201
|
CX3CR1 promotes recruitment of human glioma-infiltrating microglia/macrophages (GIMs). Exp Cell Res 2010; 316:1553-66. [DOI: 10.1016/j.yexcr.2010.02.018] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Revised: 02/03/2010] [Accepted: 02/16/2010] [Indexed: 11/22/2022]
|
202
|
Lorger M, Felding-Habermann B. Capturing changes in the brain microenvironment during initial steps of breast cancer brain metastasis. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:2958-71. [PMID: 20382702 DOI: 10.2353/ajpath.2010.090838] [Citation(s) in RCA: 228] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Brain metastases are difficult to treat and mostly develop late during progressive metastatic disease. Patients at risk would benefit from the development of prevention and improved treatments. This requires knowledge of the initial events that lead to brain metastasis. The present study reveals cellular events during the initiation of brain metastasis by breast cancer cells and documents the earliest host responses to incoming cancer cells after carotid artery injection in immunodeficient and immunocompetent mouse models. Our findings capture and characterize heterogeneous astrocytic and microglial reactions to the arrest and extravasation of cancer cells in the brain, showing immediate and drastic changes in the brain microenvironment on arrival of individual cancer cells. We identified reactive astrocytes as the most active host cell population that immediately localizes to individual invading tumor cells and continuously associates with growing metastatic lesions. Up-regulation of matrix metalloproteinase-9 associated with astrocyte activation in the immediate vicinity of extravasating cancer cells might support their progression. Early involvement of different host cell types indicates environmental clues that might codetermine whether a single cancer cell progresses to macrometastasis or remains dormant. Thus, information on the initial interplay between brain homing tumor cells and reactive host cells may help develop strategies for prevention and treatment of symptomatic breast cancer brain metastases.
Collapse
Affiliation(s)
- Mihaela Lorger
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, MEM-150, La Jolla, CA 92037, USA.
| | | |
Collapse
|
203
|
Ghosh A, Chaudhuri S. Microglial action in glioma: a boon turns bane. Immunol Lett 2010; 131:3-9. [PMID: 20338195 DOI: 10.1016/j.imlet.2010.03.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2010] [Revised: 03/13/2010] [Accepted: 03/16/2010] [Indexed: 12/25/2022]
Abstract
Microglia has the potential to shape the neuroimmune defense with vast array of functional attributes. The cells prime infiltrated lymphocytes to retain their effector functions, play crucial role in controlling microenvironmental milieu and significantly participate in glioma. Reports demonstrate microglial accumulation in glioma and predict their assistance in glioma growth and spreading. Clarification of the 'double-edged' appearance of microglia is necessary to unfold its role in glioma biology. In this article the interpretation of microglial activities has been attempted to reveal their actual function in glioma. Contrary to the trendy acceptance of its glioma promoting infamy, accumulated evidences make an effort to view the state of affairs in favor of the cell. Critical scrutiny indicates that microglial immune assaults are intended to demolish the neoplastic cells in brain. But the weaponry of microglia has been tactically utilized by glioma in their favor as the survival strategy. Hence the defender appears as enemy in advanced glioma.
Collapse
Affiliation(s)
- Anirban Ghosh
- Immunobiology Lab, Department of Zoology, Panihati Mahavidyalaya (West Bengal State University), Barasat Road, Sodepur, Kolkata, West Bengal, India.
