101
|
The Crosstalk between Ovarian Cancer Stem Cell Niche and the Tumor Microenvironment. Stem Cells Int 2017; 2017:5263974. [PMID: 28819364 PMCID: PMC5551518 DOI: 10.1155/2017/5263974] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 07/03/2017] [Indexed: 02/07/2023] Open
Abstract
Ovarian cancer is one of the most important causes of cancer-related death among women in the world. Despite advances in ovarian cancer treatment, 70–80% of women who initially respond to therapy eventually relapse and die. There is evidence that a small population of cells within the tumors called cancer stem cells (CSCs) could be responsible for treatment failure due to their enhanced chemoresistance and tumorigenicity. These cells reside in a niche that maintains the principal properties of CSCs. These properties are associated with the capacity of CSCs to interact with different cells of the tumor microenvironment including mesenchymal stem cells, endothelial cells, immune cells, and fibroblasts, promoting cancer progression. This interaction can be mediated by cytokines, growth factors, lipids, and/or extracellular vesicles released in the CSC niche. In this review, we will discuss how the interaction between ovarian CSCs and the tumor microenvironment can contribute to the maintenance of the CSC niche and consequently to tumor progression in ovarian cancer.
Collapse
|
102
|
Abstract
Converging evidence from numerous laboratories has revealed that malignant brain cancers are complex ecological systems composed of distinct cellular and acellular elements that collectively dictate glioblastoma biology. Our understanding of the individual contributions of each of these components is vital to the design of effective therapies against these cancers. In this issue of Genes & Development, Zanca and colleagues (pp. 1212-1227) demonstrate that one subpopulation of glioblastoma cells expressing a mutant epidermal growth factor receptor (EGFRvIII) is responsible for the survival of non-EGFRvIII-expressing tumor cells as well as for evading molecularly targeted therapy.
Collapse
Affiliation(s)
- Ran Chen
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Yuan Pan
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|
103
|
Peitzsch C, Tyutyunnykova A, Pantel K, Dubrovska A. Cancer stem cells: The root of tumor recurrence and metastases. Semin Cancer Biol 2017; 44:10-24. [DOI: 10.1016/j.semcancer.2017.02.011] [Citation(s) in RCA: 191] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/26/2017] [Accepted: 02/27/2017] [Indexed: 12/11/2022]
|
104
|
Yan H, Romero-López M, Benitez LI, Di K, Frieboes HB, Hughes CCW, Bota DA, Lowengrub JS. 3D Mathematical Modeling of Glioblastoma Suggests That Transdifferentiated Vascular Endothelial Cells Mediate Resistance to Current Standard-of-Care Therapy. Cancer Res 2017; 77:4171-4184. [PMID: 28536277 DOI: 10.1158/0008-5472.can-16-3094] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/24/2017] [Accepted: 05/16/2017] [Indexed: 01/17/2023]
Abstract
Glioblastoma (GBM), the most aggressive brain tumor in human patients, is decidedly heterogeneous and highly vascularized. Glioma stem/initiating cells (GSC) are found to play a crucial role by increasing cancer aggressiveness and promoting resistance to therapy. Recently, cross-talk between GSC and vascular endothelial cells has been shown to significantly promote GSC self-renewal and tumor progression. Furthermore, GSC also transdifferentiate into bona fide vascular endothelial cells (GEC), which inherit mutations present in GSC and are resistant to traditional antiangiogenic therapies. Here we use three-dimensional mathematical modeling to investigate GBM progression and response to therapy. The model predicted that GSCs drive invasive fingering and that GEC spontaneously form a network within the hypoxic core, consistent with published experimental findings. Standard-of-care treatments using DNA-targeted therapy (radiation/chemo) together with antiangiogenic therapies reduced GBM tumor size but increased invasiveness. Anti-GEC treatments blocked the GEC support of GSCs and reduced tumor size but led to increased invasiveness. Anti-GSC therapies that promote differentiation or disturb the stem cell niche effectively reduced tumor invasiveness and size, but were ultimately limited in reducing tumor size because GECs maintain GSCs. Our study suggests that a combinatorial regimen targeting the vasculature, GSCs, and GECs, using drugs already approved by the FDA, can reduce both tumor size and invasiveness and could lead to tumor eradication. Cancer Res; 77(15); 4171-84. ©2017 AACR.
Collapse
Affiliation(s)
- Huaming Yan
- Department of Mathematics, University of California, Irvine, California
| | - Mónica Romero-López
- Department of Biomedical Engineering, University of California, Irvine, California
| | - Lesly I Benitez
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California
| | - Kaijun Di
- Chao Comprehensive Cancer Center, University of California, Irvine, California.,Department of Neurological Surgery, University of California, Irvine, California
| | - Hermann B Frieboes
- James Graham Brown Cancer Center, University of Louisville.,Department of Bioengineering, University of Louisville, Louisville, Kentucky
| | - Christopher C W Hughes
- Department of Biomedical Engineering, University of California, Irvine, California.,Department of Molecular Biology and Biochemistry, University of California, Irvine, California.,Chao Comprehensive Cancer Center, University of California, Irvine, California.,Center for Complex Biological Systems, University of California, Irvine, California
| | - Daniela A Bota
- Chao Comprehensive Cancer Center, University of California, Irvine, California.,Department of Neurological Surgery, University of California, Irvine, California.,Department of Neurology, University of California, Irvine, California
| | - John S Lowengrub
- Department of Mathematics, University of California, Irvine, California. .,Department of Biomedical Engineering, University of California, Irvine, California.,Chao Comprehensive Cancer Center, University of California, Irvine, California.,Center for Complex Biological Systems, University of California, Irvine, California
| |
Collapse
|
105
|
Kast RE, Hill QA, Wion D, Mellstedt H, Focosi D, Karpel-Massler G, Heiland T, Halatsch ME. Glioblastoma-synthesized G-CSF and GM-CSF contribute to growth and immunosuppression: Potential therapeutic benefit from dapsone, fenofibrate, and ribavirin. Tumour Biol 2017; 39:1010428317699797. [PMID: 28459367 DOI: 10.1177/1010428317699797] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Increased ratio of circulating neutrophils to lymphocytes is a common finding in glioblastoma and other cancers. Data reviewed establish that any damage to brain tissue tends to cause an increase in G-CSF and/or GM-CSF (G(M)-CSF) synthesized by the brain. Glioblastoma cells themselves also synthesize G(M)-CSF. G(M)-CSF synthesized by brain due to damage by a growing tumor and by the tumor itself stimulates bone marrow to shift hematopoiesis toward granulocytic lineages away from lymphocytic lineages. This shift is immunosuppressive and generates the relative lymphopenia characteristic of glioblastoma. Any trauma to brain-be it blunt, sharp, ischemic, infectious, cytotoxic, tumor encroachment, or radiation-increases brain synthesis of G(M)-CSF. G(M)-CSF are growth and motility enhancing factors for glioblastomas. High levels of G(M)-CSF contribute to the characteristic neutrophilia and lymphopenia of glioblastoma. Hematopoietic bone marrow becomes entrained with, directed by, and contributes to glioblastoma pathology. The antibiotic dapsone, the lipid-lowering agent fenofibrate, and the antiviral drug ribavirin are Food and Drug Administration- and European Medicines Agency-approved medicines that have potential to lower synthesis or effects of G(M)-CSF and thus deprive a glioblastoma of some of the growth promoting contributions of bone marrow and G(M)-CSF.
Collapse
Affiliation(s)
| | - Quentin A Hill
- 2 Department of Haematology, St James's University Hospital, Leeds Teaching Hospitals, Leeds, UK
| | - Didier Wion
- 3 INSERM U1205, Centre de Recherche Biomédicale Edmond J. Safra, Grenoble, France
| | - Håkan Mellstedt
- 4 Department of Oncology, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Daniele Focosi
- 5 North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | | | - Tim Heiland
- 6 Department of Neurosurgery, University of Ulm, Ulm, Germany
| | | |
Collapse
|
106
|
Sharma I, Singh A, Sharma KC, Saxena S. Gene Expression Profiling of Chemokines and Their Receptors in Low and High Grade Astrocytoma. Asian Pac J Cancer Prev 2017; 18:1307-1313. [PMID: 28610419 PMCID: PMC5555540 DOI: 10.22034/apjcp.2017.18.5.1307] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background: Despite intense interest in molecular characterization and searches for novel therapeutic targets, the
glioblastoma remains a formidable clinical challenge. Among many contributors to gliomagenesis, chemokines have
drawn special attention due to their involvement in a plethora of biological processes and pathological conditions. In
the present study we aimed to elucidate any pro-gliomagenic chemokine axis and probable roles in development of
glioblastoma multiforme (GBM). Method: An array of 84 chemokines, chemokine receptors and related genes were
studied by real time PCR with comparison between low grade astrocytoma (diffuse astrocytoma – grade II) and high
grade astrocytoma (glioblastoma multiforme – grade IV). Gene ontology analysis and database mining were performed
to funnel down the important axis in GBM followed by validation at the protein level by immunohistochemistry on tissue
microarrays. Results: Gene expression and gene ontology analysis identified CXCL8 as an important chemokine which
was more frequently up-regulated in GBM as compared to diffuse astrocytoma. Further we demonstrated localization
of CXCL8 and its receptors in glioblastoma possibly affecting autocrine and paracrine signalling that promotes tumor
cell proliferation and neovascularisation with vascular mimicry. Conclusion: From these results CXCL8 appears to be
an important gliomagenic chemokine which may be involved in GBM growth by promoting tumor cell proliferation
and neovascularization via vascular mimicry. Further in vitro and in vivo investigations are required to explore its
potential candidature in anti-GBM therapy.
Collapse
Affiliation(s)
- Ira Sharma
- National Institute of Pathology, New Delhi, India.,Symbiosis International University, Pune, India.
| | | | | | | |
Collapse
|
107
|
Lei KF, Chang CH, Chen MJ. Paper/PMMA Hybrid 3D Cell Culture Microfluidic Platform for the Study of Cellular Crosstalk. ACS APPLIED MATERIALS & INTERFACES 2017; 9:13092-13101. [PMID: 28353331 DOI: 10.1021/acsami.7b03021] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Studying cellular crosstalk is important for understanding tumor initiation, progression, metastasis, and therapeutic resistance. Moreover, a three-dimensional (3D) cell culture model can provide a more physiologically meaningful culture microenvironment. However, studying cellular crosstalk in a 3D cell culture model involves tedious processing. In this study, a paper/poly(methyl methacrylate) (PMMA) hybrid 3D cell culture microfluidic platform was successfully developed for the study of cellular crosstalk. The platform was a paper substrate with culture microreactors placed on a PMMA substrate with hydrogel-infused channels. Different types of cells were directly seeded and cultured in the microreactors. Aberrant cell proliferation of the affected cells was induced by secretions from transfected cells, and the proliferation ratios were investigated using a colorimetric method. The results showed that the responses of cellular crosstalk were different in different types of cells. Moreover, neutralizing and competitive assays were performed to show the functionality of the platform. Additionally, the triggered signaling pathways of the affected cells were directly analyzed by a subsequent immunoassay. The microfluidic platform provides a simple method for studying cellular crosstalk and the corresponding signaling pathways in a 3D culture model.
