Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Exp Clin Cancer Res ; 43(1): 98, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38561856

ABSTRACT

BACKGROUND: The complement inhibitor CSMD1 acts as a tumor suppressor in various types of solid cancers. Despite its high level of expression in the brain, its function in gliomas, malignant brain tumors originating from glial cells, has not been investigated. METHODS: Three cohorts of glioma patients comprising 1500 patients were analyzed in our study along with their clinical data. H4, U-118 and U-87 cell lines were used to investigate the tumor suppressor function of CSMD1 in gliomas. PDGFB-induced brain tumor model was utilized for the validation of in vitro data. RESULTS: The downregulation of CSMD1 expression correlated with reduced overall and disease-free survival, elevated tumor grade, wild-type IDH genotype, and intact 1p/19q status. Moreover, enhanced activity was noted in the neuroinflammation pathway. Importantly, ectopic expression of CSMD1 in glioma cell lines led to decreased aggressiveness in vitro. Mechanically, CSMD1 obstructed the TNF-induced NF-kB and STAT3 signaling pathways, effectively suppressing the secretion of IL-6 and IL-8. There was also reduced survival in PDGFB-induced brain tumors in mice when Csmd1 was downregulated. CONCLUSIONS: Our study has identified CSMD1 as a tumor suppressor in gliomas and elucidated its role in TNF-induced neuroinflammation, contributing to a deeper understanding of glioma pathogenesis.


Subject(s)
Brain Neoplasms , Glioma , Humans , Animals , Mice , Neuroinflammatory Diseases , Proto-Oncogene Proteins c-sis/genetics , Glioma/pathology , Brain Neoplasms/pathology , Disease-Free Survival , Isocitrate Dehydrogenase/genetics , Mutation , Membrane Proteins/genetics , Tumor Suppressor Proteins/genetics
3.
Semin Cancer Biol ; 86(Pt 3): 846-856, 2022 11.
Article in English | MEDLINE | ID: mdl-35143991

ABSTRACT

Cancer cells reside amongst a complex milieu of stromal cells and structural features known as the tumor microenvironment. Often cancer cells divert and co-opt functions of stromal cells of the microenvironment to support tumor progression and treatment resistance. During therapy targeting cancer cells, the stromal cells of the microenvironment receive therapy to the same extent as cancer cells. Stromal cells therefore activate a variety of responses to the damage induced by these therapies, and some of those responses may support tumor progression and resistance. We review here the response of stromal cells to cancer therapy with a focus on radiotherapy in glioblastoma. We highlight the response of endothelial cells and the vasculature, macrophages and microglia, and astrocytes, as well as describing resulting changes in the extracellular matrix. We emphasize the complex interplay of these cellular factors in their dynamic responses. Finally, we discuss their resulting support of cancer cells in tumor progression and therapy resistance. Understanding the stromal cell response to therapy provides insight into complementary therapeutic targets to enhance tumor response to existing treatment options.


Subject(s)
Glioblastoma , Tumor Microenvironment , Humans , Endothelial Cells , Stromal Cells , Extracellular Matrix
4.
EMBO J ; 41(7): e110819, 2022 04 04.
Article in English | MEDLINE | ID: mdl-35199359

ABSTRACT

Context-specific control mechanisms of hypoxia-inducible transcription factors HIF-1alpha and HIF-2alpha in tumors exposed to oxygen shortage remain incompletely understood. In this issue, Zhang et al (2022) identify a deubiquitinase that differentially stabilizes HIF-2alpha in stem-like glioblastoma cells, suggesting potential implications for regulation of the hypoxic response in a wide array of tissues and cancers.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Glioblastoma , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Hypoxia/physiology , Deubiquitinating Enzymes , Glioblastoma/genetics , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Protein Isoforms
5.
J Histochem Cytochem ; 70(1): 17-28, 2022 01.
Article in English | MEDLINE | ID: mdl-34606325

ABSTRACT

DLK1 is a maternally imprinted, paternally expressed gene coding for the transmembrane protein Delta-like homologue 1 (DLK1), a non-canonical NOTCH ligand with well-described roles during development, and tumor-supportive functions in several aggressive cancer forms. Here, we review the many functions of DLK1 as a regulator of stem cell pools and tissue differentiation in tissues such as brain, muscle, and liver. Furthermore, we review recent evidence supporting roles for DLK1 in the maintenance of aggressive stem cell characteristics of tumor cells, specifically focusing on central nervous system tumors, neuroblastoma, and hepatocellular carcinoma. We discuss NOTCH -dependent as well as NOTCH-independent functions of DLK1, and focus particularly on the complex pattern of DLK1 expression and cleavage that is finely regulated from a spatial and temporal perspective. Progress in recent years suggest differential functions of extracellular, soluble DLK1 as a paracrine stem cell niche-secreted factor, and has revealed a role for the intracellular domain of DLK1 in cell signaling and tumor stemness. A better understanding of DLK1 regulation and signaling may enable therapeutic targeting of cancer stemness by interfering with DLK1 release and/or intracellular signaling.


