epi)genetic abnormality, growth rate, apoptosis and other hallmarks of cancer that eventually drive disease progression and treatment failure. Cancer stem cells (CSCs) have been put forward to be one of the determining factors that contribute to intra-tumor heterogeneity. However, recent findings have shown that the stem-like state in a given tumor cell is a plastic quality. A corollary to this view is that stemness traits can be acquired via (epi)genetic modification and/or interaction with the tumor microenvironment (TME). Here we discuss factors contributing to this CSC heterogeneity and the potential implications for cancer therapy.
Collapse
Affiliation(s)
- Pramudita R Prasetyanti
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental Molecular Medicine (CEMM), Academic Medical Center (AMC), University of Amsterdam, 1105AZ, Amsterdam, The Netherlands.,Cancer Center Amsterdam and Cancer Genomics Center, Amsterdam, The Netherlands
| | - Jan Paul Medema
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental Molecular Medicine (CEMM), Academic Medical Center (AMC), University of Amsterdam, 1105AZ, Amsterdam, The Netherlands. .,Cancer Center Amsterdam and Cancer Genomics Center, Amsterdam, The Netherlands. .,Academic Medical Center, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.
| |
Collapse
|
16
|
Rosa AI, Grade S, Santos SD, Bernardino L, Chen TC, Relvas J, Hofman FM, Agasse F. Heterocellular Contacts with Mouse Brain Endothelial Cells Via Laminin and α6β1 Integrin Sustain Subventricular Zone (SVZ) Stem/Progenitor Cells Properties. Front Cell Neurosci 2016; 10:284. [PMID: 28018177 PMCID: PMC5156690 DOI: 10.3389/fncel.2016.00284] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 11/28/2016] [Indexed: 01/08/2023] Open
Abstract
Neurogenesis in the subventricular zone (SVZ) is regulated by diffusible factors and cell–cell contacts. In vivo, SVZ stem cells are associated with the abluminal surface of blood vessels and such interactions are thought to regulate their neurogenic capacity. SVZ neural stem cells (NSCs) have been described to contact endothelial-derived laminin via α6β1 integrin. To elucidate whether heterocellular contacts with brain endothelial cells (BEC) regulate SVZ cells neurogenic capacities, cocultures of SVZ neurospheres and primary BEC, both obtained from C57BL/6 mice, were performed. The involvement of laminin-integrin interactions in SVZ homeostasis was tested in three ways. Firstly, SVZ cells were analyzed following incubation of BEC with the protein synthesis inhibitor cycloheximide (CHX) prior to coculture, a treatment expected to decrease membrane proteins. Secondly, SVZ cells were cocultured with BEC in the presence of an anti-α6 integrin neutralizing antibody. Thirdly, BEC were cultured with β1−/− SVZ cells. We showed that contact with BEC supports, at least in part, proliferation and stemness of SVZ cells, as evaluated by the number of BrdU positive (+) and Sox2+ cells in contact with BEC. These effects are dependent on BEC-derived laminin binding to α6β1 integrin and are decreased in cocultures incubated with anti-α6 integrin neutralizing antibody and in cocultures with SVZ β1−/− cells. Moreover, BEC-derived laminin sustains stemness in SVZ cell cultures via activation of the Notch and mTOR signaling pathways. Our results show that BEC/SVZ interactions involving α6β1 integrin binding to laminin, contribute to SVZ cell proliferation and stemness.
Collapse
Affiliation(s)
- Alexandra I Rosa
- Centre for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal; Department of Pathology, University of Southern CaliforniaLos Angeles, CA, USA; Department of Neurological Surgery, University of Southern CaliforniaLos Angeles, CA, USA
| | - Sofia Grade
- Centre for Neuroscience and Cell Biology, University of Coimbra Coimbra, Portugal
| | - Sofia D Santos
- Glial Cell Biology Group, Institute for Molecular and Cell Biology - IBMCPorto, Portugal; Instituto de Investigação e Inovação em Saúde, Universidade do PortoPorto, Portugal
| | - Liliana Bernardino
- Centre for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal; Health Sciences Research Centre, Faculty of Health Sciences, University of Beira InteriorCovilhã, Portugal
| | - Thomas C Chen
- Department of Neurological Surgery, University of Southern California Los Angeles, CA, USA
| | - João Relvas
- Glial Cell Biology Group, Institute for Molecular and Cell Biology - IBMCPorto, Portugal; Instituto de Investigação e Inovação em Saúde, Universidade do PortoPorto, Portugal
| | - Florence M Hofman
- Department of Pathology, University of Southern California Los Angeles, CA, USA
| | - Fabienne Agasse
- Centre for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal; Department of Pathology, University of Southern CaliforniaLos Angeles, CA, USA; Department of Neurological Surgery, University of Southern CaliforniaLos Angeles, CA, USA
| |
Collapse
|
17
|
Bayin NS, Frenster JD, Kane JR, Rubenstein J, Modrek AS, Baitalmal R, Dolgalev I, Rudzenski K, Scarabottolo L, Crespi D, Redaelli L, Snuderl M, Golfinos JG, Doyle W, Pacione D, Parker EC, Chi AS, Heguy A, MacNeil DJ, Shohdy N, Zagzag D, Placantonakis DG. GPR133 (ADGRD1), an adhesion G-protein-coupled receptor, is necessary for glioblastoma growth. Oncogenesis 2016; 5:e263. [PMID: 27775701 PMCID: PMC5117849 DOI: 10.1038/oncsis.2016.63] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 08/09/2016] [Accepted: 08/24/2016] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is a deadly primary brain malignancy with extensive intratumoral hypoxia. Hypoxic regions of GBM contain stem-like cells and are associated with tumor growth and angiogenesis. The molecular mechanisms that regulate tumor growth in hypoxic conditions are incompletely understood. Here, we use primary human tumor biospecimens and cultures to identify GPR133 (ADGRD1), an orphan member of the adhesion family of G-protein-coupled receptors, as a critical regulator of the response to hypoxia and tumor growth in GBM. GPR133 is selectively expressed in CD133+ GBM stem cells (GSCs) and within the hypoxic areas of PPN in human biospecimens. GPR133 mRNA is transcriptionally upregulated by hypoxia in hypoxia-inducible factor 1α (Hif1α)-dependent manner. Genetic inhibition of GPR133 with short hairpin RNA reduces the prevalence of CD133+ GSCs, tumor cell proliferation and tumorsphere formation in vitro. Forskolin rescues the GPR133 knockdown phenotype, suggesting that GPR133 signaling is mediated by cAMP. Implantation of GBM cells with short hairpin RNA-mediated knockdown of GPR133 in the mouse brain markedly reduces tumor xenograft formation and increases host survival. Analysis of the TCGA data shows that GPR133 expression levels are inversely correlated with patient survival. These findings indicate that GPR133 is an important mediator of the hypoxic response in GBM and has significant protumorigenic functions. We propose that GPR133 represents a novel molecular target in GBM and possibly other malignancies where hypoxia is fundamental to pathogenesis.