Collapse
Affiliation(s)
- Kin Fong Lei
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Linkou Branch , Taoyuan 333, Taiwan
| | | | | |
Collapse
|
108
|
Sandén E, Dyberg C, Krona C, Gallo-Oller G, Olsen TK, Enríquez Pérez J, Wickström M, Estekizadeh A, Kool M, Visse E, Ekström TJ, Siesjö P, Johnsen JI, Darabi A. Establishment and characterization of an orthotopic patient-derived Group 3 medulloblastoma model for preclinical drug evaluation. Sci Rep 2017; 7:46366. [PMID: 28417956 PMCID: PMC5394470 DOI: 10.1038/srep46366] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 03/15/2017] [Indexed: 12/25/2022] Open
Abstract
Medulloblastomas comprise a heterogeneous group of tumours and can be subdivided into four molecular subgroups (WNT, SHH, Group 3 and Group 4) with distinct prognosis, biological behaviour and implications for targeted therapies. Few experimental models exist of the aggressive and poorly characterized Group 3 tumours. In order to establish a reproducible transplantable Group 3 medulloblastoma model for preclinical therapeutic studies, we acquired a patient-derived tumour sphere culture and inoculated low-passage spheres into the cerebellums of NOD-scid mice. Mice developed symptoms of brain tumours with a latency of 17–18 weeks. Neurosphere cultures were re-established and serially transplanted for 3 generations, with a negative correlation between tumour latency and numbers of injected cells. Xenografts replicated the phenotype of the primary tumour, including high degree of clustering in DNA methylation analysis, high proliferation, expression of tumour markers, MYC amplification and elevated MYC expression, and sensitivity to the MYC inhibitor JQ1. Xenografts maintained maintained expression of tumour-derived VEGFA and stromal-derived COX-2. VEGFA, COX-2 and c-Myc are highly expressed in Group 3 compared to other medulloblastoma subgroups, suggesting that these molecules are relevant therapeutic targets in Group 3 medulloblastoma.
Collapse
Affiliation(s)
- Emma Sandén
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Neurosurgery, Lund, Sweden
| | - Cecilia Dyberg
- Karolinska Institutet, Department of Women´s and Children´s Health, Childhood Cancer Research Unit, Stockholm, Sweden
| | - Cecilia Krona
- Uppsala University, Department of Immunology, Genetics and Pathology, Uppsala, Sweden
| | - Gabriel Gallo-Oller
- Karolinska Institutet, Department of Women´s and Children´s Health, Childhood Cancer Research Unit, Stockholm, Sweden
| | - Thale Kristin Olsen
- Karolinska Institutet, Department of Women´s and Children´s Health, Childhood Cancer Research Unit, Stockholm, Sweden
| | - Julio Enríquez Pérez
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Neurosurgery, Lund, Sweden
| | - Malin Wickström
- Karolinska Institutet, Department of Women´s and Children´s Health, Childhood Cancer Research Unit, Stockholm, Sweden
| | - Atosa Estekizadeh
- Karolinska University Hospital, Solna, Center for Molecular Medicine, and Karolinska Institutet, Department of Clinical Neuroscience, Stockholm, Sweden
| | - Marcel Kool
- German Cancer Research Center DKFZ, Division of Pediatric Neurooncology, Heidelberg, Germany
| | - Edward Visse
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Neurosurgery, Lund, Sweden
| | - Tomas J Ekström
- Karolinska University Hospital, Solna, Center for Molecular Medicine, and Karolinska Institutet, Department of Clinical Neuroscience, Stockholm, Sweden
| | - Peter Siesjö
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Neurosurgery, Lund, Sweden.,Lund University, Skane University Hospital, Department of Clinical Sciences Lund, Neurosurgery, Lund, Sweden
| | - John Inge Johnsen
- Karolinska Institutet, Department of Women´s and Children´s Health, Childhood Cancer Research Unit, Stockholm, Sweden
| | - Anna Darabi
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Neurosurgery, Lund, Sweden
| |
Collapse
|
109
|
Integrating the glioblastoma microenvironment into engineered experimental models. Future Sci OA 2017; 3:FSO189. [PMID: 28883992 PMCID: PMC5583655 DOI: 10.4155/fsoa-2016-0094] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/22/2017] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is the most lethal cancer originating in the brain. Its high mortality rate has been attributed to therapeutic resistance and rapid, diffuse invasion - both of which are strongly influenced by the unique microenvironment. Thus, there is a need to develop new models that mimic individual microenvironmental features and are able to provide clinically relevant data. Current understanding of the effects of the microenvironment on GBM progression, established experimental models of GBM and recent developments using bioengineered microenvironments as ex vivo experimental platforms that mimic the biochemical and physical properties of GBM tumors are discussed.
Collapse
|
110
|
Relationship of inflammatory profile of elderly patients serum and senescence-associated secretory phenotype with human breast cancer cells proliferation: Role of IL6/IL8 ratio. Cytokine 2017; 91:13-29. [DOI: 10.1016/j.cyto.2016.12.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 11/15/2016] [Accepted: 12/02/2016] [Indexed: 12/19/2022]
|
111
|
McConnell HL, Kersch CN, Woltjer RL, Neuwelt EA. The Translational Significance of the Neurovascular Unit. J Biol Chem 2016; 292:762-770. [PMID: 27920202 DOI: 10.1074/jbc.r116.760215] [Citation(s) in RCA: 209] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The mammalian brain is supplied with blood by specialized vasculature that is structurally and functionally distinct from that of the periphery. A defining feature of this vasculature is a physical blood-brain barrier (BBB). The BBB separates blood components from the brain microenvironment, regulating the entry and exit of ions, nutrients, macromolecules, and energy metabolites. Over the last two decades, physiological studies of cerebral blood flow dynamics have demonstrated that substantial intercellular communication occurs between cells of the vasculature and the neurons and glia that abut the vasculature. These findings suggest that the BBB does not function independently, but as a module within the greater context of a multicellular neurovascular unit (NVU) that includes neurons, astrocytes, pericytes, and microglia as well as the blood vessels themselves. Here, we describe the roles of these NVU components as well as how they act in concert to modify cerebrovascular function and permeability in health and in select diseases.
Collapse
Affiliation(s)
- Heather L McConnell
- From the Departments of Neurology, Pathology, Neurosurgery, and Veterans Affairs, Oregon Health & Science University, Portland, Oregon 97239-2941
| | - Cymon N Kersch
- From the Departments of Neurology, Pathology, Neurosurgery, and Veterans Affairs, Oregon Health & Science University, Portland, Oregon 97239-2941
| | - Randall L Woltjer
- From the Departments of Neurology, Pathology, Neurosurgery, and Veterans Affairs, Oregon Health & Science University, Portland, Oregon 97239-2941
| | - Edward A Neuwelt
- From the Departments of Neurology, Pathology, Neurosurgery, and Veterans Affairs, Oregon Health & Science University, Portland, Oregon 97239-2941
| |
Collapse
|
112
|
Boccellino M, Quagliuolo L, Alaia C, Grimaldi A, Addeo R, Nicoletti GF, Kast RE, Caraglia M. The strange connection between epidermal growth factor receptor tyrosine kinase inhibitors and dapsone: from rash mitigation to the increase in anti-tumor activity. Curr Med Res Opin 2016; 32:1839-1848. [PMID: 27398628 DOI: 10.1080/03007995.2016.1211522] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The presence of an aberrantly activated epidermal growth factor receptor (EGFR) in many epithelial tumors, due to its overexpression, activating mutations, gene amplification and/or overexpression of receptor ligands, represent the fundamental basis underlying the use of EGFR tyrosine kinase inhibitors (EGFR-TKIs). Drugs inhibiting the EGFR have different mechanisms of action; while erlotinib and gefitinib inhibit the intracellular tyrosine kinase, monoclonal antibodies like cetuximab and panitumumab bind the extracellular domain of the EGFR both activating immunomediated anti-cancer effect and inhibiting receptor function. On the other hand, interleukin-8 has tumor promoting as well as neo-angiogenesis enhancing effects and several attempts have been made to inhibit its activity. One of these is based on the use of the old sulfone antibiotic dapsone that has demonstrated several interleukin-8 system inhibiting actions. Erlotinib typically gives a rash that has recently been proven to come out via up-regulated keratinocyte interleukin-8 synthesis with histological features reminiscent of typical neutrophilic dermatoses. In this review, we report experimental evidence that shows the use of dapsone to improve quality of life in erlotinib-treated patients by ameliorating rash as well as short-circuiting a growth-enhancing aspect of erlotinib based on increased interleukin-8 secretion.
Collapse
Affiliation(s)
- Mariarosaria Boccellino
- a Department of Biochemistry, Biophysics and General Pathology , Second University of Naples , Naples , Italy
| | - Lucio Quagliuolo
- a Department of Biochemistry, Biophysics and General Pathology , Second University of Naples , Naples , Italy
| | - Concetta Alaia
- a Department of Biochemistry, Biophysics and General Pathology , Second University of Naples , Naples , Italy
| | - Anna Grimaldi
- a Department of Biochemistry, Biophysics and General Pathology , Second University of Naples , Naples , Italy
| | - Raffaele Addeo
- b Oncology DH ASL Napoli 3 Nord, Frattamaggiore Hospital , Frattamaggiore , Naples , Italy
| | | | | | - Michele Caraglia
- a Department of Biochemistry, Biophysics and General Pathology , Second University of Naples , Naples , Italy
| |
Collapse
|
113
|
McCoy MG, Seo BR, Choi S, Fischbach C. Collagen I hydrogel microstructure and composition conjointly regulate vascular network formation. Acta Biomater 2016; 44:200-8. [PMID: 27545811 PMCID: PMC5045803 DOI: 10.1016/j.actbio.2016.08.028] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 07/22/2016] [Accepted: 08/17/2016] [Indexed: 11/18/2022]
Abstract
UNLABELLED Neovascularization is a hallmark of physiological and pathological tissue remodeling that is regulated in part by the extracellular matrix (ECM). Collagen I hydrogels or Matrigel are frequently used to study vascular network formation; however, in isolation these materials do not typically mimic the integrated effects of ECM structure and composition that may influence endothelial cells in vivo. Here, we have utilized microfabricated 3D culture models to control collagen I microstructure in the presence and absence of Matrigel and tested the effect of these variations on vascular network formation by human cerebral microvascular endothelial cells (hCMECs). Varied collagen microarchitecture was achieved by adjusting the gelation temperature and subsequently confirmed by structural analysis. Casting at colder temperature increased collagen fiber thickness and length, and inclusion of Matrigel further pronounced these differences. Interestingly, the presence of Matrigel affected vascular network formation by modulating hCMEC growth, whereas altered collagen fiber structure impacted the morphology and maturity of the developed vascular network. These differences were related to substrate-dependent changes in interleukin-8 (IL-8) secretion and were functionally relevant as vascular networks preformed in more fibrillar, Matrigel-containing hydrogels promoted angiogenic sprouting. Our studies indicate that collagen hydrogel microstructure and composition conjointly regulate vascular network formation with implications for translational and basic science approaches. STATEMENT OF SIGNIFICANCE Neovascularization is a hallmark of both tissue homeostasis and disease and is in part regulated by cell remodeling that occurs in the extracellular matrix (ECM). The use of bio-mimetic hydrogel cell culture systems has been used to study the effects of the ECM on cell behavior. Here, we employ a hydrogel system that enables control over both the structure and composition of the ECM and subsequently investigated the effects that these have on blood vessel dynamics. Finally, we linked these differences to changes in protein secretion and the implications that this may play in scientific translation.