Subject(s)
Calcium-Binding Proteins/metabolism , Membrane Proteins/metabolism , Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Stem Cell Niche , Calcium-Binding Proteins/genetics , Humans , Membrane Proteins/genetics , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Signal Transduction
6.
Cells ; 10(3)2021 03 10.
Article in English | MEDLINE | ID: mdl-33802060

ABSTRACT

Glioblastoma is characterized by extensive necrotic areas with surrounding hypoxia. The cancer cell response to hypoxia in these areas is well-described; it involves a metabolic shift and an increase in stem cell-like characteristics. Less is known about the hypoxic response of tumor-associated astrocytes, a major component of the glioma tumor microenvironment. Here, we used primary human astrocytes and a genetically engineered glioma mouse model to investigate the response of this stromal cell type to hypoxia. We found that astrocytes became reactive in response to intermediate and severe hypoxia, similarly to irradiated and temozolomide-treated astrocytes. Hypoxic astrocytes displayed a potent hypoxia response that appeared to be driven primarily by hypoxia-inducible factor 2-alpha (HIF-2α). This response involved the activation of classical HIF target genes and the increased production of hypoxia-associated cytokines such as TGF-ß1, IL-3, angiogenin, VEGF-A, and IL-1 alpha. In vivo, astrocytes were present in proximity to perinecrotic areas surrounding HIF-2α expressing cells, suggesting that hypoxic astrocytes contribute to the glioma microenvironment. Extracellular matrix derived from hypoxic astrocytes increased the proliferation and drug efflux capability of glioma cells. Together, our findings suggest that hypoxic astrocytes are implicated in tumor growth and potentially stemness maintenance by remodeling the tumor microenvironment.


Subject(s)
Astrocytes/metabolism , Glioma/physiopathology , Animals , Cell Hypoxia , Humans , Mice , Tumor Microenvironment
7.
Cancer Res ; 81(8): 2101-2115, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33483373

ABSTRACT

The tumor microenvironment plays an essential role in supporting glioma stemness and radioresistance. Following radiotherapy, recurrent gliomas form in an irradiated microenvironment. Here we report that astrocytes, when pre-irradiated, increase stemness and survival of cocultured glioma cells. Tumor-naïve brains increased reactive astrocytes in response to radiation, and mice subjected to radiation prior to implantation of glioma cells developed more aggressive tumors. Extracellular matrix derived from irradiated astrocytes were found to be a major driver of this phenotype and astrocyte-derived transglutaminase 2 (TGM2) was identified as a promoter of glioma stemness and radioresistance. TGM2 levels increased after radiation in vivo and in recurrent human glioma, and TGM2 inhibitors abrogated glioma stemness and survival. These data suggest that irradiation of the brain results in the formation of a tumor-supportive microenvironment. Therapeutic targeting of radiation-induced, astrocyte-derived extracellular matrix proteins may enhance the efficacy of standard-of-care radiotherapy by reducing stemness in glioma. SIGNIFICANCE: These findings presented here indicate that radiotherapy can result in a tumor-supportive microenvironment, the targeting of which may be necessary to overcome tumor cell therapeutic resistance and recurrence. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/8/2101/F1.large.jpg.