| | | |
Collapse
|
204
|
Donson AM, Birks DK, Barton VN, Wei Q, Kleinschmidt-DeMasters BK, Handler MH, Waziri AE, Wang M, Foreman NK. Immune Gene and Cell Enrichment Is Associated with a Good Prognosis in Ependymoma. THE JOURNAL OF IMMUNOLOGY 2009; 183:7428-40. [DOI: 10.4049/jimmunol.0902811] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
205
|
Non-apoptotic Fas signaling regulates invasiveness of glioma cells and modulates MMP-2 activity via NFkappaB-TIMP-2 pathway. Cell Signal 2009; 22:212-20. [PMID: 19788921 DOI: 10.1016/j.cellsig.2009.09.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Revised: 09/04/2009] [Accepted: 09/14/2009] [Indexed: 12/31/2022]
Abstract
Fas (CD95/APO-1) is a cell surface "death receptor" that mediates apoptosis upon engagement by its ligand, FasL. Paradoxically, Fas/FasL can also promote cell invasion among non-apoptotic cells; here, we show that Fas/FasL signaling can promote tumor invasion when apoptosis is compromised. We have developed a recombinant FasL Interfering Protein (FIP) to interfere with Fas signaling in C6 glioma cells expressing both Fas receptor and its ligand. FIP administration did not affect cell viability but impaired cell motility and invasiveness of glioma cells. Blockade of Fas signaling reduced MMP-2 activity in glioma cells, that was associated with down-regulation of MAPK signaling, and AP-1 and NFkappaB-driven transcription. FIP treatment did not affect mmp-2 and mt1-mmp expression but significantly attenuated timp-2 expression and TIMP-2 amount in the culture medium. Studies with pharmacological inhibitors of JNK/c-Jun (SP600125) and NFkappaB (BAY11-7082) signaling pathways demonstrated that timp-2 expression is regulated by NFkappaB transcription factor. Our findings show that non-apoptotic Fas signaling activated in the autocrine manner or through microenvironment derived factors can regulate invasiveness of glioma cells via modulation of MMP-2 activation, likely by controlling TIMP-2 expression.
Collapse
|
206
|
Lin HC, Song TY, Hu ML. S-Adenosylhomocysteine promotes the invasion of C6 glioma cells via increased secretion of matrix metalloproteinase-2 in murine microglial BV2 cells. Toxicol Sci 2009; 112:322-30. [PMID: 19770485 DOI: 10.1093/toxsci/kfp218] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
S-Adenosylhomocysteine (SAH) is a risk factor for many diseases, including tumor progression and neurodegenerative disease. In this study, we examined the hypothesis that SAH may indirectly enhance the invasion of C6 glioma cells by induction of matrix metalloproteinase-2 (MMP-2) secreted from the murine microglia BV2 cells. We obtained conditioned medium (CM) by incubating BV2 cells with SAH (1-50nM) for 24 h. We found that the SAH-containing CM (SAH-BV2-CM) strongly enhanced the invasiveness of C6 glioma cells and that this effect increased with increasing concentrations of SAH in the SAH-BV2-CM. The effect of CM could be attributed to its MMP-2 activity, as a result of increased protein and messenger RNA expression of MMP-2 in BV2 cells induced by SAH. In BV2 cells treated with SAH, the binding abilities of nuclear factor-kappa B (NF-kappaB) and stimulatory protein-1 (Sp1) to the MMP-2 promoter were increased, whereas the level of NF-kappaB inhibitor was decreased. In addition, SAH significantly increased the phosphorylation of extracellular signal-regulated kinase (ERK) and phosphatidylinositol-3-kinase/serine/threonine protein kinase (or protein kinase B) (PI3K/Akt) proteins but did not affect that of c-Jun NH2-terminal kinase or p38. Pretreatment of BV2 cells with an inhibitor specific for ERK (U0126) markedly abated the expression of ERK and MMP-2. Furthermore, SAH significantly and dose dependently decreased tissue inhibitor of metalloproteinase-2 (TIMP-2) in BV2 cells. Thus, SAH may induce the invasiveness of C6 glioma cells by decreased TIMP-2 expression and increased MMP-2 expression in BV2 cells. The latter effect is likely mediated through the ERK and PI3K/Akt pathways, with increased binding activities of NF-kappaB and Sp1 to the MMP-2 gene promoter.