Collapse
Affiliation(s)
- N S Bayin
- Department of Neurosurgery, New York University School of Medicine, New York, NY, USA
- Kimmel Center for Stem Cell Biology, New York University School of Medicine, New York, NY, USA
| | - J D Frenster
- Department of Neurosurgery, New York University School of Medicine, New York, NY, USA
- Kimmel Center for Stem Cell Biology, New York University School of Medicine, New York, NY, USA
| | - J R Kane
- Department of Neurosurgery, New York University School of Medicine, New York, NY, USA
| | - J Rubenstein
- Department of Neurosurgery, New York University School of Medicine, New York, NY, USA
| | - A S Modrek
- Department of Neurosurgery, New York University School of Medicine, New York, NY, USA
| | - R Baitalmal
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - I Dolgalev
- Genome Technology Center, New York University School of Medicine, New York, NY, USA
| | - K Rudzenski
- Office for Therapeutic Alliances, New York University School of Medicine, New York, NY, USA
| | | | | | | | - M Snuderl
- Department of Pathology, New York University School of Medicine, New York, NY, USA
- Brain Tumor Center, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - J G Golfinos
- Department of Neurosurgery, New York University School of Medicine, New York, NY, USA
- Brain Tumor Center, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - W Doyle
- Department of Neurosurgery, New York University School of Medicine, New York, NY, USA
| | - D Pacione
- Department of Neurosurgery, New York University School of Medicine, New York, NY, USA
| | - E C Parker
- Department of Neurosurgery, New York University School of Medicine, New York, NY, USA
| | - A S Chi
- Brain Tumor Center, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
- Department of Neurology, New York University School of Medicine, New York, NY, USA
| | - A Heguy
- Genome Technology Center, New York University School of Medicine, New York, NY, USA
| | - D J MacNeil
- Office for Therapeutic Alliances, New York University School of Medicine, New York, NY, USA
| | - N Shohdy
- Office for Therapeutic Alliances, New York University School of Medicine, New York, NY, USA
| | - D Zagzag
- Department of Neurosurgery, New York University School of Medicine, New York, NY, USA
- Department of Pathology, New York University School of Medicine, New York, NY, USA
- Brain Tumor Center, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - D G Placantonakis
- Department of Neurosurgery, New York University School of Medicine, New York, NY, USA
- Kimmel Center for Stem Cell Biology, New York University School of Medicine, New York, NY, USA
- Brain Tumor Center, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
18
|
Feinberg AP, Koldobskiy MA, Göndör A. Epigenetic modulators, modifiers and mediators in cancer aetiology and progression. Nat Rev Genet 2016; 17:284-99. [PMID: 26972587 DOI: 10.1038/nrg.2016.13] [Citation(s) in RCA: 598] [Impact Index Per Article: 74.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This year is the tenth anniversary of the publication in this journal of a model suggesting the existence of 'tumour progenitor genes'. These genes are epigenetically disrupted at the earliest stages of malignancies, even before mutations, and thus cause altered differentiation throughout tumour evolution. The past decade of discovery in cancer epigenetics has revealed a number of similarities between cancer genes and stem cell reprogramming genes, widespread mutations in epigenetic regulators, and the part played by chromatin structure in cellular plasticity in both development and cancer. In the light of these discoveries, we suggest here a framework for cancer epigenetics involving three types of genes: 'epigenetic mediators', corresponding to the tumour progenitor genes suggested earlier; 'epigenetic modifiers' of the mediators, which are frequently mutated in cancer; and 'epigenetic modulators' upstream of the modifiers, which are responsive to changes in the cellular environment and often linked to the nuclear architecture. We suggest that this classification is helpful in framing new diagnostic and therapeutic approaches to cancer.