Collapse
Affiliation(s)
- Michael G McCoy
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, United States
| | - Bo Ri Seo
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, United States
| | - Siyoung Choi
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, United States
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, United States; Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY 14853, United States.
| |
Collapse
|
114
|
Florczyk SJ, Kievit FM, Wang K, Erickson AE, Ellenbogen RG, Zhang M. 3D Porous Chitosan-Alginate Scaffolds Promote Proliferation and Enrichment of Cancer Stem-Like Cells. J Mater Chem B 2016; 4:6326-6334. [PMID: 28133535 PMCID: PMC5260821 DOI: 10.1039/c6tb01713d] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancer stem cells are increasingly becoming a primary target for new cancer treatment development. The ability to study their transient behavior in vitro will provide the opportunity for high-throughput testing of more effective therapies. We have previously demonstrated the use of 3D porous chitosan-alginate (CA) scaffolds to promote cancer stem-like cell (CSC) proliferation and enrichment in glioblastoma. Here we use 3D porous CA scaffolds to promote cancer stem-like cell enrichment in cell lines from prostate, liver, and breast cancers, and investigate the proliferation, morphology, and gene expressions of cells cultured in CA scaffolds as compared to 2D controls. The 3D CA scaffold cultures for all three cancer types showed reduced proliferation, formation of tumor spheroids, and increased expression of CSC associated mark genes (CD133 and NANOG), as opposed to monolayers. Additionally, we present a putative mechanism for the cancer stem-like cell enrichment on CA scaffolds. This study demonstrates that the cancer stem-like cell enrichment in CA scaffolds is a robust process that is not restricted to particular cancer types.
Collapse
Affiliation(s)
- Stephen J. Florczyk
- Department of Materials Science & Engineering, University of Washington, Seattle, WA 98195
| | - Forrest M. Kievit
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195
| | - Kui Wang
- Department of Materials Science & Engineering, University of Washington, Seattle, WA 98195
| | - Ariane E. Erickson
- Department of Materials Science & Engineering, University of Washington, Seattle, WA 98195
| | | | - Miqin Zhang
- Department of Materials Science & Engineering, University of Washington, Seattle, WA 98195
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195
| |
Collapse
|
115
|
Ping YF, Zhang X, Bian XW. Cancer stem cells and their vascular niche: Do they benefit from each other? Cancer Lett 2016; 380:561-567. [DOI: 10.1016/j.canlet.2015.05.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 04/07/2015] [Accepted: 05/08/2015] [Indexed: 12/22/2022]
|
116
|
Hu CT, Guo LL, Feng N, Zhang L, Zhou N, Ma LL, Shen L, Tong GH, Yan QW, Zhu SJ, Bian XW, Lai MD, Deng YJ, Ding YQ. MIF, secreted by human hepatic sinusoidal endothelial cells, promotes chemotaxis and outgrowth of colorectal cancer in liver prometastasis. Oncotarget 2016; 6:22410-23. [PMID: 26087187 PMCID: PMC4673172 DOI: 10.18632/oncotarget.4198] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 05/20/2015] [Indexed: 12/27/2022] Open
Abstract
Growth and invasion of metastatic colorectal cancer (CRC) cells in the liver depend on microenvironment. Here, we showed that human hepatic sinusoidal endothelial cells (HHSECs) induce chemotaxis and outgrowth of CRC cells. Macrophage migration inhibitory factor (MIF), released by HHSECs, stimulated chemotaxis of CRC cells. MIF secreted by HHSECs, but not by CRC cells themselves, promoted migration and epithelial-mesenchymal transition (EMT) and facilitated proliferation and apoptotic resistance of CRC cells. In orthotopic implantation models in nude mice, exogenous MIF stimulated growth of CRC cells and metastasis. Furthermore, MIF accelerated mobility of CRC cells by suppressing F-actin depolymerization and phosphorylating cofilin. Noteworthy, MIF levels were correlated with the size of hepatic metastases. We suggest that HHSECs and paracrine MIF promote initial migration and proliferation of CRC cells in the hepatic sinusoids to generate liver metastases.
Collapse
Affiliation(s)
- Chun-Ting Hu
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Li-Li Guo
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Na Feng
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Lei Zhang
- Department of General Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Na Zhou
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Li-Li Ma
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lan Shen
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Gui-Hui Tong
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Qian-Wen Yan
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shi-Jie Zhu
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiu-Wu Bian
- Department of Pathology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Mao D Lai
- Department of Pathology, School of Medical Sciences, Zhejiang University, Hangzhou 310006, China
| | - Yong-Jian Deng
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Yan-Qing Ding
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| |
Collapse
|
117
|
Tabatabaei P, Visse E, Bergström P, Brännström T, Siesjö P, Bergenheim AT. Radiotherapy induces an immediate inflammatory reaction in malignant glioma: a clinical microdialysis study. J Neurooncol 2016; 131:83-92. [PMID: 27664151 PMCID: PMC5258803 DOI: 10.1007/s11060-016-2271-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 08/30/2016] [Indexed: 11/27/2022]
Abstract
The knowledge of response to radiation in the immuno-microenvironment of high grade gliomas is sparse. In vitro results have indicated an inflammatory response of myeloid cells after irradiation. Therefore, microdialysis was used to verify whether this is operative in tumor tissue and brain adjacent to tumor (BAT) after clinical radiotherapy of patients with high grade glioma. Stereotactic biopsies and implantation of microdialysis catheters in tumor tissue and BAT were performed in eleven patients with high-grade glioma. The patients were given daily radiation fractions of 2–3.4 Gy. Microdialysis samples were collected before radiotherapy and during the first five days of radiation. Cytokines, glucose metabolites, glutamate and glycerol were analyzed. Immunohistochemistry was performed to detect macrophages (CD68) and monocytes (CD163) as well as IL-6, IL-8 and MCP-1. A significant increase of IL-8, MCP-1 and MIP-1a were detected in tumor tissue already after the first dose of radiation and increased further during 5 days of radiation. IL-6 did also increase but after five fractions of radiation. In BAT, the cytokine response was modest with significant increase of IL-8 after third dose of radiation. We found a positive correlation between baseline IL-8 and IL-6 microdialysis levels in tumor tissue and survival. Glucose metabolites or glycerol and glutamate did not change during radiation. In all tumors staining for macrophages was demonstrated. IL-6 was found in viable tumor cells while MCP-1 was demonstrated in macrophages or tumor matrix. Our findings suggest that radiation induces a rapid enhancement of the prevailing inflammation in high-grade glioma tissue. The microdialysis technique is feasible for this type of study and could be used to monitor metabolic changes after different interventions.
Collapse
Affiliation(s)
- Pedram Tabatabaei
- Department of Clinical Neuroscience, Neurosurgery, Umea University, 901 85, Umeå, Sweden.
| | - Eward Visse
- Department of Clinical Science, Lund University Hospital, 221 85, Lund, Sweden
| | - Per Bergström
- Department of Radiation Science, Umeå University, 901 85, Umeå, Sweden
| | - Thomas Brännström
- Department of Medical Bioscience, Umeå University, 901 85, Umeå, Sweden
| | - Peter Siesjö
- Department of Clinical Science, Lund University Hospital, 221 85, Lund, Sweden
| | - A Tommy Bergenheim
- Department of Clinical Neuroscience, Neurosurgery, Umea University, 901 85, Umeå, Sweden
| |
Collapse
|
118
|
Vascular Transdifferentiation in the CNS: A Focus on Neural and Glioblastoma Stem-Like Cells. Stem Cells Int 2016; 2016:2759403. [PMID: 27738435 PMCID: PMC5055959 DOI: 10.1155/2016/2759403] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 09/05/2016] [Indexed: 01/12/2023] Open
Abstract
Glioblastomas are devastating and extensively vascularized brain tumors from which glioblastoma stem-like cells (GSCs) have been isolated by many groups. These cells have a high tumorigenic potential and the capacity to generate heterogeneous phenotypes. There is growing evidence to support the possibility that these cells are derived from the accumulation of mutations in adult neural stem cells (NSCs) as well as in oligodendrocyte progenitors. It was recently reported that GSCs could transdifferentiate into endothelial-like and pericyte-like cells both in vitro and in vivo, notably under the influence of Notch and TGFβ signaling pathways. Vascular cells derived from GBM cells were also observed directly in patient samples. These results could lead to new directions for designing original therapeutic approaches against GBM neovascularization but this specific reprogramming requires further molecular investigations. Transdifferentiation of nontumoral neural stem cells into vascular cells has also been described and conversely vascular cells may generate neural stem cells. In this review, we present and discuss these recent data. As some of them appear controversial, further validation will be needed using new technical approaches such as high throughput profiling and functional analyses to avoid experimental pitfalls and misinterpretations.
Collapse
|
119
|
Wittmann K, Fischbach C. Contextual Control of Adipose-Derived Stem Cell Function: Implications for Engineered Tumor Models. ACS Biomater Sci Eng 2016; 3:1483-1493. [DOI: 10.1021/acsbiomaterials.6b00328] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Katharina Wittmann
- Nancy E. and Peter C. Meinig School of Biomedical
Engineering and ‡Kavli Institute
at Cornell for Nanoscale Science, Cornell University, Ithaca, New York 14850, United States
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical
Engineering and ‡Kavli Institute
at Cornell for Nanoscale Science, Cornell University, Ithaca, New York 14850, United States
| |
Collapse
|
120
|
Nassar D, Blanpain C. Cancer Stem Cells: Basic Concepts and Therapeutic Implications. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2016; 11:47-76. [DOI: 10.1146/annurev-pathol-012615-044438] [Citation(s) in RCA: 405] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Dany Nassar
- IRIBHM, Université Libre de Bruxelles, Brussels B-1070, Belgium;
| | - Cédric Blanpain
- IRIBHM, Université Libre de Bruxelles, Brussels B-1070, Belgium;
- WELBIO, Université Libre de Bruxelles, Brussels B-1070, Belgium
| |
Collapse
|
121
|
Raju ENS, Kuechler J, Behling S, Sridhar S, Hirseland E, Tronnier V, Zechel C. Maintenance of Stemlike Glioma Cells and Microglia in an Organotypic Glioma Slice Model. Neurosurgery 2016; 77:629-43; discussion 643. [PMID: 26308638 DOI: 10.1227/neu.0000000000000891] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The therapeutic resistance of gliomas is, at least in part, due to stemlike glioma cells (SLGCs), which self-renew, generate the bulk of tumor cells, and sustain tumor growth. SLGCs from glioblastomas (GB) have been studied in cell cultures or mouse models, whereas little is known about SLGCs from lower grade gliomas. OBJECTIVE To compare cell and organotypic slice cultures from GBs and lower grade gliomas and study the maintenance of SLGCs. METHODS Cells and tissue slices from astrocytomas, oligodendrogliomas, oligoastrocytomas, and GBs were cultivated in (1) serum-free medium supplemented with the growth factors epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF), (2) medium containing 10% serum plus EGF and bFGF (F+GF medium), or (3) medium containing 10% fetal calf serum (F medium). Maintenance of cells and cytoarchitecture was addressed, using several candidate SLGC markers (Nestin, Sox2, CD133, CD44, CD49f/integrin α6, and Notch) as well as CD31 (endothelial cells), ionized calcium-binding adapter molecule 1 (microglia), and vimentin. Cell vitality was determined. RESULTS SLGCs were present in tissue slices from lower and higher grade gliomas. Preservation of the cytoarchitecture in slices was possible for >3 weeks. Maintenance of SLGCs required the presence of EGF/bFGF in cell and slice cultures, in which F+GF appeared superior to N medium. Constraints were observed regarding the preservation of the microglia but not of the endothelial cells. Maintenance of the microglia was improved by addition of the cytokine macrophage colony-stimulating factor. CONCLUSION Medium supplemented with serum and growth factors EGF, bFGF, and macrophage colony-stimulating factor permits the preservation of SLGCs and non-SLGCs in the original glioma microenvironment.