Collapse
Affiliation(s)
- Hung-Chi Lin
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 40227, Taiwan, Republic of China
| | | | | |
Collapse
|
207
|
Gliomas induce and exploit microglial MT1-MMP expression for tumor expansion. Proc Natl Acad Sci U S A 2009; 106:12530-5. [PMID: 19617536 DOI: 10.1073/pnas.0804273106] [Citation(s) in RCA: 293] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Diffuse infiltration of glioma cells into normal brain tissue is considered to be a main reason for the unfavorable outcomes of patients with malignant gliomas. Invasion of glioma cells into the brain parenchyma is facilitated by metalloprotease-mediated degradation of the extracellular matrix. Metalloproteases are released as inactive pro-forms and get activated upon cleavage by membrane bound metalloproteases. Here, we show that membrane type 1 metalloprotease (MT1-MMP) is up-regulated in glioma-associated microglia, but not in the glioma cells. Overexpression of MT1-MMP is even lethal for glioma cells. Glioma-released factors trigger the expression and activity of MT1-MMP via microglial toll-like receptors and the p38 MAPK pathway, as deletion of the toll-like receptor adapter protein MyD88 or p38 inhibition prevented MT1-MMP expression and activity in cultured microglial cells. Microglial MT1-MMP in turn activates glioma-derived pro-MMP-2 and promotes glioma expansion, as shown in an ex vivo model using MT1-MMP-deficient brain tissue and a microglia depletion paradigm. Finally, MyD88 deficiency or microglia depletion largely attenuated glioma expansion in 2 independent in vivo models.
Collapse
|
208
|
Kreutz S, Koch M, Böttger C, Ghadban C, Korf HW, Dehghani F. 2-Arachidonoylglycerol elicits neuroprotective effects on excitotoxically lesioned dentate gyrus granule cells via abnormal-cannabidiol-sensitive receptors on microglial cells. Glia 2009; 57:286-94. [PMID: 18837048 DOI: 10.1002/glia.20756] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Endocannabinoids like 2-arachidonoylglycerol (2-AG) exert neuroprotective effects after brain injuries. According to current concepts, these neuroprotective effects are due to interactions between 2-AG and cannabinoid (CB)1 receptors on neurons. Moreover, 2-AG modulates migration and proliferation of microglial cells which are rapidly activated after brain lesion. This effect is mediated via CB2- and abnormal-cannabidiol (abn-CBD)-sensitive receptors. In the present study, we investigated whether the abn-CBD-sensitive receptor on microglial cells contributes to 2-AG-mediated neuroprotection in organotypic hippocampal slice cultures (OHSCs) after excitotoxic lesion induced by NMDA (50 microM) application for 4 h. This lesion caused neuronal damage and accumulation of microglial cells within the granule cell layer. To analyze the role of abn-CBD-sensitive receptors for neuroprotection and microglial cell accumulation, two agonists of the abn-CBD-sensitive receptor, abn-CBD or 2-AG, two antagonists, 1,3-dimethoxy-5-methyl-2-[(1R,6R)-3-methyl-6-(1-methylethenyl)-2-cyclohexen1-yl]-benzene (O-1918) or cannabidiol (CBD), and the CB1 receptor antagonist AM251, were applied to NMDA-lesioned OHSC. Propidium iodide (PI) labeling was used as a marker of degenerating neurons and isolectin B(4) (IB(4)) as a marker of microglial cells. Application of both, abn-CBD or 2-AG to lesioned OHSC significantly decreased the number of IB(4)(+) microglial cells and PI(+) neurons in the dentate gyrus. In contrast to AM251, application of O-1918 or CBD antagonized these effects. When microglial cells were depleted by preincubation of OHSC with the bisphosphonate clodronate (100 microg/mL) for 5 days before excitotoxic lesion, 2-AG and abn-CBD lost their neuroprotective effects. We therefore propose that the endocannabinoid 2-AG exerts its neuroprotective effects via activation of abn-CBD-sensitive receptors on microglial cells.