Collapse
Affiliation(s)
- Andrew P Feinberg
- Center for Epigenetics, Johns Hopkins University School of Medicine, 855 N. Wolfe Street, Rangos 570, Baltimore, Maryland 21205, USA
| | - Michael A Koldobskiy
- Center for Epigenetics, Johns Hopkins University School of Medicine, 855 N. Wolfe Street, Rangos 570, Baltimore, Maryland 21205, USA
| | - Anita Göndör
- Department of Microbiology, Tumour and Cell Biology, Nobels väg 16, Karolinska Institutet, S-171 77 Stockholm, Sweden
| |
Collapse
|
19
|
Fessler E, Borovski T, Medema JP. Endothelial cells induce cancer stem cell features in differentiated glioblastoma cells via bFGF. Mol Cancer 2015; 14:157. [PMID: 26282129 PMCID: PMC4539660 DOI: 10.1186/s12943-015-0420-3] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 07/23/2015] [Indexed: 12/19/2022] Open
Abstract
Background Glioblastoma multiforme (GBM) is a rapidly growing malignant brain tumor, which has been reported to be organized in a hierarchical fashion with cancer stem cells (CSCs) at the apex. Recent studies demonstrate that this hierarchy does not follow a one-way route but can be reverted with more differentiated cells giving rise to cells possessing CSC features. We investigated the role of tumor microvascular endothelial cells (tMVECs) in reverting differentiated glioblastoma cells to CSC-like cells. Methods We made use of primary GBM lines and tMVECs. To ensure differentiation, CSC-enriched cultures were forced into differentiation using several stimuli and cultures consisting solely of differentiated cells were obtained by sorting on the oligodendrocyte marker O4. Reversion to the CSC state was assessed phenotypically by CSC marker expression and functionally by evaluating clonogenic and multilineage differentiation potential. Results Conditioned medium of tMVECs was able to replenish the CSC pool by phenotypically and functionally reverting differentiated GBM cells to the CSC state. Basic fibroblast growth factor (bFGF), secreted by tMVECs, recapitulated the effects of the conditioned medium in inducing re-expression of CSC markers and increasing neurosphere formation ability of differentiated GBM cells. Conclusions Our findings demonstrate that the CSC-based hierarchy displays a high level of plasticity showing that differentiated GBM cells can acquire CSC features when placed in the right environment. These results point to the need to intersect the elaborate network of tMVECs and GBM CSCs for efficient elimination of GBM CSCs. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0420-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Evelyn Fessler
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental Molecular Medicine (CEMM), Academic Medical Center (AMC), University of Amsterdam, Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Tijana Borovski
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental Molecular Medicine (CEMM), Academic Medical Center (AMC), University of Amsterdam, Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands. .,Present address: Department of Surgery, University Medical Center (UMC) Utrecht, Utrecht, The Netherlands.
| | - Jan Paul Medema
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental Molecular Medicine (CEMM), Academic Medical Center (AMC), University of Amsterdam, Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
20
|
Kim SJ, Lee HJ, Kim MS, Choi HJ, He J, Wu Q, Aldape K, Weinberg JS, Yung WKA, Conrad CA, Langley RR, Lehembre F, Regenass U, Fidler IJ. Macitentan, a Dual Endothelin Receptor Antagonist, in Combination with Temozolomide Leads to Glioblastoma Regression and Long-term Survival in Mice. Clin Cancer Res 2015; 21:4630-41. [PMID: 26106074 DOI: 10.1158/1078-0432.ccr-14-3195] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 05/30/2015] [Indexed: 12/22/2022]
Abstract
PURPOSE The objective of the study was to determine whether astrocytes and brain endothelial cells protect glioma cells from temozolomide through an endothelin-dependent signaling mechanism and to examine the therapeutic efficacy of the dual endothelin receptor antagonist, macitentan, in orthotopic models of human glioblastoma. EXPERIMENTAL DESIGN We evaluated several endothelin receptor antagonists for their ability to inhibit astrocyte- and brain endothelial cell-induced protection of glioma cells from temozolomide in chemoprotection assays. We compared survival in nude mice bearing orthotopically implanted LN-229 glioblastomas or temozolomide-resistant (LN-229(Res) and D54(Res)) glioblastomas that were treated with macitentan, temozolomide, or both. Tumor burden was monitored weekly with bioluminescence imaging. The effect of therapy on cell division, apoptosis, tumor-associated vasculature, and pathways associated with cell survival was assessed by immunofluorescent microscopy. RESULTS Only dual endothelin receptor antagonism abolished astrocyte- and brain endothelial cell-mediated protection of glioma cells from temozolomide. In five independent survival studies, including temozolomide-resistant glioblastomas, 46 of 48 (96%) mice treated with macitentan plus temozolomide had no evidence of disease (P < 0.0001), whereas all mice in other groups died. In another analysis, macitentan plus temozolomide therapy was stopped in 16 mice after other groups had died. Only 3 of 16 mice eventually developed recurrent disease, 2 of which responded to additional cycles of macitentan plus temozolomide. Macitentan downregulated proteins associated with cell division and survival in glioma cells and associated endothelial cells, which enhanced their sensitivity to temozolomide. CONCLUSIONS Macitentan plus temozolomide are well tolerated, produce durable responses, and warrant clinical evaluation in glioblastoma patients.
Collapse
Affiliation(s)
- Sun-Jin Kim
- Department of Cancer Biology, Metastasis Research Laboratory, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ho Jeong Lee
- Department of Cancer Biology, Metastasis Research Laboratory, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mark Seungwook Kim
- Department of Cancer Biology, Metastasis Research Laboratory, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hyun Jin Choi
- Department of Cancer Biology, Metastasis Research Laboratory, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Junqin He
- Department of Cancer Biology, Metastasis Research Laboratory, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Qiuyu Wu
- Department of Cancer Biology, Metastasis Research Laboratory, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kenneth Aldape
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeffrey S Weinberg
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - W K Alfred Yung
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Charles A Conrad
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert R Langley
- Department of Cancer Biology, Metastasis Research Laboratory, University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Urs Regenass
- Actelion Pharmaceuticals, Ltd., Allschwil, Switzerland
| | - Isaiah J Fidler
- Department of Cancer Biology, Metastasis Research Laboratory, University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
21
|
Islam F, Qiao B, Smith RA, Gopalan V, Lam AKY. Cancer stem cell: fundamental experimental pathological concepts and updates. Exp Mol Pathol 2015; 98:184-91. [PMID: 25659759 DOI: 10.1016/j.yexmp.2015.02.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 02/04/2015] [Indexed: 01/01/2023]
Abstract
Cancer stem cells (CSCs) are a subset of cancer cells which play a key role in predicting the biological aggressiveness of cancer due to its ability of self-renewal and multi-lineage differentiation (stemness). The CSC model is a dynamic one with a functional subpopulation of cancer cells rather than a stable cell population responsible for tumour regeneration. Hypotheses regarding the origins of CSCs include (1) malignant transformation of normal stem cells; (2) mature cancer cell de-differentiation with epithelial-mesenchymal transition and (3) induced pluripotent cancer cells. Surprisingly, the cancer stem cell hypothesis originated in the late nineteenth century and the existence of haematopoietic stem cells was demonstrated a century later, demonstrating that the concept was possible. In the last decade, CSCs have been identified and isolated in different cancers. The hallmark traits of CSCs include their heterogeneity, interaction with microenvironments and plasticity. Understanding these basic concepts of CSCs is important for translational applications using CSCs in the management of patients with cancer.