Collapse
Affiliation(s)
- E N Sanjaya Raju
- Department of Neurosurgery, University of Lübeck, Lübeck, Germany
| | | | | | | | | | | | | |
Collapse
|
122
|
Ahn SH, Park H, Ahn YH, Kim S, Cho MS, Kang JL, Choi YH. Necrotic cells influence migration and invasion of glioblastoma via NF-κB/AP-1-mediated IL-8 regulation. Sci Rep 2016; 6:24552. [PMID: 27076368 PMCID: PMC4830983 DOI: 10.1038/srep24552] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 03/31/2016] [Indexed: 01/01/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common primary intracranial tumor in adults and has poor prognosis. Diffuse infiltration into normal brain parenchyma, rapid growth, and the presence of necrosis are remarkable hallmarks of GBM. However, the effect of necrotic cells on GBM growth and metastasis is poorly understood at present. In this study, we examined the biological significance of necrotic tissues by exploring the molecular mechanisms underlying the signaling network between necrotic tissues and GBM cells. The migration and invasion of the GBM cell line CRT-MG was significantly enhanced by treatment with necrotic cells, as shown by assays for scratch wound healing and spheroid invasion. Incubation with necrotic cells induced IL-8 secretion in CRT-MG cells in a dose-dependent manner. In human GBM tissues, IL-8 positive cells were mainly distributed in the perinecrotic region, as seen in immunohistochemistry and immunofluorescence analysis. Necrotic cells induced NF-κB and AP-1 activation and their binding to the IL-8 promoter, leading to enhanced IL-8 production and secretion in GBM cells. Our data demonstrate that when GBM cells are exposed to and stimulated by necrotic cells, the migration and invasion of GBM cells are enhanced and facilitated via NF-κB/AP-1 mediated IL-8 upregulation.
Collapse
Affiliation(s)
- So-Hee Ahn
- Department of Physiology, Ewha Womans University School of Medicine, Seoul 911-1, Korea.,Tissue Injury Defense Research Center , School of Medicine, Ewha Womans University, Seoul, Korea
| | - Hyunju Park
- Department of Physiology, Ewha Womans University School of Medicine, Seoul 911-1, Korea.,Tissue Injury Defense Research Center , School of Medicine, Ewha Womans University, Seoul, Korea
| | - Young-Ho Ahn
- Tissue Injury Defense Research Center , School of Medicine, Ewha Womans University, Seoul, Korea.,Department of Molecular Medicine , School of Medicine, Ewha Womans University, Seoul, Korea
| | - Sewha Kim
- Department of Pathology, School of Medicine, Ewha Womans University, Seoul, Korea
| | - Min-Sun Cho
- Department of Pathology, School of Medicine, Ewha Womans University, Seoul, Korea
| | - Jihee Lee Kang
- Department of Physiology, Ewha Womans University School of Medicine, Seoul 911-1, Korea.,Tissue Injury Defense Research Center , School of Medicine, Ewha Womans University, Seoul, Korea
| | - Youn-Hee Choi
- Department of Physiology, Ewha Womans University School of Medicine, Seoul 911-1, Korea.,Tissue Injury Defense Research Center , School of Medicine, Ewha Womans University, Seoul, Korea
| |
Collapse
|
123
|
Bronisz A, Godlewski J, Chiocca EA. Extracellular Vesicles and MicroRNAs: Their Role in Tumorigenicity and Therapy for Brain Tumors. Cell Mol Neurobiol 2016; 36:361-76. [PMID: 26983830 DOI: 10.1007/s10571-015-0293-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/22/2015] [Indexed: 12/12/2022]
Abstract
MicroRNAs are small non-coding RNAs which mediate post-transcriptional gene regulation. Recently, microRNAs have also been found to be localized to the extracellular space, often encapsulated in secreted extracellular vesicles (EVs). This tandem of EVs and tissue-specific expressed/secreted microRNAs that can be taken up by neighboring or distant recipient cells, leading to changes in gene expression-suggests a cell-specialized role in physiological and pathological conditions. The complexity of solid tumors and their distinct pathophysiology relies on interactive communications between the various cell types in the neoplasm (tumor, endothelial, or macrophages, for instance). Understanding how such EV/microRNA-mediated communication occurs may actually lead to avenues for therapeutic exploitation and/or intervention, particularly for the most formidable cancers, such as those in the brain. In this review, the role of microRNAs/EVs in brain tumors will be discussed with emphasis on how these molecules could be utilized for tumor therapy.
Collapse
Affiliation(s)
- Agnieszka Bronisz
- Department of Neurosurgery, Harvey Cushing Neuro-oncology Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jakub Godlewski
- Department of Neurosurgery, Harvey Cushing Neuro-oncology Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - E Antonio Chiocca
- Department of Neurosurgery, Harvey Cushing Neuro-oncology Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
124
|
Liu YN, Chang TH, Tsai MF, Wu SG, Tsai TH, Chen HY, Yu SL, Yang JCH, Shih JY. IL-8 confers resistance to EGFR inhibitors by inducing stem cell properties in lung cancer. Oncotarget 2016; 6:10415-31. [PMID: 25871388 PMCID: PMC4496364 DOI: 10.18632/oncotarget.3389] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 02/15/2015] [Indexed: 01/17/2023] Open
Abstract
Epidermal growth factor receptor (EGFR)-targeted strategy is limited by resistance. We identify the potential genes involved in EGFR TKI (tyrosine kinase inhibitor) resistance and study the therapeutic mechanism in the non-small cell lung cancers. Potential genes involved in resistance were examined by analyzing datasets from a pair of EGFR TKI-sensitive (PC9) and TKI-resistant cells (PC9/gef). Blood specimens from patients taking EGFR TKI as first-line treatment were used to examine the correlation between drug's efficacy and IL-8 level. The effects of IL-8 on gefitinib-induced apoptosis, stemness, and in vivo tumorigenicity were investigated using established cell lines. We identified IL-8 was up-regulated in gefitinib-resistant cells, and high plasma IL-8 level was correlated with shorter progression-free-survival time. IL-8 overexpression suppressed gefitinib-induced apoptosis in gefitinib-sensitive cells. By contrast, suppression of IL-8 enhanced gefitinib-induced cell death in gefitinib-resistant cells. IL-8 also increased stem-like characteristics including aldehyde dehydrogenase activity, expression of stemness-related genes, clonogenic activity, side-population, and in vivo tumorigenicity. Consistently, knockdown of IL-8 leads to loss of stem cell-like characteristics in gefitinib-resistant cells. Our study demonstrates an important role for IL-8, and suggests IL-8 is a potential therapeutic target for overcoming EGFR TKI resistance.
Collapse
Affiliation(s)
- Yi-Nan Liu
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tzu-Hua Chang
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Meng-Feng Tsai
- Department of Molecular Biotechnology, Dayeh University, Changhua, Taiwan
| | - Shang-Gin Wu
- Department of Internal Medicine, National Taiwan University Hospital, Yun-Lin Branch, Yun-Lin, Taiwan
| | - Tzu-Hsiu Tsai
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsuan-Yu Chen
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
| | - Sung-Liang Yu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | - James Chih-Hsin Yang
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan.,Graduate Institute of Oncology, Cancer Research Center, National Taiwan University, Taipei, Taiwan
| | - Jin-Yuan Shih
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
125
|
Alfaro C, Teijeira A, Oñate C, Pérez G, Sanmamed MF, Andueza MP, Alignani D, Labiano S, Azpilikueta A, Rodriguez-Paulete A, Garasa S, Fusco JP, Aznar A, Inogés S, De Pizzol M, Allegretti M, Medina-Echeverz J, Berraondo P, Perez-Gracia JL, Melero I. Tumor-Produced Interleukin-8 Attracts Human Myeloid-Derived Suppressor Cells and Elicits Extrusion of Neutrophil Extracellular Traps (NETs). Clin Cancer Res 2016; 22:3924-36. [PMID: 26957562 DOI: 10.1158/1078-0432.ccr-15-2463] [Citation(s) in RCA: 299] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 02/03/2016] [Indexed: 12/17/2022]
Abstract
PURPOSE Myeloid-derived suppressor cells (MDSC) are considered an important T-cell immunosuppressive component in cancer-bearing hosts. The factors that attract these cells to the tumor microenvironment are poorly understood. IL8 (CXCL8) is a potent chemotactic factor for neutrophils and monocytes. EXPERIMENTAL DESIGN MDSC were characterized and sorted by multicolor flow cytometry on ficoll-gradient isolated blood leucokytes from healthy volunteers (n = 10) and advanced cancer patients (n = 28). In chemotaxis assays, sorted granulocytic and monocytic MDSC were tested in response to recombinant IL8, IL8 derived from cancer cell lines, and patient sera. Neutrophil extracellular traps (NETs) formation was assessed by confocal microscopy, fluorimetry, and time-lapse fluorescence confocal microscopy on short-term MDSC cultures. RESULTS IL8 chemoattracts both granulocytic (GrMDSC) and monocytic (MoMDSC) human MDSC. Monocytic but not granulocytic MDSC exerted a suppressor activity on the proliferation of autologous T cells isolated from the circulation of cancer patients. IL8 did not modify the T-cell suppressor activity of human MDSC. However, IL8 induced the formation of NETs in the GrMDSC subset. CONCLUSIONS IL8 derived from tumors contributes to the chemotactic recruitment of MDSC and to their functional control. Clin Cancer Res; 22(15); 3924-36. ©2016 AACR.