Collapse
Affiliation(s)
- Susanne Kreutz
- Dr. Senckenbergische Anatomie, Institut für Anatomie II, J. W. Goethe-Universität, Frankfurt am Main, Germany
| | | | | | | | | | | |
Collapse
|
209
|
Daginakatte GC, Gianino SM, Zhao NW, Parsadanian AS, Gutmann DH. Increased c-Jun-NH2-kinase signaling in neurofibromatosis-1 heterozygous microglia drives microglia activation and promotes optic glioma proliferation. Cancer Res 2009; 68:10358-66. [PMID: 19074905 DOI: 10.1158/0008-5472.can-08-2506] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Neurofibromatosis-1 (NF1) is a common tumor predisposition syndrome in which affected individuals develop benign and malignant tumors. Previous studies from our laboratory and others have shown that benign tumor formation in Nf1 genetically engineered mice (GEM) requires a permissive tumor microenvironment. In the central nervous system, Nf1 loss in glia is insufficient for glioma formation unless coupled with Nf1 heterozygosity in the brain. Our subsequent studies identified Nf1+/- microglia as a critical cellular determinant of optic glioma growth in Nf1 GEM. Using NF1 as an experimental paradigm to further characterize the role of microglia in glioma growth, we first examined the properties of Nf1+/- microglia in vitro and in vivo. Nf1+/- microglia exhibit increased proliferation and motility and express elevated levels of genes associated with microglia activation. We further show that Nf1+/- microglia harbor high levels of activated c-Jun-NH(2)-kinase (JNK) without any significant changes in Akt, mitogen-activated protein kinase (MAPK), or p38-MAPK activity. In contrast, Nf1-/- astrocytes do not exhibit increased JNK activation. SP600125 inhibition of JNK activity in Nf1+/- microglia results in amelioration of the increased proliferation and motility phenotypes and reduces the levels of expression of activated microglia-associated transcripts. Moreover, SP600125 treatment of Nf1 optic glioma-bearing GEM results in reduced optic glioma proliferation in vivo. Collectively, these findings suggest that Nf1+/- microglia represent a good model system to study the role of specialized microglia in brain tumorigenesis and identify a unique Nf1 deregulated pathway for therapeutic studies aimed at abrogating microenvironmental signals that promote brain tumor growth.
Collapse
Affiliation(s)
- Girish C Daginakatte
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | | | | | | | | |
Collapse
|
210
|
Abstract
Tumor-selective replication-competent viral vectors, such as oncolytic herpes simplex virus (HSV) type I (HSV-1), represent an attractive strategy for tumor-based therapies because these viruses can replicate and spread in situ exhibiting cytopathic effects through direct oncolytic activity. These lytic viruses offer a distinct advantage over other forms of cancer therapies in that they are self-perpetuating and can spread not only in the tumor itself, but also to distant micrometastases. Translational studies aimed at identifying novel virotherapies for human cancers are incumbent upon the appropriate experimental models. While animal models are the preferred choice for efficacy studies of HSV virotherapy, we have developed a novel complementary approach toward assessing the effectiveness of oncolytic HSV therapy in both brain and prostate cancers. This experimental model takes advantage of previously published work in which human prostate cancer biopsies and rodent brain slices can be easily maintained ex vivo. The advantage of these systems is that the three-dimensional structure remains intact. Thus, all of the factors that may affect viral entry and replication, such as cell-cell and cell-matrix interactions, and interstitial fluid within this three-dimensional milieu remain preserved. Moreover, with respect to the brain, this system offers the advantage of direct access to brain cells, such as microglia and astrocytes, and circumvents the problems associated with the presence of the blood-brain barrier.