Collapse
Affiliation(s)
- Farhadul Islam
- Cancer Molecular Pathology, School of Medicine and Griffith Health Institute, Griffith University, Gold Coast, Queensland, Australia
| | - Bin Qiao
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Robert A Smith
- Cancer Molecular Pathology, School of Medicine and Griffith Health Institute, Griffith University, Gold Coast, Queensland, Australia; Genomics Research Centre, Institute of Health and Biomedical Innovation, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Vinod Gopalan
- Cancer Molecular Pathology, School of Medicine and Griffith Health Institute, Griffith University, Gold Coast, Queensland, Australia
| | - Alfred K-Y Lam
- Cancer Molecular Pathology, School of Medicine and Griffith Health Institute, Griffith University, Gold Coast, Queensland, Australia; Department of Pathology, Gold Coast University Hospital, Gold Coast, Queensland, Australia.
| |
Collapse
|
22
|
Cho DY, Lin SZ, Yang WK, Lee HC, Hsu DM, Lin HL, Chen CC, Liu CL, Lee WY, Ho LH. Targeting cancer stem cells for treatment of glioblastoma multiforme. Cell Transplant 2014; 22:731-9. [PMID: 23594862 DOI: 10.3727/096368912x655136] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cancer stem cells (CSCs) in glioblastoma multiforme (GBM) are radioresistant and chemoresistant, which eventually results in tumor recurrence. Targeting CSCs for treatment is the most crucial issue. There are five methods for targeting the CSCs of GBM. One is to develop a new chemotherapeutic agent specific to CSCs. A second is to use a radiosensitizer to enhance the radiotherapy effect on CSCs. A third is to use immune cells to attack the CSCs. In a fourth method, an agent is used to promote CSCs to differentiate into normal cells. Finally, ongoing gene therapy may be helpful. New therapeutic agents for targeting a signal pathway, such as epidermal growth factor (EGF) and vascular epidermal growth factor (VEGF) or protein kinase inhibitors, have been used for GBM but for CSCs the effects still require further evaluation. Nonsteroidal anti-inflammatory drugs (NSAIDs) such as cyclooxygenase-2 (Cox-2) inhibitors have proven to be effective for increasing radiation sensitivity of CSCs in culture. Autologous dendritic cells (DCs) are one of the promising immunotherapeutic agents in clinical trials and may provide another innovative method for eradication of CSCs. Bone-morphogenetic protein 4 (BMP4) is an agent used to induce CSCs to differentiate into normal glial cells. Research on gene therapy by viral vector is also being carried out in clinical trials. Targeting CSCs by eliminating the GBM tumor may provide an innovative way to reduce tumor recurrence by providing a synergistic effect with conventional treatment. The combination of conventional surgery, chemotherapy, and radiotherapy with stem cell-orientated therapy may provide a new promising treatment for reducing GBM recurrence and improving the survival rate.
Collapse
Affiliation(s)
- Der-Yang Cho
- Department of Neurosurgery, Neuropsychiatry Center, China Medical University Hospital, Taichung, Taiwan, ROC
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Glioblastoma multiforme therapy and mechanisms of resistance. Pharmaceuticals (Basel) 2013; 6:1475-506. [PMID: 24287492 PMCID: PMC3873674 DOI: 10.3390/ph6121475] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 11/04/2013] [Accepted: 11/12/2013] [Indexed: 12/26/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a grade IV brain tumor characterized by a heterogeneous population of cells that are highly infiltrative, angiogenic and resistant to chemotherapy. The current standard of care, comprised of surgical resection followed by radiation and the chemotherapeutic agent temozolomide, only provides patients with a 12–14 month survival period post-diagnosis. Long-term survival for GBM patients remains uncommon as cells with intrinsic or acquired resistance to treatment repopulate the tumor. In this review we will describe the mechanisms of resistance, and how they may be overcome to improve the survival of GBM patients by implementing novel chemotherapy drugs, new drug combinations and new approaches relating to DNA damage, angiogenesis and autophagy.
Collapse
|
24
|
Molecular imaging in the development of a novel treatment paradigm for glioblastoma (GBM): an integrated multidisciplinary commentary. Drug Discov Today 2013; 18:1052-66. [DOI: 10.1016/j.drudis.2013.06.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 06/03/2013] [Accepted: 06/11/2013] [Indexed: 12/29/2022]
|
25
|
Abstract
Cancer stem cells (CSCs) have been proposed as the driving force of tumorigenesis and the seeds of metastases. However, their existence and role remain a topic of intense debate. Recently, the identification of CSCs in endogenously developing mouse tumours has provided further support for this concept. Here I discuss the challenges in identifying CSCs, their dependency on a supportive niche and their role in metastasis, and propose that stemness is a flexible — rather than fixed — quality of tumour cells that can be lost and gained.
Collapse
Affiliation(s)
- Jan Paul Medema
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental Molecular Medicine, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| |
Collapse
|
26
|
Cancer stem cell dynamics in tumor progression and metastasis: is the microenvironment to blame? Cancer Lett 2012; 341:97-104. [PMID: 23089245 DOI: 10.1016/j.canlet.2012.10.015] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 10/02/2012] [Accepted: 10/13/2012] [Indexed: 12/13/2022]
Abstract
Stem cells are defined by their self-renewal capacity and the ability to give rise to all differentiated progeny necessary for one specific organ. These two characteristics are also inherent in cancer stem cells (CSCs), which are thought to be the only subpopulation within a tumor endowed with tumorigenic potential. CSCs combine many features that render cancer one of the leading causes of death in the Western world: metastasis, tumor recurrence, and therapy refractoriness. Strikingly, CSCs are not a fixed entity, but differentiated tumor cells are able to revert to a stem-like state. Thus, CSCs are not only intrinsically programmed to fulfill their detrimental roles, but are orchestrated by stromal cells residing in their vicinity and forming the CSC niche. Yet, this relationship is not a one-way road: CSCs are able to manipulate stromal cells to their needs, not only in the primary tumor, but also in distant organs and thus prime the foreign soil for their arrival by inducing a premetastatic niche. The suggested plasticity between the differentiation states of cancer cells and the regulation by microenvironmental cues provides new starting-points for novel cancer therapies.