Subject(s)
Astrocytes/enzymology , Brain Neoplasms/radiotherapy , Brain/radiation effects , GTP-Binding Proteins/metabolism , Glioblastoma/radiotherapy , Neoplastic Stem Cells , Transglutaminases/metabolism , Tumor Microenvironment/radiation effects , Animals , Astrocytes/radiation effects , Brain/cytology , Brain/physiology , Brain Neoplasms/pathology , Cell Survival/physiology , Enzyme Inhibitors/pharmacology , Extracellular Matrix/metabolism , Extracellular Matrix/radiation effects , Female , GTP-Binding Proteins/antagonists & inhibitors , Glioblastoma/pathology , Glioma/pathology , Glioma/radiotherapy , Humans , Male , Mice , Neoplasm Recurrence, Local/enzymology , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/physiology , Protein Glutamine gamma Glutamyltransferase 2 , Radiation Tolerance , Transglutaminases/antagonists & inhibitors , Tumor Microenvironment/physiology
8.
Neoplasia ; 22(12): 689-701, 2020 12.
Article in English | MEDLINE | ID: mdl-33142235

ABSTRACT

Tumor cell behaviors associated with aggressive tumor growth such as proliferation, therapeutic resistance, and stem cell characteristics are regulated in part by soluble factors derived from the tumor microenvironment. Tumor-associated astrocytes represent a major component of the glioma tumor microenvironment, and astrocytes have an active role in maintenance of normal neural stem cells in the stem cell niche, in part via secretion of soluble delta-like noncanonical Notch ligand 1 (DLK1). We found that astrocytes, when exposed to stresses of the tumor microenvironment such as hypoxia or ionizing radiation, increased secretion of soluble DLK1. Tumor-associated astrocytes in a glioma mouse model expressed DLK1 in perinecrotic and perivascular tumor areas. Glioma cells exposed to recombinant DLK1 displayed increased proliferation, enhanced self-renewal and colony formation abilities, and increased levels of stem cell marker genes. Mechanistically, DLK1-mediated effects on glioma cells involved increased and prolonged stabilization of hypoxia-inducible factor 2alpha, and inhibition of hypoxia-inducible factor 2alpha activity abolished effects of DLK1 in hypoxia. Forced expression of soluble DLK1 resulted in more aggressive tumor growth and shortened survival in a genetically engineered mouse model of glioma. Together, our data support DLK1 as a soluble mediator of glioma aggressiveness derived from the tumor microenvironment.


Subject(s)
Biomarkers, Tumor , Calcium-Binding Proteins/metabolism , Glioma/metabolism , Tumor Microenvironment , Animals , Astrocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Calcium-Binding Proteins/genetics , Cell Proliferation , Cell Survival/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Glioma/pathology , Hypoxia , Mice , Mice, Knockout , Tumor Burden
9.
Glia ; 68(12): 2486-2502, 2020 12.
Article in English | MEDLINE | ID: mdl-32621641

ABSTRACT

Radiation therapy is part of the standard of care for gliomas and kills a subset of tumor cells, while also altering the tumor microenvironment. Tumor cells with stem-like properties preferentially survive radiation and give rise to glioma recurrence. Various techniques for enriching and quantifying cells with stem-like properties have been used, including the fluorescence activated cell sorting (FACS)-based side population (SP) assay, which is a functional assay that enriches for stem-like tumor cells. In these analyses, mouse models of glioma have been used to understand the biology of this disease and therapeutic responses, including the radiation response. We present combined SP analysis and single-cell RNA sequencing of genetically-engineered mouse models of glioma to show a time course of cellular response to radiation. We identify and characterize two distinct tumor cell populations that are inherently radioresistant and also distinct effects of radiation on immune cell populations within the tumor microenvironment.


Subject(s)
Brain Neoplasms , Glioma , Stem Cells , Animals , Brain Neoplasms/radiotherapy , Mice , Neoplastic Stem Cells , Single-Cell Analysis , Tumor Microenvironment
10.
Oncogene ; 39(20): 4028-4044, 2020 05.
Article in English | MEDLINE | ID: mdl-32205867