Collapse
Affiliation(s)
- Giulia Fulci
- Brain Tumor Research Center, Simches Research Building, Neurosurgery Service, Massachussetts General Hospital, Boston, MA, USA
| | | |
Collapse
|
211
|
Fishman P, Bar-Yehuda S, Synowitz M, Powell JD, Klotz KN, Gessi S, Borea PA. Adenosine receptors and cancer. Handb Exp Pharmacol 2009:399-441. [PMID: 19639290 PMCID: PMC3598010 DOI: 10.1007/978-3-540-89615-9_14] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The A(1), A(2A), A(2B) and A(3) G-protein-coupled cell surface adenosine receptors (ARs) are found to be upregulated in various tumor cells. Activation of the receptors by specific ligands, agonists or antagonists, modulates tumor growth via a range of signaling pathways. The A(1)AR was found to play a role in preventing the development of glioblastomas. This antitumor effect of the A(1)AR is mediated via tumor-associated microglial cells. Activation of the A(2A)AR results in inhibition of the immune response to tumors via suppression of T regulatory cell function and inhibition of natural killer cell cytotoxicity and tumor-specific CD4+/CD8+ activity. Therefore, it is suggested that pharmacological inhibition of A(2A)AR activation by specific antagonists may enhance immunotherapeutics in cancer therapy. Activation of the A(2B)AR plays a role in the development of tumors via upregulation of the expression levels of angiogenic factors in microvascular endothelial cells. In contrast, it was evident that activation of A(2B)AR results in inhibition of ERK1/2 phosphorylation and MAP kinase activity, which are involved in tumor cell growth signals. Finally, A(3)AR was found to be highly expressed in tumor cells and tissues while low expression levels were noted in normal cells or adjacent tissue. Receptor expression in the tumor tissues was directly correlated to disease severity. The high receptor expression in the tumors was attributed to overexpression of NF-kappaB, known to act as an A(3)AR transcription factor. Interestingly, high A(3)AR expression levels were found in peripheral blood mononuclear cells (PBMCs) derived from tumor-bearing animals and cancer patients, reflecting receptor status in the tumors. A(3)AR agonists were found to induce tumor growth inhibition, both in vitro and in vivo, via modulation of the Wnt and the NF-kappaB signaling pathways. Taken together, A(3)ARs that are abundantly expressed in tumor cells may be targeted by specific A(3)AR agonists, leading to tumor growth inhibition. The unique characteristics of these A(3)AR agonists make them attractive as drug candidates.
Collapse
Affiliation(s)
- P Fishman
- Can-Fite BioPharma, Kiryat Matalon, Petach Tikva, 49170, Israel.
| | | | | | | | | | | | | |
Collapse
|
212
|
Abstract
The development of effective immunotherapy strategies for glioma requires adequate understanding of the unique immunological microenvironment in the central nervous system (CNS) and CNS tumors. Although the CNS is often considered to be an immunologically privileged site and poses unique challenges for the delivery of effector cells and molecules, recent advances in technology and discoveries in CNS immunology suggest novel mechanisms that may significantly improve the efficacy of immunotherapy against gliomas. In this review, we first summarize recent advances in the CNS and CNS tumor immunology. We address factors that may promote immune escape of gliomas. We also review advances in passive and active immunotherapy strategies for glioma, with an emphasis on lessons learned from recent early-phase clinical trials. We also discuss novel immunotherapy strategies that have been recently tested in non-CNS tumors and show great potential for application to gliomas. Finally, we discuss how each of these promising strategies can be combined to achieve clinical benefit for patients with gliomas.
Collapse
Affiliation(s)
- Hideho Okada
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA.
| | | | | | | | | | | | | |
Collapse
|
213
|
Abstract
T-cell mediated immunotherapy is a conceptually attractive treatment option to envisage for glioma, since T lymphocytes can actively seek out neoplastic cells in the brain, and they have the potential to safely and specifically eliminate tumor. Some antigenic targets on glioma cells are already defined, and we can be optimistic that more will be discovered from progress in T-cell epitope identification and gene expression profiling of brain tumors. In parallel, advances in immunology (regional immunology, neuroimmunology, tumor immunology) now equip us to build upon the results from current immunotherapy trials in which the safety and feasibility of brain tumor immunotherapy have already been confirmed. We can now look to the next phase of immunotherapy, in which we must harness the most promising basic science advances and existing clinical expertise, and apply these to randomized clinical trials to determine the real clinical impact and applicability of these approaches for treating patients with currently incurable malignant brain tumors.