Collapse
|
27
|
Angiogenic signalling pathways altered in gliomas: selection mechanisms for more aggressive neoplastic subpopulations with invasive phenotype. JOURNAL OF SIGNAL TRANSDUCTION 2012; 2012:597915. [PMID: 22852079 PMCID: PMC3407647 DOI: 10.1155/2012/597915] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 05/22/2012] [Indexed: 12/18/2022]
Abstract
The angiogenesis process is a key event for glioma survival, malignancy and growth. The start of angiogenesis is mediated by a cascade of intratumoural events: alteration of the microvasculature network; a hypoxic microenvironment; adaptation of neoplastic cells and synthesis of pro-angiogenic factors. Due to a chaotic blood flow, a consequence of an aberrant microvasculature, tissue hypoxia phenomena are induced. Hypoxia inducible factor 1 is a major regulator in glioma invasiveness and angiogenesis. Clones of neoplastic cells with stem cell characteristics are selected by HIF-1. These cells, called "glioma stem cells" induce the synthesis of vascular endothelial growth factor. This factor is a pivotal mediator of angiogenesis. To elucidate the role of these angiogenic mediators during glioma growth, we have used a rat endogenous glioma model. Gliomas induced by prenatal ENU administration allowed us to study angiogenic events from early to advanced tumour stages. Events such as microvascular aberrations, hypoxia, GSC selection and VEGF synthesis may be studied in depth. Our data showed that for the treatment of gliomas, developing anti-angiogenic therapies could be aimed at GSCs, HIF-1 or VEGF. The ENU-glioma model can be considered to be a useful option to check novel designs of these treatment strategies.
Collapse
|
28
|
Choi SA, Wang KC, Phi JH, Lee JY, Park CK, Park SH, Kim SK. A distinct subpopulation within CD133 positive brain tumor cells shares characteristics with endothelial progenitor cells. Cancer Lett 2012; 324:221-30. [PMID: 22652175 DOI: 10.1016/j.canlet.2012.05.026] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 05/02/2012] [Accepted: 05/22/2012] [Indexed: 01/07/2023]
Abstract
The cell surface marker CD133 has been proposed as a brain tumor stem cell marker. However, there have been substantial controversies regarding the necessity and role of CD133 in tumorigenesis. This study aimed to characterize CD133(+) cells in brain tumors. Human brain tumor specimens and whole blood were collected from the same patients (N=12). We carried out dual FACS staining for CD133/CD34 and functional tumorigenesis and angiogenesis analyses of CD133(+) cells from different origins. We also investigated the in vivo tumorigenic potential and histological characteristics of four distinct groups on the basis of expression of CD133/CD34 markers (CD133(+), CD133(+)/CD34(+), CD133(+)/CD34(-), and CD133(-)). CD133(+) brain tumor cells coexpressed significantly higher positivity for CD34 (70.7±5.2% in CD133(+) vs. 12.3±4.2% in CD133(-) cells, P<0.001). CD133(+) brain tumor cells formed neurosphere-like spheroids and differentiated into multiple nervous system lineages unlike CD133(+) blood cells. They showed biological characteristics of endothelial cells, including vWF expression, LDL uptake and tube formation in vitro, unlike CD133(-) brain tumors cells. Pathologic analysis of brains implanted with CD133(+) cells showed large, markedly hypervascular tumors with well-demarcated boundary. CD133(+)/CD34(-) cells produced smaller but highly infiltrative tumors. Notably, pure angiogenic cell fractions (CD133(+)/CD34(+)) and CD133(-) tumor cells did not generate tumors in vivo. Our data suggest the presence of a distinct subpopulation of CD133(+) cells isolated from human brain tumors, with characteristics of endothelial progenitor cells (EPCs).
Collapse
Affiliation(s)
- Seung Ah Choi
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
29
|
Therapy-resistant tumor microvascular endothelial cells contribute to treatment failure in glioblastoma multiforme. Oncogene 2012; 32:1539-48. [DOI: 10.1038/onc.2012.172] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
30
|
Rao S, Sengupta R, Choe EJ, Woerner BM, Jackson E, Sun T, Leonard J, Piwnica-Worms D, Rubin JB. CXCL12 mediates trophic interactions between endothelial and tumor cells in glioblastoma. PLoS One 2012; 7:e33005. [PMID: 22427929 PMCID: PMC3299723 DOI: 10.1371/journal.pone.0033005] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 02/08/2012] [Indexed: 12/11/2022] Open
Abstract
Emerging evidence suggests endothelial cells (EC) play a critical role in promoting Glioblastoma multiforme (GBM) cell proliferation and resistance to therapy. The molecular basis for GBM-EC interactions is incompletely understood. We hypothesized that the chemokine CXCL12 and its receptor CXCR4 could mediate direct interactions between GBM cells and tumor-associated endothelial cells and that disruption of this interaction might be the molecular basis for the anti-tumor effects of CXCR4 antagonists. We investigated this possibility in vivo and in an in vitro co-culture model that incorporated extracellular matrix, primary human brain microvascular ECs (HBMECs) and either an established GBM cell line or primary GBM specimens. Depletion of CXCR4 in U87 GBM cells blocked their growth as intracranial xenografts indicating that tumor cell CXCR4 is required for tumor growth in vivo. In vitro, co-culture of either U87 cells or primary GBM cells with HBMECs resulted in their co-localization and enhanced GBM cell growth. Genetic manipulation of CXCL12 expression and pharmacological inhibition of its receptors CXCR4 and CXCR7 revealed that the localizing and trophic effects of endothelial cells on GBM cells were dependent upon CXCL12 and CXCR4. These findings indicate that the CXCL12/CXCR4 pathway directly mediates endothelial cell trophic function in GBMs and that inhibition of CXCL12-CXCR4 signaling may uniquely target this activity. Therapeutic disruption of endothelial cell trophic functions could complement the structural disruption of anti-angiogenic regimens and, in combination, might also improve the efficacy of radiation and chemotherapy in treating GBMs.