Collapse
Affiliation(s)
- Carlos Alfaro
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain. Department of Oncology, University Clinic of Navarra, Pamplona, Spain. Department of Immunology, University Clinic of Navarra, Pamplona, Spain
| | - Alvaro Teijeira
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain. Department of Oncology, University Clinic of Navarra, Pamplona, Spain. Department of Immunology, University Clinic of Navarra, Pamplona, Spain
| | - Carmen Oñate
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain. Department of Oncology, University Clinic of Navarra, Pamplona, Spain. Department of Immunology, University Clinic of Navarra, Pamplona, Spain
| | - Guiomar Pérez
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain. Department of Oncology, University Clinic of Navarra, Pamplona, Spain. Department of Immunology, University Clinic of Navarra, Pamplona, Spain
| | - Miguel F Sanmamed
- Department of Oncology, University Clinic of Navarra, Pamplona, Spain. Department of Immunology, University Clinic of Navarra, Pamplona, Spain
| | - Maria Pilar Andueza
- Department of Oncology, University Clinic of Navarra, Pamplona, Spain. Department of Immunology, University Clinic of Navarra, Pamplona, Spain
| | - Diego Alignani
- Cytometry Platform, Centre for Applied Medical Research (CIMA), Pamplona, Spain
| | - Sara Labiano
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain
| | - Arantza Azpilikueta
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain
| | - Alfonso Rodriguez-Paulete
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain
| | - Saray Garasa
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain
| | - Juan P Fusco
- Department of Oncology, University Clinic of Navarra, Pamplona, Spain. Department of Immunology, University Clinic of Navarra, Pamplona, Spain
| | - Angela Aznar
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain
| | - Susana Inogés
- Department of Oncology, University Clinic of Navarra, Pamplona, Spain. Department of Immunology, University Clinic of Navarra, Pamplona, Spain
| | | | | | - Jose Medina-Echeverz
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain
| | - Pedro Berraondo
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain
| | - Jose L Perez-Gracia
- Department of Oncology, University Clinic of Navarra, Pamplona, Spain. Department of Immunology, University Clinic of Navarra, Pamplona, Spain
| | - Ignacio Melero
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain. Department of Oncology, University Clinic of Navarra, Pamplona, Spain. Department of Immunology, University Clinic of Navarra, Pamplona, Spain.
| |
Collapse
|
126
|
Katt ME, Placone AL, Wong AD, Xu ZS, Searson PC. In Vitro Tumor Models: Advantages, Disadvantages, Variables, and Selecting the Right Platform. Front Bioeng Biotechnol 2016; 4:12. [PMID: 26904541 PMCID: PMC4751256 DOI: 10.3389/fbioe.2016.00012] [Citation(s) in RCA: 494] [Impact Index Per Article: 54.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 01/28/2016] [Indexed: 12/19/2022] Open
Abstract
In vitro tumor models have provided important tools for cancer research and serve as low-cost screening platforms for drug therapies; however, cancer recurrence remains largely unchecked due to metastasis, which is the cause of the majority of cancer-related deaths. The need for an improved understanding of the progression and treatment of cancer has pushed for increased accuracy and physiological relevance of in vitro tumor models. As a result, in vitro tumor models have concurrently increased in complexity and their output parameters further diversified, since these models have progressed beyond simple proliferation, invasion, and cytotoxicity screens and have begun recapitulating critical steps in the metastatic cascade, such as intravasation, extravasation, angiogenesis, matrix remodeling, and tumor cell dormancy. Advances in tumor cell biology, 3D cell culture, tissue engineering, biomaterials, microfabrication, and microfluidics have enabled rapid development of new in vitro tumor models that often incorporate multiple cell types, extracellular matrix materials, and spatial and temporal introduction of soluble factors. Other innovations include the incorporation of perfusable microvessels to simulate the tumor vasculature and model intravasation and extravasation. The drive toward precision medicine has increased interest in adapting in vitro tumor models for patient-specific therapies, clinical management, and assessment of metastatic potential. Here, we review the wide range of current in vitro tumor models and summarize their advantages, disadvantages, and suitability in modeling specific aspects of the metastatic cascade and drug treatment.
Collapse
Affiliation(s)
- Moriah E Katt
- Institute for Nanobiotechnology (INBT), Johns Hopkins University, Baltimore, MD, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Amanda L Placone
- Institute for Nanobiotechnology (INBT), Johns Hopkins University, Baltimore, MD, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Andrew D Wong
- Institute for Nanobiotechnology (INBT), Johns Hopkins University, Baltimore, MD, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Zinnia S Xu
- Institute for Nanobiotechnology (INBT), Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Peter C Searson
- Institute for Nanobiotechnology (INBT), Johns Hopkins University, Baltimore, MD, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
127
|
Glioma Stem Cells and Their Microenvironments: Providers of Challenging Therapeutic Targets. Stem Cells Int 2016; 2016:5728438. [PMID: 26977157 PMCID: PMC4764748 DOI: 10.1155/2016/5728438] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/12/2015] [Accepted: 01/06/2016] [Indexed: 12/26/2022] Open
Abstract
Malignant gliomas are aggressive brain tumors with limited therapeutic options, possibly because of highly tumorigenic subpopulations of glioma stem cells. These cells require specific microenvironments to maintain their “stemness,” described as perivascular and hypoxic niches. Each of those niches induces particular signatures in glioma stem cells (e.g., activation of Notch signaling, secretion of VEGF, bFGF, SDF1 for the vascular niche, activation of HIF2α, and metabolic reprogramming for hypoxic niche). Recently, accumulated knowledge on tumor-associated macrophages, possibly delineating a third niche, has underlined the role of immune cells in glioma progression, via specific chemoattractant factors and cytokines, such as macrophage-colony stimulation factor (M-CSF). The local or myeloid origin of this new component of glioma stem cells niche is yet to be determined. Such niches are being increasingly recognized as key regulators involved in multiple stages of disease progression, therapy resistance, immune-escaping, and distant metastasis, thereby substantially impacting the future development of frontline interventions in clinical oncology. This review focuses on the microenvironment impact on the glioma stem cell biology, emphasizing GSCs cross talk with hypoxic, perivascular, and immune niches and their potential use as targeted therapy.
Collapse
|
128
|
Parajuli P, Anand R, Mandalaparty C, Suryadevara R, Sriranga PU, Michelhaugh SK, Cazacu S, Finniss S, Thakur A, Lum LG, Schalk D, Brodie C, Mittal S. Preferential expression of functional IL-17R in glioma stem cells: potential role in self-renewal. Oncotarget 2016; 7:6121-35. [PMID: 26755664 PMCID: PMC4868744 DOI: 10.18632/oncotarget.6847] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 12/26/2015] [Indexed: 02/07/2023] Open
Abstract
Gliomas are the most common primary brain tumor and one of the most lethal solid tumors. Mechanistic studies into identification of novel biomarkers are needed to develop new therapeutic strategies for this deadly disease. The objective for this study was to explore the potential direct impact of IL-17-IL-17R interaction in gliomas. Immunohistochemistry and flow cytometry analysis of 12 tumor samples obtained from patients with high grade gliomas revealed that a considerable population (2-19%) of cells in all malignant gliomas expressed IL-17RA, with remarkable co-expression of the glioma stem cell (GSC) markers CD133, Nestin, and Sox2. IL-17 enhanced the self-renewal of GSCs as determined by proliferation and Matrigel® colony assays. IL-17 also induced cytokine/chemokine (IL-6, IL-8, interferon-γ-inducible protein [IP-10], and monocyte chemoattractant protein-1 [MCP-1]) secretion in GSCs, which were differentially blocked by antibodies against IL-17R and IL-6R. Western blot analysis showed that IL-17 modulated the activity of signal transducer and activator of transcription 3 (STAT3), nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB), glycogen synthase kinase-3β (GSK-3β) and β-catenin in GSCs. While IL-17R-mediated secretion of IL-6 and IL-8 were significantly blocked by inhibitors of NF-κB and STAT3; NF-κB inhibitor was more potent than STAT3 inhibitor in blocking IL-17-induced MCP-1 secretion. Overall, our results suggest that IL-17-IL-17R interaction in GSCs induces an autocrine/paracrine cytokine feedback loop, which may provide an important signaling component for maintenance/self-renewal of GSCs via constitutive activation of both NF-κB and STAT3. The results also strongly implicate IL-17R as an important functional biomarker for therapeutic targeting of GSCs.
Collapse
Affiliation(s)
- Prahlad Parajuli
- Department of Neurosurgery, Wayne State University and Karmanos Cancer Institute, Detroit, MI, USA
| | - Rohit Anand
- Department of Neurosurgery, Wayne State University and Karmanos Cancer Institute, Detroit, MI, USA
| | | | - Raviteja Suryadevara
- Department of Neurosurgery, Wayne State University and Karmanos Cancer Institute, Detroit, MI, USA
| | - Preethi U. Sriranga
- Department of Neurosurgery, Wayne State University and Karmanos Cancer Institute, Detroit, MI, USA
| | - Sharon K. Michelhaugh
- Department of Neurosurgery, Wayne State University and Karmanos Cancer Institute, Detroit, MI, USA
| | - Simona Cazacu
- Hermelin Brain Tumor Center, Henry Ford Hospital, Detroit, MI, USA
| | - Susan Finniss
- Hermelin Brain Tumor Center, Henry Ford Hospital, Detroit, MI, USA
| | - Archana Thakur
- Department of Oncology, Wayne State University and Karmanos Cancer Institute, Detroit, MI, USA
| | - Lawrence G. Lum
- Department of Oncology, Wayne State University and Karmanos Cancer Institute, Detroit, MI, USA
- Departments of Internal Medicine, Immunology and Microbiology, and Pediatrics, Wayne State University, Detroit, MI, USA
| | - Dana Schalk
- Department of Oncology, Wayne State University and Karmanos Cancer Institute, Detroit, MI, USA
| | - Chaya Brodie
- Hermelin Brain Tumor Center, Henry Ford Hospital, Detroit, MI, USA
| | - Sandeep Mittal
- Department of Neurosurgery, Wayne State University and Karmanos Cancer Institute, Detroit, MI, USA
- Department of Oncology, Wayne State University and Karmanos Cancer Institute, Detroit, MI, USA
| |
Collapse
|
129
|
Glioblastoma Stem Cells Microenvironment: The Paracrine Roles of the Niche in Drug and Radioresistance. Stem Cells Int 2016; 2016:6809105. [PMID: 26880981 PMCID: PMC4736577 DOI: 10.1155/2016/6809105] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 11/09/2015] [Accepted: 11/10/2015] [Indexed: 12/13/2022] Open
Abstract
Among all solid tumors, the high-grade glioma appears to be the most vascularized one. In fact, "microvascular hyperplasia" is a hallmark of GBM. An altered vascular network determines irregular blood flow, so that tumor cells spread rapidly beyond the diffusion distance of oxygen in the tissue, with the consequent formation of hypoxic or anoxic areas, where the bulk of glioblastoma stem cells (GSCs) reside. The response to this event is the induction of angiogenesis, a process mediated by hypoxia inducible factors. However, this new capillary network is not efficient in maintaining a proper oxygen supply to the tumor mass, thereby causing an oxygen gradient within the neoplastic zone. This microenvironment helps GSCs to remain in a "quiescent" state preserving their potential to proliferate and differentiate, thus protecting them by the effects of chemo- and radiotherapy. Recent evidences suggest that responses of glioblastoma to standard therapies are determined by the microenvironment of the niche, where the GSCs reside, allowing a variety of mechanisms that contribute to the chemo- and radioresistance, by preserving GSCs. It is, therefore, crucial to investigate the components/factors of the niche in order to formulate new adjuvant therapies rendering more efficiently the gold standard therapies for this neoplasm.