Collapse
Affiliation(s)
- Erwin G. Meir
- School of Medicine, Emory University, Clifton Road 1365C, Atlanta, 30322 U.S.A
| |
Collapse
|
214
|
Cheung G, Kann O, Kohsaka S, Făerber K, Kettenmann H. GABAergic activities enhance macrophage inflammatory protein-1alpha release from microglia (brain macrophages) in postnatal mouse brain. J Physiol 2008; 587:753-68. [PMID: 19047202 DOI: 10.1113/jphysiol.2008.163923] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Microglial cells (brain macrophages) invade the brain during embryonic and early postnatal development, migrate preferentially along fibre tracts to their final position and transform from an amoeboid to a ramified morphology. Signals by which the invading microglia communicate with other brain cells are largely unknown. Here, we studied amoeboid microglia in postnatal corpus callosum obtained from 6- to 8-day-old mice. These cells accumulated on the surface of acute brain slices. Whole-cell patch-clamp recordings revealed that the specific GABA(A) receptor agonist muscimol triggered a transient increase in conductance typical for inward rectifying potassium channels in microglia. This current increase was not mediated by microglial GABA(A) receptors since microglial cells removed from the slice surface no longer reacted and cultured microglia only responded when a brain slice was placed in their close vicinity. Muscimol triggered a transient increase in extracellular potassium concentration ([K(+)](o)) in brain slices and an experimental elevation of [K(+)](o) mimicked the muscimol response in microglial cells. Moreover, in adult brain slices, muscimol led only to a minute increase in [K(+)](o) and microglial cells failed to respond to muscimol. In turn, an increase in [K(+)](o) stimulated the release of chemokine macrophage inflammatory protein-1alpha (MIP1-alpha) from brain slices and from cultures of microglia but not astrocytes. Our observations indicate that invading microglia in early postnatal development sense GABAergic activities indirectly via sensing changes in [K(+)](o) which results in an increase in MIP1-alpha release.
Collapse
Affiliation(s)
- Giselle Cheung
- Cellular Neurosciences, Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | | | | | | | | |
Collapse
|
215
|
An α5β1 integrin inhibitor attenuates glioma growth. Mol Cell Neurosci 2008; 39:579-85. [DOI: 10.1016/j.mcn.2008.08.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2008] [Revised: 07/24/2008] [Accepted: 08/13/2008] [Indexed: 11/19/2022] Open
|
216
|
Nowicki MO, Dmitrieva N, Stein AM, Cutter JL, Godlewski J, Saeki Y, Nita M, Berens ME, Sander LM, Newton HB, Chiocca EA, Lawler S. Lithium inhibits invasion of glioma cells; possible involvement of glycogen synthase kinase-3. Neuro Oncol 2008; 10:690-9. [PMID: 18715951 DOI: 10.1215/15228517-2008-041] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Therapies targeting glioma cells that diffusely infiltrate normal brain are highly sought after. Our aim was to identify novel approaches to this problem using glioma spheroid migration assays. Lithium, a currently approved drug for the treatment of bipolar illnesses, has not been previously examined in the context of glioma migration. We found that lithium treatment potently blocked glioma cell migration in spheroid, wound-healing, and brain slice assays. The effects observed were dose dependent and reversible, and worked using every glioma cell line tested. In addition, there was little effect on cell viability at lithium concentrations that inhibit migration, showing that this is a specific effect. Lithium treatment was associated with a marked change in cell morphology, with cells retracting the long extensions at their leading edge. Examination of known targets of lithium showed that inositol monophosphatase inhibition had no effect on glioma migration, whereas inhibition of glycogen synthase kinase-3 (GSK-3) did. This suggested that the effects of lithium on glioma cell migration could possibly be mediated through GSK-3. Specific pharmacologic GSK-3 inhibitors and siRNA knockdown of GSK-3alpha or GSK-3beta isoforms both reduced cell motility. These data outline previously unidentified pathways and inhibitors that may be useful for the development of novel anti-invasive therapeutics for the treatment of brain tumors.