ABSTRACT

Glioblastoma multiforme is characterized in part by severe hypoxia associated with tumor necrosis. The cellular response to hypoxia can influence several properties of tumor cells associated with aggressive tumor growth, including metabolic adaptations and tumor cell migration and invasion. Here, we found that Delta Like Non-Canonical Notch Ligand 1 (DLK1) expression was elevated as compared with normal brain in a genetically engineered mouse model of glioma, and that DLK1 expression increased with tumor grade in human glioma samples. DLK1 expression was highest in hypoxic and perivascular tumor areas, and we found that hypoxia induced the release and nuclear translocation of an intracellular fragment of DLK1 in murine glioma as well as in human glioma cultures. Release of the intracellular fragment was dependent on ADAM17 and Hypoxia-inducible Factor 1alpha and 2alpha (HIF-1alpha/HIF-2alpha), as ADAM17 inhibitors and HIF1A/HIF2A siRNA blocked DLK1 cleavage. Expression of a cleavable form of DLK1 amplified several hypoxia-induced traits of glioma cells such as colony formation, stem cell marker gene expression, a PI3K-pathway-mediated metabolic shift, and enhanced invasiveness. Effects of DLK1 were dependent on DLK1-cleavage by ADAM17, as expression of non-cleavable DLK1 could not replicate the DLK1-induced hypoxic phenotype. Finally, forced expression of DLK1 resulted in more invasive tumor growth in a PDGFB-induced glioma mouse model without affecting overall survival. Together, our findings suggest a previously undescribed role for DLK1 as an intracellular signaling molecule.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Nucleus/metabolism , Glioma/metabolism , Neoplasm Proteins/metabolism , Signal Transduction , Active Transport, Cell Nucleus/genetics , Animals , Calcium-Binding Proteins/genetics , Cell Hypoxia/genetics , Cell Nucleus/genetics , Cell Nucleus/pathology , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/pathology , Mice , Mice, Knockout , Neoplasm Proteins/genetics
11.
Exp Cell Res ; 371(1): 122-129, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30092219

ABSTRACT

Tumor hypoxia is associated with several features of aggressive glioma growth, including migration, invasion, and stemness. Most of the cellular adaptation to hypoxia is mediated by the hypoxia-inducible factors HIF-1α and HIF-2α, but regulation of these factors by both oxygen-dependent and -independent mechanisms in brain tumors is only partially understood. Here, we show that the p75 neurotrophin receptor (p75NTR) is stabilized at hypoxia in murine glioma in vivo, as well as in primary human glioma cultures in vitro. Expression of p75NTR resulted in increased stabilization of HIF-1α and HIF-2α, and RNAi or pharmacologic targeting of p75NTR diminished HIF stabilization and HIF-dependent signaling at hypoxia. Consequentially, p75NTR inhibition resulted in decreased migration, invasion, and stemness in response to hypoxia, suggesting that p75NTR is a central regulator of hypoxia-induced glioma aggressiveness. Together, our findings support the literature that identifies p75NTR as a potential therapeutic target in brain tumors.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glioma/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia/genetics , Receptors, Nerve Growth Factor/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Glioma/metabolism , Glioma/pathology , Humans , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Neoplasm Invasiveness , Neoplasm Transplantation , Primary Cell Culture , Protein Stability , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Nerve Growth Factor/antagonists & inhibitors , Receptors, Nerve Growth Factor/metabolism , Signal Transduction
13.
Cell Rep ; 20(7): 1641-1653, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28813675

ABSTRACT

Hypoxia-inducible factors enhance glioma stemness, and glioma stem cells have an amplified hypoxic response despite residing within a perivascular niche. Still, little is known about differential HIF regulation in stem versus bulk glioma cells. We show that the intracellular domain of stem cell marker CD44 (CD44ICD) is released at hypoxia, binds HIF-2α (but not HIF-1α), enhances HIF target gene activation, and is required for hypoxia-induced stemness in glioma. In a glioma mouse model, CD44 was restricted to hypoxic and perivascular tumor regions, and in human glioma, a hypoxia signature correlated with CD44. The CD44ICD was sufficient to induce hypoxic signaling at perivascular oxygen tensions, and blocking CD44 cleavage decreased HIF-2α stabilization in CD44-expressing cells. Our data indicate that the stem cell marker CD44 modulates the hypoxic response of glioma cells and that the pseudo-hypoxic phenotype of stem-like glioma cells is achieved by stabilization of HIF-2α through interaction with CD44, independently of oxygen.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Hyaluronan Receptors/metabolism , Hypoxia/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Hypoxia , Cell Line, Tumor , Glioma/genetics , Glioma/pathology , Humans , Hyaluronan Receptors/antagonists & inhibitors , Hyaluronan Receptors/genetics , Hypoxia/genetics , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Knockout , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phenotype , Protein Binding , Proteolysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Stem Cell Niche/genetics
14.
Sci Rep ; 6: 25956, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27456282