Collapse
|
130
|
Su S, Hong F, Liang Y, Zhou J, Liang Y, Chen K, Wang X, Wang Z, Wang Z, Chang C, Han W, Gong W, Qin H, Jiang B, Xiong H, Peng L. Lgr5 Methylation in Cancer Stem Cell Differentiation and Prognosis-Prediction in Colorectal Cancer. PLoS One 2015; 10:e0143513. [PMID: 26599100 PMCID: PMC4657969 DOI: 10.1371/journal.pone.0143513] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 11/05/2015] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE Leucine-rich-repeat-containing G-protein-coupled receptor 5 (lgr5) is a candidate marker for colorectal cancer stem cells (CSC). In the current study, we investigated the methylation status within thelgr5 promoter and evaluated its relationship with CSC differentiation, prognosis for colorectal cancer, and its clinicopathological features. METHODS The methylation status within Lgr5 promoter was detected with a methylation-specific PCR in six colorectal cancer cell lines as well as 169 primary colorectal tumor tissues. Differentiation of CSC was examined with immunofluorescence and immunocytochemistry. Down-regulation of lgr5 was achieved with gene-specific siRNA. The associations between lgr5 methylation and the clinicopathological features as well as survival of patients were analyzed with statistical methods. RESULTS The lgr5 promoter was methylated to different degrees for the six colorectal cell lines examined, with complete methylation observed in HCT116 cells in which the lgr5 expression was partially recovered following DAC treatment. The stem-cell sphere formation from HCT116 cells was accompanied by increasing methylation within the lgr5 promoter and decreasing expression of lgr5. Knocking down lgr5 by siRNA also led to stem-cell spheres formation. Among primary colorectal tumors, 40% (67/169) were positive for lgr5 methylation, while none of the normal colon tissues were positive for lgr5 methylation. Furthermore, lgr5 methylation significantly associated with higher tumor grade, and negative distant metastasis (p < 0.05), as well as better prognosis (p = 0.001) in patients with colorectal cancer. CONCLUSIONS Our data suggests that lgr5 methylation, through the regulation of lgr5 expression and colorectal CSC differentiation, may constitute a novel prognostic marker for colorectal cancer patients.
Collapse
Affiliation(s)
- Shasha Su
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Feng Hong
- Institute of liver diseases, Affiliated Hospital of Jining Medical University, Shandong, 273100, China
| | - Yanling Liang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jieqiong Zhou
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yan Liang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Kequan Chen
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xinying Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhongqiu Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhiqing Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Cassie Chang
- Immunology Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, New York 10029, United States of America
| | - Weihua Han
- Second affiliated hospital of XingTai medical college, Hebei, 054002, China
| | - Wei Gong
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Haitao Qin
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Bo Jiang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Huabao Xiong
- Immunology Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, New York 10029, United States of America
| | - Liang Peng
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- * E-mail:
| |
Collapse
|
131
|
Yang L, Liu Z, Wu R, Yao Q, Gu Z, Liu M. Correlation of C-X-C chemokine receptor 2 upregulation with poor prognosis and recurrence in human glioma. Onco Targets Ther 2015; 8:3203-9. [PMID: 26586954 PMCID: PMC4636088 DOI: 10.2147/ott.s91626] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
C-X-C chemokine receptor 2 (CXCR2), a member of the G-protein-coupled receptor family, is an interleukin-8 receptor and results in the activation of neutrophils. To date, CXCR2 has been identified with many cell events, including inflammation, neovascularization, metastasis, and cell carcinogenesis. This study aimed to investigate alterations in the expression of CXCR2 in patients with brain gliomas and relationships with pathological grades and clinicopathological characteristics. Brain tissue specimens from 60 patients with glioma and 15 patients undergoing surgery for epilepsy (controls) were detected using streptavidin-perosidase immunohistochemistry. Western blotting was used to evaluate CXCR2 protein levels with fresh tissues derived from glioma cases or controls. Correlations between CXCR2 expression and clinicopathological characteristics were analyzed using SPSS software. The results showed high-grade gliomas with high CXCR2 expression as compared with normal tissues. The expression of CXCR2 was significantly related to high grades and recurrence of tumor but not to age or sex. During an in vitro wound healing assay, U251 migration was reduced when the CXCR2-specific inhibitor SB225002 was applied. Our results suggested that the high expression of CXCR2 in gliomas was closely correlated to the degree of malignancy and recurrence and that CXCR2 inhibition decreased the migration of glioma cells. Therefore, CXCR2 may serve as a potential therapeutic target for the recurrence and migration of gliomas.
Collapse
Affiliation(s)
- Liu Yang
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Zenghui Liu
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Ronghua Wu
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Qi Yao
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Zhikai Gu
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Mei Liu
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, People's Republic of China
| |
Collapse
|
132
|
Kast RE. Erlotinib augmentation with dapsone for rash mitigation and increased anti-cancer effectiveness. SPRINGERPLUS 2015; 4:638. [PMID: 26543772 PMCID: PMC4628020 DOI: 10.1186/s40064-015-1441-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 10/14/2015] [Indexed: 01/10/2023]
Abstract
BACKGROUND The epidermal growth factor receptor tyrosine kinase inhibitor erlotinib has failed in many ways to be as potent in the anti-cancer role as pre-clinical studies would have suggested. This paper traces some aspects of this failure to a compensatory erlotinib-mediated increase in interleukin-8. Many other-but not all- cancer chemotherapeutic cytotoxic drugs also provoke a compensatory increase in a malignant clone's interleukin-8 synthesis. Untreated glioblastoma and other cancer cells themselves natively synthesize interleukin-8. Interleukin-8 has tumor growth promoting, mobility and metastasis formation enhancing, effects as well as pro-angiogenesis effects. FINDINGS The old sulfone antibiotic dapsone- one of the very first antibiotics in clinical use- has demonstrated several interleukin-8 system inhibiting actions. Review of these indicates dapsone has potential to augment erlotinib effectiveness. Erlotinib typically gives a rash that has recently been proven to come about via an erlotinib triggered up-regulated keratinocyte interleukin-8 synthesis. The erlotinib rash shares histological features reminiscent of typical neutrophilic dermatoses. Dapsone has an established therapeutic role in current treatment of other neutrophilic dermatoses. CONCLUSION Thus, dapsone has potential to both improve the quality of life in erlotinib treated patients by amelioration of rash as well as to short-circuit a growth-enhancing aspect of erlotinib when used in the anti-cancer role.
Collapse
Affiliation(s)
- R E Kast
- IIAIGC Study Center, 22 Church Street, Burlington, VT 05401 USA
| |
Collapse
|
133
|
Albini A, Bruno A, Gallo C, Pajardi G, Noonan DM, Dallaglio K. Cancer stem cells and the tumor microenvironment: interplay in tumor heterogeneity. Connect Tissue Res 2015; 56:414-25. [PMID: 26291921 PMCID: PMC4673538 DOI: 10.3109/03008207.2015.1066780] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Tumor cells able to recapitulate tumor heterogeneity have been tracked, isolated and characterized in different tumor types, and are commonly named Cancer Stem Cells or Cancer Initiating Cells (CSC/CIC). CSC/CIC are disseminated in the tumor mass and are resistant to anti-cancer therapies and adverse conditions. They are able to divide into another stem cell and a "proliferating" cancer cell. They appear to be responsible for disease recurrence and metastatic dissemination even after apparent eradication of the primary tumor. The modulation of CSC/CIC activities by the tumor microenvironment (TUMIC) is still poorly known. CSC/CIC may mutually interact with the TUMIC in a special and unique manner depending on the TUMIC cells or proteins encountered. The TUMIC consists of extracellular matrix components as well as cellular players among which endothelial, stromal and immune cells, providing and responding to signals to/from the CSC/CIC. This interplay can contribute to the mechanisms through which CSC/CIC may reside in a dormant state in a tissue for years, later giving rise to tumor recurrence or metastasis in patients. Different TUMIC components, including the connective tissue, can differentially activate CIC/CSC in different areas of a tumor and contribute to the generation of cancer heterogeneity. Here, we review possible networking activities between the different components of the tumor microenvironment and CSC/CIC, with a focus on its role in tumor heterogeneity and progression. We also summarize novel therapeutic options that could target both CSC/CIC and the microenvironment to elude resistance mechanisms activated by CSC/CIC, responsible for disease recurrence and metastases.
Collapse
Affiliation(s)
- Adriana Albini
- Department of Research and Statistics, IRCCS Arcispedale Santa Maria Nuova,
Reggio Emilia,
Italy,Correspondence: Adriana Albini, Director of the Department of Research and Statistics,
IRCCS Arcispedale Santa Maria Nuova, Reggio Emilia, Viale Risorgimento 80, 42123,
Reggio Emilia,
Italy. E-mail:
| | - Antonino Bruno
- Scientific and Technology Park, IRCCS MultiMedica,
Milan,
Italy
| | - Cristina Gallo
- Department of Research and Statistics, IRCCS Arcispedale Santa Maria Nuova,
Reggio Emilia,
Italy
| | - Giorgio Pajardi
- Department of Hand Surgery, San Giuseppe MultiMedica Hospital of Milan,
Milan,
Italy,Department of Clinical Sciences and Community, Plastic Surgery School, University of Milan,
Milan,
Italy
| | - Douglas M. Noonan
- Scientific and Technology Park, IRCCS MultiMedica,
Milan,
Italy,Department of Biotechnology and Life Sciences, University of Insubria,
Varese,
Italy
| | - Katiuscia Dallaglio
- Department of Research and Statistics, IRCCS Arcispedale Santa Maria Nuova,
Reggio Emilia,
Italy
| |
Collapse
|
134
|
Mendes B, Marques C, Carvalho I, Costa P, Martins S, Ferreira D, Sarmento B. Influence of glioma cells on a new co-culture in vitro blood–brain barrier model for characterization and validation of permeability. Int J Pharm 2015; 490:94-101. [PMID: 25981617 DOI: 10.1016/j.ijpharm.2015.05.027] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Revised: 05/07/2015] [Accepted: 05/09/2015] [Indexed: 01/11/2023]
|
135
|
Zhou W, Jiang Z, Li X, Xu Y, Shao Z. Cytokines: shifting the balance between glioma cells and tumor microenvironment after irradiation. J Cancer Res Clin Oncol 2015; 141:575-89. [PMID: 25005789 DOI: 10.1007/s00432-014-1772-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 06/30/2014] [Indexed: 12/13/2022]
Abstract
Malignant gliomas invariably recur after irradiation, showing radioresistance. Meanwhile, cranial irradiation can bring some risk for developing cognitive dysfunction. There is increasing evidence that cytokines play their peculiar roles in these processes. On the one hand, cytokines directly influence the progression of malignant glioma, promoting or suppressing tumor progression. On the other hand, cytokines indirectly contribute to the immunologic response against gliomas, exhibiting pro-inflammatory or immunosuppressive activities. We propose that cytokines are not simply unregulated products from tumor cells or immune cells, but mediators finely adjust the balance between glioma cells and tumor microenvironment after irradiation. The paper, therefore, focuses on the changes of cytokines after irradiation, analyzing how these mediate the response of tumor cells and normal cells to irradiation. In addition, cytokine-based immunotherapeutic strategies, accompanied with irradiation, for the treatment of gliomas are also discussed.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Radiation Oncology, Cancer Centre, Qilu Hospital, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, Shandong, China
| | | | | | | | | |
Collapse
|
136
|
IL8-CXCR2 pathway inhibition as a therapeutic strategy against MDS and AML stem cells. Blood 2015; 125:3144-52. [PMID: 25810490 DOI: 10.1182/blood-2015-01-621631] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/15/2015] [Indexed: 12/16/2022] Open
Abstract
Acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) are associated with disease-initiating stem cells that are not eliminated by conventional therapies. Novel therapeutic targets against preleukemic stem cells need to be identified for potentially curative strategies. We conducted parallel transcriptional analysis of highly fractionated stem and progenitor populations in MDS, AML, and control samples and found interleukin 8 (IL8) to be consistently overexpressed in patient samples. The receptor for IL8, CXCR2, was also significantly increased in MDS CD34(+) cells from a large clinical cohort and was predictive of increased transfusion dependence. High CXCR2 expression was also an adverse prognostic factor in The Cancer Genome Atlas AML cohort, further pointing to the critical role of the IL8-CXCR2 axis in AML/MDS. Functionally, CXCR2 inhibition by knockdown and pharmacologic approaches led to a significant reduction in proliferation in several leukemic cell lines and primary MDS/AML samples via induction of G0/G1 cell cycle arrest. Importantly, inhibition of CXCR2 selectively inhibited immature hematopoietic stem cells from MDS/AML samples without an effect on healthy controls. CXCR2 knockdown also impaired leukemic growth in vivo. Together, these studies demonstrate that the IL8 receptor CXCR2 is an adverse prognostic factor in MDS/AML and is a potential therapeutic target against immature leukemic stem cell-enriched cell fractions in MDS and AML.