Collapse
Affiliation(s)
- Michal O Nowicki
- Dardinger Laboratory for Neuro-oncology and Neurosciences, Department of Neurological Surgery, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
217
|
Brain inflammation and adult neurogenesis: the dual role of microglia. Neuroscience 2008; 158:1021-9. [PMID: 18662748 DOI: 10.1016/j.neuroscience.2008.06.052] [Citation(s) in RCA: 568] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2008] [Revised: 06/18/2008] [Accepted: 06/20/2008] [Indexed: 12/16/2022]
Abstract
In the adult mammalian brain, neurogenesis from neural stem/progenitor cells continues in two regions: the subgranular zone in the dentate gyrus and the subventricular zone lining the lateral ventricles. The generated neuroblasts migrate to their appropriate location and differentiate to mature granule cells and olfactory bulb interneurons, respectively. Following injury such as stroke, neuroblasts generated in the subventricular zone migrate also into areas which are not normally neurogenic, e.g. striatum and cerebral cortex. In the initial studies in rodents, brain inflammation and microglia activation were found to be detrimental for the survival of the new hippocampal neurons early after they had been born. The role of inflammation for adult neurogenesis has, however, turned out to be much more complex. Recent experimental evidence indicates that microglia under certain circumstances can be beneficial and support the different steps in neurogenesis, progenitor proliferation, survival, migration, and differentiation. Here we summarize the current knowledge on the role of inflammation and in particular of microglia in adult neurogenesis in the intact and injured mammalian brain. We conclude that microglia activation, as an indicator of inflammation, is not pro- or antineurogenic per se but the net outcome is dependent on the balance between secreted molecules with pro- and antiinflammatory action.
Collapse
|
218
|
MacKay JA, Li W, Huang Z, Dy EE, Huynh G, Tihan T, Collins R, Deen DF, Szoka FC. HIV TAT Peptide Modifies the Distribution of DNA Nanolipoparticles Following Convection-enhanced Delivery. Mol Ther 2008; 16:893-900. [DOI: 10.1038/mt.2008.36] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
219
|
Tanaka Y, Sasaki A, Ishiuchi S, Nakazato Y. Diversity of glial cell components in pilocytic astrocytoma. Neuropathology 2008; 28:399-407. [PMID: 18312545 DOI: 10.1111/j.1440-1789.2008.00896.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
To characterize the cellular density and proliferative activity of GFAP-negative cells in pilocytic astrocytoma (PA), surgically excised tissues of PAs (n=37) and diffuse astrocytomas (DAs) (n=11) were examined morphologically and immunohistochemically using antibodies against GFAP, Olig2, Iba1 and Ki-67 (MIB-1). In PA, Olig2 immunoreactivity was significantly expressed in protoplasmic astrocytes in microcystic, loose areas and cells in oligodendroglioma-like areas. Iba1-positive, activated microglia/macrophages were also commonly observed in microcystic areas. In compact areas, a prominent reaction for GFAP was observed, but for Olig2 and Iba1 to a lesser degree. On semiquantitative analysis, the number of Olig2-positive cells was significantly higher in PAs (mean labeling index (LI) +/- standard deviation (SD): 46.8+/-15.4%) than in DAs (13.3+/-7.8%) (P<0.001). Many Iba1-positive, microglia/macrophages were observed in PAs (19.9+/-6.5%), similarly to DAs (20.9+/-9.9%). Re-immunostaining of PA demonstrated that most Ki-67-positive, proliferating cells expressed Olig2, whereas GFAP or Iba1 expression in Ki-67-positive cells was less frequent (14.7+/-13.7%, and 8.8+/-13.6%) in a double immunostaining study. Conversely, the percentage of Olig2-positive, proliferating cells in total Olig2-positive cells (7.2+/-3.9%) was higher than that of Iba1-positive, proliferating cells in total Iba1-positive cells (0.9+/-0.6%). In conclusion, the present study found that PA consisted of numerous GFAP-negative cells, including Olig2-positive cells with high proliferation. Semiquantitative analysis of Olig2 immunohistochemistry in microcystic areas might therefore be useful for the differential diagnosis of PA and DA.