ABSTRACT

Glioma cells with stem cell traits are thought to be responsible for tumor maintenance and therapeutic failure. Such cells can be enriched based on their inherent drug efflux capability mediated by the ABC transporter ABCG2 using the side population assay, and their characteristics include increased self-renewal, high stem cell marker expression and high tumorigenic capacity in vivo. Here, we show that ABCG2 can actively drive expression of stem cell markers and self-renewal in glioma cells. Stem cell markers and self-renewal was enriched in cells with high ABCG2 activity, and could be specifically inhibited by pharmacological and genetic ABCG2 inhibition. Importantly, despite regulating these key characteristics of stem-like tumor cells, ABCG2 activity did not affect radiation resistance or tumorigenicity in vivo. ABCG2 effects were Notch-independent and mediated by diverse mechanisms including the transcription factor Mef. Our data demonstrate that characteristics of tumor stem cells are separable, and highlight ABCG2 as a potential driver of glioma stemness.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Biomarkers, Tumor/genetics , Brain Neoplasms/pathology , Glioma/pathology , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Animals , Biomarkers, Tumor/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/radiotherapy , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/radiation effects , Glioma/genetics , Glioma/metabolism , Glioma/radiotherapy , Humans , Mice , Neoplasm Transplantation , Neoplastic Stem Cells/radiation effects , Radiation Tolerance , Receptors, Notch/metabolism , Signal Transduction/radiation effects , Up-Regulation
15.
Cell Stem Cell ; 14(3): 357-69, 2014 Mar 06.
Article in English | MEDLINE | ID: mdl-24607407

ABSTRACT

Stem-like glioma cells reside within a perivascular niche and display hallmark radiation resistance. An understanding of the mechanisms underlying these properties will be vital for the development of effective therapies. Here, we show that the stem cell marker CD44 promotes cancer stem cell phenotypes and radiation resistance. In a mouse model of glioma, Cd44(-/-) and Cd44(+/-) animals showed improved survival compared to controls. The CD44 ligand osteopontin shared a perivascular expression pattern with CD44 and promoted glioma stem cell-like phenotypes. These effects were mediated via the γ-secretase-regulated intracellular domain of CD44, which promoted aggressive glioma growth in vivo and stem cell-like phenotypes via CBP/p300-dependent enhancement of HIF-2α activity. In human glioblastoma multiforme, expression of CD44 correlated with hypoxia-induced gene signatures and poor survival. Altogether, these data suggest that in the glioma perivascular niche, osteopontin promotes stem cell-like properties and radiation resistance in adjacent tumor cells via activation of CD44 signaling.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Hyaluronan Receptors/metabolism , Neoplastic Stem Cells/pathology , Osteopontin/metabolism , Signal Transduction , Stem Cell Niche , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , E1A-Associated p300 Protein/metabolism , Glioblastoma/metabolism , Humans , Hyaluronan Receptors/chemistry , Ligands , Mice , Neoplasm Invasiveness , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Phenotype , Platelet-Derived Growth Factor/pharmacology , Protein Structure, Tertiary , Signal Transduction/drug effects , Stem Cell Niche/drug effects , Survival Analysis
17.
Adv Cancer Res ; 112: 255-81, 2011.
Article in English | MEDLINE | ID: mdl-21925307

ABSTRACT

Cancer cells within a given tumor were long regarded as a largely homogeneous group of cells originating from a common progenitor cell. However, it is increasingly appreciated that there is a considerable heterogeneity within tumors also on the tumor cell level. This heterogeneity extends to virtually all measurable properties of cancer cells, ranging from differentiation state, proliferation rate, migratory and invasive capacity to size, and therapeutic response. Such heterogeneity likely represents a major therapeutic hurdle, but the mechanisms underlying its emergence remain poorly understood and a controversial topic. The cancer stem cell model of tumor progression has gained increasing support during the past several years. In this review, I will discuss some major implications of the cancer stem cell hypothesis on the origins of tumor heterogeneity, focusing both on heterogeneity within the tumor cells proper and on potential transdifferentiation of cancer stem cells into stromal and endothelial lineages, as well as on heterogeneity of the therapeutic response. Evidence for and against a direct and causal role of cancer stem cells in the emergence of tumor heterogeneity will be weighed and alternative explanations for apparently contradictory observations discussed. Finally, I will discuss the potential origins of cancer stem cells and the various implications of origin to the contribution to tumor heterogeneity, and outline some future directions.