Collapse
Affiliation(s)
- Shyam Rao
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Rajarshi Sengupta
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Eun Joo Choe
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - B. Mark Woerner
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Erin Jackson
- Bridging Research with Imaging, Genomics and High-Throughput Institute, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Tao Sun
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Jeffrey Leonard
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - David Piwnica-Worms
- Bridging Research with Imaging, Genomics and High-Throughput Institute, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Joshua B. Rubin
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
31
|
Woerner BM, Luo J, Brown KR, Jackson E, Dahiya SM, Mischel P, Benovic JL, Piwnica-Worms D, Rubin JB. Suppression of G-protein-coupled receptor kinase 3 expression is a feature of classical GBM that is required for maximal growth. Mol Cancer Res 2011; 10:156-66. [PMID: 22086906 DOI: 10.1158/1541-7786.mcr-11-0411] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
G-protein-coupled receptor kinases (GRK) regulate the function of G-protein-coupled receptors (GPCR). Previously, we found that GPCR (CXCR4)-mediated astrocytoma growth was dependent upon abnormally sustained CXCR4 signaling and was correlated with decreased GRK-mediated receptor phosphorylation. As CXCR4 has also been implicated in the stimulation of high-grade glioma growth, we sought to determine whether dysregulation of GRK expression and/or function might also be present in high-grade gliomas. In an analysis of data from The Cancer Genome Atlas, we found that GRK3 expression is frequently decreased in glioblastoma (GBM) of the classical subtype, which possesses signature amplification or mutational activation of the epidermal growth factor (EGF) receptor. We tested the correlation between GRK3 expression and GBM subtypes, as well as the relationship between the activation of the EGF and other growth factor receptor pathways and GRK expression. In analyses of primary GBM tissue and RNA specimens, we found that GRK3 expression is correlated with established criteria for GBM subtyping including expression of EGF receptor, platelet-derived growth factor receptor (PDGFR)α, NF1, PTEN, CDKN2A, and neurofilament. We also found that established drivers of gliomagenesis, the EGF, PDGF, and TGF-β pathways, all regulate GRK expression. Coculture experiments, designed to mimic critical interactions between tumor and brain microvascular endothelial cells, showed that specifically increasing GRK3 expression reduced the trophic effect of endothelial cells on tumor cells. Together, these experiments show that GRK3 is a negative regulator of cell growth whose expression is preferentially reduced in GBM of the classical subtype as a consequence of activity in primary gliomagenic pathways.
Collapse
Affiliation(s)
- B Mark Woerner
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Salsman VS, Chow KKH, Shaffer DR, Kadikoy H, Li XN, Gerken C, Perlaky L, Metelitsa LS, Gao X, Bhattacharjee M, Hirschi K, Heslop HE, Gottschalk S, Ahmed N. Crosstalk between medulloblastoma cells and endothelium triggers a strong chemotactic signal recruiting T lymphocytes to the tumor microenvironment. PLoS One 2011; 6:e20267. [PMID: 21647415 PMCID: PMC3103535 DOI: 10.1371/journal.pone.0020267] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 04/16/2011] [Indexed: 12/22/2022] Open
Abstract
Cancer cells can live and grow if they succeed in creating a favorable niche that often includes elements from the immune system. While T lymphocytes play an important role in the host response to tumor growth, the mechanism of their trafficking to the tumor remains poorly understood. We show here that T lymphocytes consistently infiltrate the primary brain cancer, medulloblastoma. We demonstrate, both in vitro and in vivo, that these T lymphocytes are attracted to tumor deposits only after the tumor cells have interacted with tumor vascular endothelium. Macrophage Migration Inhibitory Factor (MIF)" is the key chemokine molecule secreted by tumor cells which induces the tumor vascular endothelial cells to secrete the potent T lymphocyte attractant "Regulated upon Activation, Normal T-cell Expressed, and Secreted (RANTES)." This in turn creates a chemotactic gradient for RANTES-receptor bearing T lymphocytes. Manipulation of this pathway could have important therapeutic implications.