Collapse
|
137
|
[The stem cell niche in glioblastoma: from fundamental aspects to targeted therapies]. Bull Cancer 2015; 102:24-33. [PMID: 25609493 DOI: 10.1016/j.bulcan.2014.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Accepted: 07/21/2014] [Indexed: 12/11/2022]
Abstract
The concept of cancer stem cell (CSC) was established from models of leukemogenesis explaining tumor repopulation by the clonogenic properties of this specific population of tumoral cells. Among solid tumors, glioblastoma are currently the most documented models. Cancer stem cells reside in specific locations within tumors called niches. Anatomically, two complementary niches have been described in glioblastoma. The first one is a perivascular niche composed of vessels (endothelial cells, pericytes) and their microenvironment (integrins, interleukins) constitutive the nest of "normal" neural stem cells and cancer stem cells. The second one is a hypoxic niche found in regions with low oxygen tension such as the core of the tumor. In these niches, mutual interactions between CSC and their microenvironment involving the activation of multiple signaling pathways promote stemness maintenance and tumor propagation. The median overall survival of glioblastoma does not exceed 15 months despite an aggressive multimodal treatment, thus the therapeutic targeting of these niches, by systemic agents or radiotherapy, in order to inhibit the signaling pathways involved in the maintenance of the CSC niches, represents a major challenge. The combination of these two strategies appears promising and many clinical trials are underway.
Collapse
|
138
|
Guo H, Liu C, Yang L, Dong L, Wang L, Wang Q, Li H, Zhang J, Lin P, Wang X. Morusin inhibits glioblastoma stem cell growth in vitro and in vivo through stemness attenuation, adipocyte transdifferentiation, and apoptosis induction. Mol Carcinog 2014; 55:77-89. [PMID: 25557841 DOI: 10.1002/mc.22260] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Revised: 10/27/2014] [Accepted: 11/03/2014] [Indexed: 02/05/2023]
Affiliation(s)
- Huijie Guo
- Laboratory of Experimental Oncology; State Key Laboratory of Biotherapy; West China Hospital; West China Clinical Medical School; Sichuan University; Chengdu China
- Department of Immunology; School of Basic Medical Sciences, Chengdu Medical College; Chengdu China
| | - Chuanlan Liu
- Laboratory of Experimental Oncology; State Key Laboratory of Biotherapy; West China Hospital; West China Clinical Medical School; Sichuan University; Chengdu China
| | - Liuqi Yang
- Laboratory of Experimental Oncology; State Key Laboratory of Biotherapy; West China Hospital; West China Clinical Medical School; Sichuan University; Chengdu China
| | - Lihua Dong
- Laboratory of Experimental Oncology; State Key Laboratory of Biotherapy; West China Hospital; West China Clinical Medical School; Sichuan University; Chengdu China
| | - Li Wang
- Laboratory of Experimental Oncology; State Key Laboratory of Biotherapy; West China Hospital; West China Clinical Medical School; Sichuan University; Chengdu China
| | - Qiaoping Wang
- Laboratory of Experimental Oncology; State Key Laboratory of Biotherapy; West China Hospital; West China Clinical Medical School; Sichuan University; Chengdu China
| | - Haiyan Li
- Laboratory of Experimental Oncology; State Key Laboratory of Biotherapy; West China Hospital; West China Clinical Medical School; Sichuan University; Chengdu China
| | - Jie Zhang
- Laboratory of Experimental Oncology; State Key Laboratory of Biotherapy; West China Hospital; West China Clinical Medical School; Sichuan University; Chengdu China
| | - Ping Lin
- Laboratory of Experimental Oncology; State Key Laboratory of Biotherapy; West China Hospital; West China Clinical Medical School; Sichuan University; Chengdu China
| | - Xiujie Wang
- Laboratory of Experimental Oncology; State Key Laboratory of Biotherapy; West China Hospital; West China Clinical Medical School; Sichuan University; Chengdu China
| |
Collapse
|
139
|
Jhaveri N, Chen TC, Hofman FM. Tumor vasculature and glioma stem cells: Contributions to glioma progression. Cancer Lett 2014; 380:545-551. [PMID: 25527451 DOI: 10.1016/j.canlet.2014.12.028] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 12/09/2014] [Accepted: 12/11/2014] [Indexed: 01/13/2023]
Abstract
Glioblastoma multiforme (GBM), the most malignant of brain tumors, is characterized by extensive vascularization and a high degree of invasion. The current standard of care is not very effective, resulting in tumor recurrence with patients rarely surviving over 2 years. This tumor recurrence is attributed to the presence of chemo and radiation resistant glioma stem cells (GSCs). These cells are associated with vascular niches which regulate GSC self-renewal and survival. Recent studies suggest that while blood vessels support glioma stem cells, these tumor cells in turn may regulate and contribute to the tumor vasculature by transdifferentiating into endothelial cells directly or through the secretion of regulatory growth factors such as vascular endothelial growth factor (VEGF) and hepatoma derived growth factor (HDGF). The relationship between the tumor vasculature and the glioma stem cells is the subject of this review.
Collapse
Affiliation(s)
- Niyati Jhaveri
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Thomas C Chen
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA; Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Florence M Hofman
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA; Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
140
|
Rape A, Ananthanarayanan B, Kumar S. Engineering strategies to mimic the glioblastoma microenvironment. Adv Drug Deliv Rev 2014; 79-80:172-83. [PMID: 25174308 PMCID: PMC4258440 DOI: 10.1016/j.addr.2014.08.012] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 04/23/2014] [Accepted: 08/20/2014] [Indexed: 12/12/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common and deadly brain tumor, with a mean survival time of only 21months. Despite the dramatic improvements in our understanding of GBM fueled by recent revolutions in molecular and systems biology, treatment advances for GBM have progressed inadequately slowly, which is due in part to the wide cellular and molecular heterogeneity both across tumors and within a single tumor. Thus, there is increasing clinical interest in targeting cell-extrinsic factors as way of slowing or halting the progression of GBM. These cell-extrinsic factors, collectively termed the microenvironment, include the extracellular matrix, blood vessels, stromal cells that surround tumor cells, and all associated soluble and scaffold-bound signals. In this review, we will first describe the regulation of GBM tumors by these microenvironmental factors. Next, we will discuss the various in vitro approaches that have been exploited to recapitulate and model the GBM tumor microenvironment in vitro. We conclude by identifying future challenges and opportunities in this field, including the development of microenvironmental platforms amenable to high-throughput discovery and screening. We anticipate that these ongoing efforts will prove to be valuable both as enabling tools for accelerating our understanding of microenvironmental regulation in GBM and as foundations for next-generation molecular screening platforms that may serve as a conceptual bridge between traditional reductionist systems and animal or clinical studies.
Collapse
Affiliation(s)
- Andrew Rape
- Department of Bioengineering, University of California-Berkeley, Berkeley, CA, USA
| | | | - Sanjay Kumar
- Department of Bioengineering, University of California-Berkeley, Berkeley, CA, USA.
| |
Collapse
|
141
|
Shamir ER, Ewald AJ. Three-dimensional organotypic culture: experimental models of mammalian biology and disease. Nat Rev Mol Cell Biol 2014; 15:647-64. [PMID: 25237826 PMCID: PMC4352326 DOI: 10.1038/nrm3873] [Citation(s) in RCA: 525] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Mammalian organs are challenging to study as they are fairly inaccessible to experimental manipulation and optical observation. Recent advances in three-dimensional (3D) culture techniques, coupled with the ability to independently manipulate genetic and microenvironmental factors, have enabled the real-time study of mammalian tissues. These systems have been used to visualize the cellular basis of epithelial morphogenesis, to test the roles of specific genes in regulating cell behaviours within epithelial tissues and to elucidate the contribution of microenvironmental factors to normal and disease processes. Collectively, these novel models can be used to answer fundamental biological questions and generate replacement human tissues, and they enable testing of novel therapeutic approaches, often using patient-derived cells.
Collapse
Affiliation(s)
- Eliah R Shamir
- Departments of Cell Biology and Oncology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, 855 North Wolfe Street, Baltimore, Maryland 21205, USA
| | - Andrew J Ewald
- Departments of Cell Biology and Oncology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, 855 North Wolfe Street, Baltimore, Maryland 21205, USA
| |
Collapse
|
142
|
Sanmamed MF, Carranza-Rua O, Alfaro C, Oñate C, Martín-Algarra S, Perez G, Landazuri SF, Gonzalez A, Gross S, Rodriguez I, Muñoz-Calleja C, Rodríguez-Ruiz M, Sangro B, López-Picazo JM, Rizzo M, Mazzolini G, Pascual JI, Andueza MP, Perez-Gracia JL, Melero I. Serum interleukin-8 reflects tumor burden and treatment response across malignancies of multiple tissue origins. Clin Cancer Res 2014; 20:5697-707. [PMID: 25224278 DOI: 10.1158/1078-0432.ccr-13-3203] [Citation(s) in RCA: 195] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE Interleukin-8 (IL8) is a chemokine produced by malignant cells of multiple cancer types. It exerts various functions in shaping protumoral vascularization and inflammation/immunity. We evaluated sequential levels of serum IL8 in preclinical tumor models and in patients to assess its ability to estimate tumor burden. EXPERIMENTAL DESIGN IL8 levels were monitored by sandwich ELISAs in cultured tumor cells supernatants, tumor-xenografted mice serum, and in samples from 126 patients with cancer. We correlated IL8 serum levels with baseline tumor burden and with treatment-induced changes in tumor burden, as well as with prognosis. RESULTS IL8 concentrations correlated with the number of IL8-producing tumor cells in culture. In xenografted neoplasms, IL8 serum levels rapidly dropped after surgical excision, indicating an accurate correlation with tumor burden. In patients with melanoma (n = 16), renal cell carcinoma (RCC; n = 23), non-small cell lung cancer (NSCLC; n = 21), or hepatocellular carcinoma (HCC; n = 30), serum IL8 concentrations correlated with tumor burden and stage, survival (melanoma, n = 16; RCC, n = 23; HCC, n = 33), and objective responses to therapy, including those to BRAF inhibitors (melanoma, n = 16) and immunomodulatory monoclonal antibodies (melanoma, n = 8). IL8 concentrations in urine (n = 18) were mainly elevated in tumors with direct contact with the urinary tract. CONCLUSIONS IL8 levels correlate with tumor burden in preclinical models and in patients with cancer. IL8 is a potentially useful biomarker to monitor changes in tumor burden following anticancer therapy, and has prognostic significance.