Subject(s)
Cell Transformation, Neoplastic/pathology , Neoplasms/genetics , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Humans
18.
PLoS One ; 6(7): e20605, 2011.
Article in English | MEDLINE | ID: mdl-21754979

ABSTRACT

BACKGROUND: Gliomas are thought to form by clonal expansion from a single cell-of-origin, and progression-associated mutations to occur in its progeny cells. Glioma progression is associated with elevated growth factor signaling and loss of function of tumor suppressors Ink4a, Arf and Pten. Yet, gliomas are cellularly heterogeneous; they recruit and trap normal cells during infiltration. METHODOLOGY/PRINCIPAL FINDINGS: We performed lineage tracing in a retrovirally mediated, molecularly and histologically accurate mouse model of hPDGFb-driven gliomagenesis. We were able to distinguish cells in the tumor that were derived from the cell-of-origin from those that were not. Phenotypic, tumorigenic and expression analyses were performed on both populations of these cells. Here we show that during progression of hPDGFb-induced murine gliomas, tumor suppressor loss can expand the recruited cell population not derived from the cell-of-origin within glioma microenvironment to dominate regions of the tumor, with essentially no contribution from the progeny of glioma cell-of-origin. Moreover, the recruited cells can give rise to gliomas upon transplantation and passaging, acquire polysomal expression profiles and genetic aberrations typically present in glioma cells rather than normal progenitors, aid progeny cells in glioma initiation upon transplantation, and become independent of PDGFR signaling. CONCLUSIONS/SIGNIFICANCE: These results indicate that non-cell-of-origin derived cells within glioma environment in the mouse can be corrupted to become bona fide tumor, and deviate from the generally established view of gliomagenesis.


Subject(s)
Cell Movement , Cell Transformation, Neoplastic/pathology , Disease Progression , Glioma/pathology , Platelet-Derived Growth Factor/metabolism , AC133 Antigen , Animals , Antigens, CD/metabolism , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glycoproteins/metabolism , Homozygote , Humans , Mice , Neoplasm Transplantation , Peptides/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism
19.
Mol Cancer Res ; 9(5): 626-36, 2011 May.
Article in English | MEDLINE | ID: mdl-21402725

ABSTRACT

Several studies have revealed links between hypoxia and activation of Notch in solid tumors. While most reports have focused on intracellular domain of the Notch1 receptor (icN1) stabilization by direct interaction with HIF proteins, little attention has been given to Notch ligand regulation during hypoxia. Here we show that the Notch ligand JAG2 is transcriptionally activated by hypoxia in a HIF-1α dependent manner. Hypoxic JAG2 induction resulted in elevated Notch activity in tumor cells, as was measured by increased icN1 levels and induction of the Notch target gene HEY1. In primary tumor material, JAG2 expression correlated with vascular development and angiogenesis gene signatures. In line with this, coculture experiments of endothelial cells with hypoxic breast cancer cells displayed a reduction in number of capillary-like tubes formed upon JAG2 siRNA treatment of the breast cancer cells. Together these results suggest that a hypoxic induction of JAG2 in tumor cells mediates a hypoxia-regulated cross-talk between tumor and endothelial cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Breast Neoplasms/genetics , Cell Cycle Proteins/metabolism , Endothelial Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Receptor, Notch1/metabolism , Angiogenic Proteins , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Cell Cycle Proteins/genetics , Cell Hypoxia , Coculture Techniques , Endothelial Cells/pathology , Endothelium, Vascular/metabolism , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Intercellular Signaling Peptides and Proteins/genetics , Jagged-2 Protein , Membrane Proteins/genetics , Mice , Neovascularization, Pathologic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering , Receptor Cross-Talk , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/metabolism
20.
J Proteome Res ; 10(4): 1645-56, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21235201

ABSTRACT

Nontransient hypoxia is strongly associated with malignant lesions, resulting in aggressive behavior and resistance to treatment. We present an analysis of mRNA and protein expression changes in neuroblastoma cell lines occurring upon the transition from normoxia to hypoxia. The correlation between mRNA and protein level changes was poor, although some known hypoxia-driven genes and proteins correlated well. We present previously undescribed membrane proteins expressed under hypoxic conditions that are candidates for evaluation as biomarkers.


Subject(s)
Biomarkers/chemistry , Cell Membrane/chemistry , Hypoxia/metabolism , Membrane Proteins/chemistry , Biomarkers/metabolism , Cell Line, Tumor , Humans , Mass Spectrometry/methods , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microarray Analysis , Neoplasms/chemistry , Neoplasms/metabolism , Neoplasms/pathology , RNA, Messenger/metabolism , Two-Dimensional Difference Gel Electrophoresis/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...