Collapse
Affiliation(s)
- Vita S. Salsman
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Kevin K. H. Chow
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Donald R. Shaffer
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Huseyin Kadikoy
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Xiao-Nan Li
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Claudia Gerken
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Laszlo Perlaky
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Leonid S. Metelitsa
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Xiuhua Gao
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Meena Bhattacharjee
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Karen Hirschi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Helen E. Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Stephen Gottschalk
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Nabil Ahmed
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
33
|
Tafani M, Di Vito M, Frati A, Pellegrini L, De Santis E, Sette G, Eramo A, Sale P, Mari E, Santoro A, Raco A, Salvati M, De Maria R, Russo MA. Pro-inflammatory gene expression in solid glioblastoma microenvironment and in hypoxic stem cells from human glioblastoma. J Neuroinflammation 2011; 8:32. [PMID: 21489226 PMCID: PMC3098164 DOI: 10.1186/1742-2094-8-32] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Accepted: 04/13/2011] [Indexed: 01/14/2023] Open
Abstract
Background Adaptation to hypoxia and consequent pro-inflammatory gene expression of prostate and breast carcinomas have been implicated in the progression toward cancer malignant phenotype. Only partial data are available for the human tumor glioblastoma multiforme (GBM). The aim of our study was to analyze the hypoxic and pro-inflammatory microenvironment in GBMs and to demonstrate that in a stem/progenitor cell line derived from human glioblastoma (GBM-SCs), hypoxia activates a coordinated inflammatory response, evidencing an invasive and migratory phenotype. Methods From each of 10 human solid glioblastomas, clinically and histopathologically characterized, we obtained three surgical samples taken from the center and the periphery of the tumor, and from adjacent host normal tissue. Molecular and morphological analyses were carried out using quantitative real-time PCR and western blot (WB). GBM stem and differentiated cells were incubated under hypoxic conditions and analyzed for pro-inflammatory gene expression and for invasive/migratory behavior. Results A panel of selected representative pro-inflammatory genes (RAGE and P2X7R, COX2, NOS2 and, PTX3) were analyzed, comparing tumor, peritumor and host normal tissues. Tumors containing leukocyte infiltrates (as assessed using CD45 immunohistochemistry) were excluded. Selected genes were overexpressed in the central regions of the tumors (i.e. in the more hypoxic areas), less expressed in peripheral regions, and poorly expressed or absent in adjacent normal host tissues. Western blot analysis confirmed that the corresponding pro-inflammatory proteins were also differently expressed. Hypoxic stem cell lines showed a clear time-dependent activation of the entire panel of pro-inflammatory genes as compared to differentiated tumor cells. Biological assays showed that invasive and migratory behavior was strengthened by hypoxia only in GBM stem cells. Conclusions In human solid glioblastoma we have observed a coordinated overexpression of a panel of pro-inflammatory genes as compared to host normal tissue. We have also evidenced a similar pattern of overexpressed genes in GBM-SCs after hypoxic treatment, showing also a gain of invasive and migratory function that was lost when these stem cells differentiated. We suggest that, as has been previously described for prostatic and mammary carcinoma, in human glioblastoma acquisition of a proinflammatory phenotype may be relevant for malignant progression.
Collapse
Affiliation(s)
- Marco Tafani
- Department of Experimental Medicine, Sapienza University of Rome, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Christensen K, Schrøder HD, Kristensen BW. CD133+ niches and single cells in glioblastoma have different phenotypes. J Neurooncol 2010; 104:129-43. [PMID: 21184132 DOI: 10.1007/s11060-010-0488-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Accepted: 12/02/2010] [Indexed: 12/19/2022]
Abstract
Putative CD133(+) brain tumor stem cells have been shown to be located in niches and as single cells. This is the first study providing insight into the different phenotypes of CD133(+) cells in glioblastoma according to localization. Paraffin sections were stained by double immunofluorescence with CD133 and the candidate stem cell markers Sox2, Bmi-1, EGFR, podoplanin and nestin, the proliferation marker Ki67 and the endothelial cell markers CD31, CD34, and VWF. Cell counting showed that the CD133(+) cells in the niches had a significantly higher expression of Sox2, EGFR and nestin compared to CD133(+) single cells, but only a 3% Ki67 labeling index versus 14% found for CD133(+) single cells. Only low endothelial cell marker expression was found in the niches or the CD133(-) tumor areas, while 43% CD133(+)/CD31(+) and 25% CD133(+)/CD34(+) single cells were found. CD133(+) blood vessels within CD133(+) niches were less proliferative and more often Bmi-1(+) than CD133(+) blood vessels outside niches. In conclusion, different CD133(+) cell phenotypes exist according to the in situ localization, and also the phenotype of CD133(+) blood vessels vary according to the localization. CD133(+) niches contain stem-like cells with a lower proliferation index than CD133(+) single cells, which have an endothelial differentiation profile suggesting a role in angiogenesis.
Collapse
Affiliation(s)
- Karina Christensen
- Department of Pathology, Odense University Hospital, Winsløwparken 15, 5000 Odense C, Denmark
| | | | | |
Collapse
|
35
|
Dirks PB. Brain tumor stem cells: the cancer stem cell hypothesis writ large. Mol Oncol 2010; 4:420-30. [PMID: 20801091 DOI: 10.1016/j.molonc.2010.08.001] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Revised: 08/03/2010] [Accepted: 08/03/2010] [Indexed: 01/06/2023] Open
Abstract
Brain tumors, which are typically very heterogeneous at the cellular level, appear to have a stem cell foundation. Recently, investigations from multiple groups have found that human as well as experimental mouse brain tumors contain subpopulations of cells that functionally behave as tumor stem cells, driving tumor growth and generating tumor cell progeny that form the tumor bulk, but which then lose tumorigenic ability. In human glioblastomas, these tumor stem cells express neural precursor markers and are capable of differentiating into tumor cells that express more mature neural lineage markers. In addition, modeling brain tumors in mice suggests that neural precursor cells more readily give rise to full blown tumors, narrowing potential cells of origin to those rarer brain cells that have a proliferative potential. Applying stem cell concepts and methodologies is giving fresh insight into brain tumor biology, cell of origin and mechanisms of growth, and is offering new opportunities for development of more effective treatments. The field of brain tumor stem cells remains very young and there is much to be learned before these new insights are translated into new patient treatments.
Collapse
Affiliation(s)
- Peter B Dirks
- Division of Neurosurgery, Developmental and Stem Cell Biology Program, Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Ontario, Canada.