Collapse
Affiliation(s)
- Miguel F Sanmamed
- Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain. Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Omar Carranza-Rua
- Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Carlos Alfaro
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Carmen Oñate
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | | | - Guiomar Perez
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Sara F Landazuri
- Department of Biochemistry, Clínica Universidad de Navarra, Pamplona, Spain
| | - Alvaro Gonzalez
- Department of Biochemistry, Clínica Universidad de Navarra, Pamplona, Spain
| | - Stefanie Gross
- Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Inmaculada Rodriguez
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | | | - María Rodríguez-Ruiz
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Bruno Sangro
- Liver Unit, Clínica Universidad de Navarra, Pamplona, Spain. Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Pamplona, Spain
| | | | - Manglio Rizzo
- Gene Therapy Laboratory, Department of Medicine, Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Argentina
| | - Guillermo Mazzolini
- Gene Therapy Laboratory, Department of Medicine, Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Argentina
| | - Juan I Pascual
- Department of Urology, Clínica Universidad de Navarra, Pamplona, Spain
| | | | | | - Ignacio Melero
- Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain. Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain.
| |
Collapse
|
143
|
Zhou J, Yi L, Ouyang Q, Xu L, Cui H, Xu M. Neurotensin signaling regulates stem-like traits of glioblastoma stem cells through activation of IL-8/CXCR1/STAT3 pathway. Cell Signal 2014; 26:2896-902. [PMID: 25200966 DOI: 10.1016/j.cellsig.2014.08.027] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 08/29/2014] [Indexed: 11/26/2022]
Abstract
We recently found that neurotensin (NTS) and its primary receptor NTSR1 play a crucial role in glioblastoma cell proliferation and invasion. However, very little is known regarding the functional role of NTS/NTSR1 signaling in glioblastoma stem cells (GSCs). Here, we showed that NTSR1 is highly expressed in GSCs than its non-GSC counterparts. Pharmacological blockade with SR48692 or lentivirus mediated knockdown of NTSR1 efficiently reduced the sphere-forming ability and expression of stem cell markers such as nestin and Sox2 in GSCs isolated from glioblastoma cell line and glioblastoma tissues. Conversely, treated GSCs with NTS led to increase of tumor sphere formation. Mechanistically, we demonstrated that EGFR-dependent enhancement of IL-8 secretion is responsible for the effect of NTS signaling in the regulation of stem-like traits. Finally, we showed that NTSR1 or IL-8 knockdown decreased the phosphorylation of transcriptional factor STAT3 at Tyr705, which is a major transcription factor implicated in the regulation of GSC stem-like traits. Although both CXCR1 and CXCR2 inhibition reduced the tumor sphere formation, we found that CXCR1, but not CXCR2, is primarily responsible for STAT3 phosphorylation. Taken together, our findings suggest that NTS/IL-8/CXCR1/STAT3 signaling is crucial for the maintenance of stem-like traits in GSCs and provides a potential therapeutic target for glioblastoma therapy.
Collapse
Affiliation(s)
- Ji Zhou
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Liang Yi
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Qing Ouyang
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Lunshan Xu
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Institute of Sericulture and Systems Biology, Southwest University, Chongqing 400716, China.
| | - Minhui Xu
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China.
| |
Collapse
|
144
|
Garcia C, Gutmann DH. Using the neurofibromatosis tumor predisposition syndromes to understand normal nervous system development. SCIENTIFICA 2014; 2014:915725. [PMID: 25243094 PMCID: PMC4163293 DOI: 10.1155/2014/915725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 05/07/2014] [Indexed: 06/03/2023]
Abstract
Development is a tightly regulated process that involves stem cell self-renewal, differentiation, cell-to-cell communication, apoptosis, and blood vessel formation. These coordinated processes ensure that tissues maintain a size and architecture that is appropriate for normal tissue function. As such, tumors arise when cells acquire genetic mutations that allow them to escape the normal growth constraints. In this regard, the study of tumor predisposition syndromes affords a unique platform to better understand normal development and the process by which normal cells transform into cancers. Herein, we review the processes governing normal brain development, discuss how brain cancer represents a disruption of these normal processes, and highlight insights into both normal development and cancer made possible by the study of tumor predisposition syndromes.
Collapse
Affiliation(s)
- Cynthia Garcia
- Department of Neurology, Washington University School of Medicine, Box 8111, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - David H. Gutmann
- Department of Neurology, Washington University School of Medicine, Box 8111, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| |
Collapse
|
145
|
Orr BA, Eberhart CG. Molecular pathways: not a simple tube--the many functions of blood vessels. Clin Cancer Res 2014; 21:18-23. [PMID: 25074609 DOI: 10.1158/1078-0432.ccr-13-1641] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Although the ability of blood vessels to carry fluid and cells through neoplastic tissue is clearly important, other functions of vascular elements that drive tumor growth and progression are increasingly being recognized. Vessels can provide physical support and help regulate the stromal microenvironment within tumors, form niches for tumor-associated stem cells, serve as avenues for local tumor spread, and promote relative immune privilege. Understanding the molecular drivers of these phenotypes will be critical if we are to therapeutically target their protumorigenic effects. The potential for neoplastic cells to transdifferentiate into vascular and perivascular elements also needs to be better understood, as it has the potential to complicate such therapies. In this review, we provide a brief overview of these less conventional vascular functions in tumors.
Collapse
Affiliation(s)
- Brent A Orr
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Charles G Eberhart
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland. Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland. Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
146
|
Zhou J, Xiang Y, Yoshimura T, Chen K, Gong W, Huang J, Zhou Y, Yao X, Bian X, Wang JM. The role of chemoattractant receptors in shaping the tumor microenvironment. BIOMED RESEARCH INTERNATIONAL 2014; 2014:751392. [PMID: 25110692 PMCID: PMC4119707 DOI: 10.1155/2014/751392] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/17/2014] [Indexed: 12/13/2022]
Abstract
Chemoattractant receptors are a family of seven transmembrane G protein coupled receptors (GPCRs) initially found to mediate the chemotaxis and activation of immune cells. During the past decades, the functions of these GPCRs have been discovered to not only regulate leukocyte trafficking and promote immune responses, but also play important roles in homeostasis, development, angiogenesis, and tumor progression. Accumulating evidence indicates that chemoattractant GPCRs and their ligands promote the progression of malignant tumors based on their capacity to orchestrate the infiltration of the tumor microenvironment by immune cells, endothelial cells, fibroblasts, and mesenchymal cells. This facilitates the interaction of tumor cells with host cells, tumor cells with tumor cells, and host cells with host cells to provide a basis for the expansion of established tumors and development of distant metastasis. In addition, many malignant tumors of the nonhematopoietic origin express multiple chemoattractant GPCRs that increase the invasiveness and metastasis of tumor cells. Therefore, GPCRs and their ligands constitute targets for the development of novel antitumor therapeutics.
Collapse
Affiliation(s)
- Jiamin Zhou
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
- Endoscopic Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yi Xiang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
- Department of Pulmonary Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Teizo Yoshimura
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Keqiang Chen
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Wanghua Gong
- Basic Research Program, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Jian Huang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ye Zhou
- Department of Gastric Cancer and Soft Tissue Surgery, Fudan University Cancer Center, Shanghai 200032, China
| | - Xiaohong Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ji Ming Wang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
147
|
Berezovsky AD, Poisson LM, Cherba D, Webb CP, Transou AD, Lemke NW, Hong X, Hasselbach LA, Irtenkauf SM, Mikkelsen T, deCarvalho AC. Sox2 promotes malignancy in glioblastoma by regulating plasticity and astrocytic differentiation. Neoplasia 2014; 16:193-206, 206.e19-25. [PMID: 24726753 DOI: 10.1016/j.neo.2014.03.006] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 02/20/2014] [Accepted: 02/24/2014] [Indexed: 01/10/2023] Open
Abstract
The high-mobility group-box transcription factor sex-determining region Y-box 2 (Sox2) is essential for the maintenance of stem cells from early development to adult tissues. Sox2 can reprogram differentiated cells into pluripotent cells in concert with other factors and is overexpressed in various cancers. In glioblastoma (GBM), Sox2 is a marker of cancer stemlike cells (CSCs) in neurosphere cultures and is associated with the proneural molecular subtype. Here, we report that Sox2 expression pattern in GBM tumors and patient-derived mouse xenografts is not restricted to a small percentage of cells and is coexpressed with various lineage markers, suggesting that its expression extends beyond CSCs to encompass more differentiated neoplastic cells across molecular subtypes. Employing a CSC derived from a patient with GBM and isogenic differentiated cell model, we show that Sox2 knockdown in the differentiated state abolished dedifferentiation and acquisition of CSC phenotype. Furthermore, Sox2 deficiency specifically impaired the astrocytic component of a biphasic gliosarcoma xenograft model while allowing the formation of tumors with sarcomatous phenotype. The expression of genes associated with stem cells and malignancy were commonly downregulated in both CSCs and serum-differentiated cells on Sox2 knockdown. Genes previously shown to be associated with pluripontency and CSCs were only affected in the CSC state, whereas embryonic stem cell self-renewal genes and cytokine signaling were downregulated, and the Wnt pathway activated in differentiated Sox2-deficient cells. Our results indicate that Sox2 regulates the expression of key genes and pathways involved in GBM malignancy, in both cancer stemlike and differentiated cells, and maintains plasticity for bidirectional conversion between the two states, with significant clinical implications.
Collapse
Affiliation(s)
| | - Laila M Poisson
- Department of Public Health Sciences, Henry Ford Hospital, Detroit, MI
| | - David Cherba
- Program of Translational Medicine, Van Andel Research Institute, Grand Rapids, MI
| | - Craig P Webb
- Program of Translational Medicine, Van Andel Research Institute, Grand Rapids, MI
| | | | - Nancy W Lemke
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI
| | - Xin Hong
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI
| | | | | | - Tom Mikkelsen
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI; Department of Neurology, Henry Ford Hospital, Detroit, MI
| | | |
Collapse
|