| |
Collapse
|
36
|
Christensen K, Aaberg-Jessen C, Andersen C, Goplen D, Bjerkvig R, Kristensen BW. Immunohistochemical expression of stem cell, endothelial cell, and chemosensitivity markers in primary glioma spheroids cultured in serum-containing and serum-free medium. Neurosurgery 2010; 66:933-47. [PMID: 20404698 DOI: 10.1227/01.neu.0000368393.45935.46] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE To investigate the influence of serum-free medium (SFM) supplemented with epidermal growth factor and basic fibroblast growth factor compared with conventional serum-containing medium (SCM) on the phenotype of organotypic primary spheroids from seven gliomas. METHODS Paraffin sections of the original surgical specimens, primary glioma spheroids, and U87 derived spheroids were stained immunohistochemically with the stem cell markers CD133, podoplanin, Sox2, Bmi-1, and nestin; the endothelial cell markers CD31, CD34, and Von Willebrand Factor (VWF); the chemosensitivity markers P-glycoprotein and tissue inhibitor of metalloproteinases-1 (TIMP-1); and glial fibrillary acidic protein, neural cell adhesion molecule CD56, and the proliferation marker Ki67. RESULTS Scoring of the immunohistochemical stainings showed that the expression of CD133 and all other markers included was preserved in primary spheroids, confirming the in vivo-like nature of these spheroids. Spheroids in SFM better mimicked the in vivo phenotype with significantly more CD133, CD34, VWF, P-glycoprotein, TIMP-1, and Ki67 compared with SCM. CONCLUSION In this first study of the influence of SFM on primary glioma spheroids, the conditions favored an in vivo-like phenotype with increased expression of CD133. More vascular structures were found in SFM, suggesting that the close relationship between blood vessels and tumor stem-like cells was better preserved in this medium.
Collapse
Affiliation(s)
- Karina Christensen
- Department of Pathology, Odense University Hospital, 5000 Odense, Denmark
| | | | | | | | | | | |
Collapse
|
37
|
Takahashi M, Shimajiri S, Izumi H, Hirano G, Kashiwagi E, Yasuniwa Y, Wu Y, Han B, Akiyama M, Nishizawa S, Sasaguri Y, Kohno K. Y-box binding protein-1 is a novel molecular target for tumor vessels. Cancer Sci 2010; 101:1367-73. [PMID: 20398058 PMCID: PMC11159253 DOI: 10.1111/j.1349-7006.2010.01534.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2009] [Revised: 02/01/2010] [Accepted: 02/04/2010] [Indexed: 11/28/2022] Open
Abstract
Y-box binding protein-1 (YB-1) is a member of the cold shock protein family and functions in transcription and translation. Many reports indicate that YB-1 is highly expressed in tumor cells and is a marker for tumor aggressiveness and clinical prognosis. Here, we show clear evidence that YB-1 is expressed in the angiogenic endothelial cells of various tumors, such as glioblastoma, esophageal cancer, gastric cancer, colon cancer, and lung cancer, as well as in tumor cells. YB-1 was highly expressed in glomeruloid microvascular endothelial cells of brain tumors and microvessels in the desmoplastic region around multiple solid tumors. On the other hand, no or low YB-1 expression was observed in normal angiogenic endothelial cells from fetal kidney, newborn lung, and placenta. The endothelial cells in inflammatory regions of granulomas were also weakly labeled. Knockdown of YB-1 expression by small-interfering RNA induced G1 cell cycle arrest and inhibited the growth of human umbilical vein endothelial cells stimulated by growth factors. Taken together, YB-1 plays an important role in the growth of not only tumor cells but also tumor-associated endothelial cells, suggesting that YB-1 is a promising target for cancer therapy.
Collapse
Affiliation(s)
- Mayu Takahashi
- Department of Molecular Biology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Vermeulen L, De Sousa E Melo F, van der Heijden M, Cameron K, de Jong JH, Borovski T, Tuynman JB, Todaro M, Merz C, Rodermond H, Sprick MR, Kemper K, Richel DJ, Stassi G, Medema JP. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat Cell Biol 2010; 12:468-76. [DOI: 10.1038/ncb2048] [Citation(s) in RCA: 1412] [Impact Index Per Article: 100.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Accepted: 03/26/2010] [Indexed: 11/09/2022]
|
39
|
Mimeault M, Batra SK. New promising drug targets in cancer- and metastasis-initiating cells. Drug Discov Today 2010; 15:354-64. [PMID: 20338259 DOI: 10.1016/j.drudis.2010.03.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2009] [Revised: 02/03/2010] [Accepted: 03/17/2010] [Indexed: 02/08/2023]
Abstract
The unique properties of cancer- and metastasis-initiating cells endowed with a high self-renewal and aberrant differentiation potential (including their elevated expression levels of anti-apoptotic factors, multidrug transporters, and DNA repair and detoxifying enzymes) might be associated with their resistance to current clinical cancer therapies and disease recurrence. The eradication of cancer- and metastasis-initiating cells by molecular targeting of distinct deregulated signaling elements that might contribute to their sustained growth, survival, and treatment resistance, therefore, is of immense therapeutic interest. These novel targeted approaches should improve the efficacy of current therapeutic treatments against highly aggressive, metastatic, recurrent, and lethal cancers.
Collapse
Affiliation(s)
- Murielle Mimeault
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA.
| | | |
Collapse
|
40
|
Sottoriva A, Verhoeff JJC, Borovski T, McWeeney SK, Naumov L, Medema JP, Sloot PMA, Vermeulen L. Cancer stem cell tumor model reveals invasive morphology and increased phenotypical heterogeneity. Cancer Res 2010; 70:46-56. [PMID: 20048071 DOI: 10.1158/0008-5472.can-09-3663] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The recently developed concept of cancer stem cells (CSC) sheds new light on various aspects of tumor growth and progression. Here, we present a mathematical model of malignancies to investigate how a hierarchical organized cancer cell population affects the fundamental properties of solid malignancies. We establish that tumors modeled in a CSC context more faithfully resemble human malignancies and show invasive behavior, whereas tumors without a CSC hierarchy do not. These findings are corroborated by in vitro studies. In addition, we provide evidence that the CSC model is accompanied by highly altered evolutionary dynamics compared with the ones predicted to exist in a stochastic, nonhierarchical tumor model. Our main findings indicate that the CSC model allows for significantly higher tumor heterogeneity, which may affect therapy resistance. Moreover, we show that therapy which fails to target the CSC population is not only unsuccessful in curing the patient, but also promotes malignant features in the recurring tumor. These include rapid expansion, increased invasion, and enhanced heterogeneity.
Collapse
Affiliation(s)
- Andrea Sottoriva
- Computational Science, Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| | | | | | | | | | | | | | | |
Collapse
|