Collapse
Affiliation(s)
- Yuko Tanaka
- Department of Human Pathology, Gumma University Graduate School of Medicine, Maebashi, Gumma, Japan
| | | | | | | |
Collapse
|
220
|
Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci 2008; 10:1387-94. [PMID: 17965659 DOI: 10.1038/nn1997] [Citation(s) in RCA: 2672] [Impact Index Per Article: 167.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Microglial cells constitute the resident macrophage population of the CNS. Recent in vivo studies have shown that microglia carry out active tissue scanning, which challenges the traditional notion of 'resting' microglia in the normal brain. Transformation of microglia to reactive states in response to pathology has been known for decades as microglial activation, but seems to be more diverse and dynamic than ever anticipated--in both transcriptional and nontranscriptional features and functional consequences. This may help to explain why engagement of microglia can be either neuroprotective or neurotoxic, resulting in containment or aggravation of disease progression. Moreover, little is known about the heterogeneity of microglial responses in different pathologic contexts that results from regional adaptations or from the progression of a disease. In this review, we focus on several key observations that illustrate the multi-faceted activities of microglia in the normal and pathologic brain.
Collapse
|
221
|
Fulci G, Dmitrieva N, Gianni D, Fontana EJ, Pan X, Lu Y, Kaufman CS, Kaur B, Lawler SE, Lee RJ, Marsh CB, Brat DJ, van Rooijen N, Rachamimov AS, Hochberg FH, Weissleder R, Martuza RL, Chiocca EA. Depletion of peripheral macrophages and brain microglia increases brain tumor titers of oncolytic viruses. Cancer Res 2007; 67:9398-406. [PMID: 17909049 PMCID: PMC2850558 DOI: 10.1158/0008-5472.can-07-1063] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Clinical trials have proven oncolytic virotherapy to be safe but not effective. We have shown that oncolytic viruses (OV) injected into intracranial gliomas established in rodents are rapidly cleared, and this is associated with up-regulation of markers (CD68 and CD163) of cells of monocytic lineage (monocytes/microglia/macrophages). However, it is unclear whether these cells directly impede intratumoral persistence of OV through phagocytosis and whether they infiltrate the tumor from the blood or the brain parenchyma. To investigate this, we depleted phagocytes with clodronate liposomes (CL) in vivo through systemic delivery and ex vivo in brain slice models with gliomas. Interestingly, systemic CL depleted over 80% of peripheral CD163+ macrophages in animal spleen and peripheral blood, thereby decreasing intratumoral infiltration of these cells, but CD68+ cells were unchanged. Intratumoral viral titers increased 5-fold. In contrast, ex vivo CL depleted only CD68+ cells from brain slices, and intratumoral viral titers increased 10-fold. These data indicate that phagocytosis by both peripheral CD163+ and brain-resident CD68+ cells infiltrating tumor directly affects viral clearance from tumor. Thus, improved therapeutic efficacy may require modulation of these innate immune cells. In support of this new therapeutic paradigm, we observed intratumoral up-regulation of CD68+ and CD163+ cells following treatment with OV in a patient with glioblastoma.
Collapse
Affiliation(s)
- Giulia Fulci
- Molecular Neuro-oncology Laboratories, Neurosurgery Service, and Center for Molecular Imaging, Massachusetts General Hospital-East Building, Charlestown, Massachusetts
- Brain Tumor Research Center, Neurosurgery Service, Simches Research Building, Boston, Massachusetts
- Department of Neurological Surgery, Dardinger Laboratory for Neuro-oncology and Neurosciences, James Cancer Hospital/Solove Research Institute, Columbus, Ohio
| | - Nina Dmitrieva
- Department of Neurological Surgery, Dardinger Laboratory for Neuro-oncology and Neurosciences, James Cancer Hospital/Solove Research Institute, Columbus, Ohio
| | - Davide Gianni
- Molecular Neuro-oncology Laboratories, Neurosurgery Service, and Center for Molecular Imaging, Massachusetts General Hospital-East Building, Charlestown, Massachusetts
| | - Elisabeth J. Fontana
- Molecular Neuro-oncology Laboratories, Neurosurgery Service, and Center for Molecular Imaging, Massachusetts General Hospital-East Building, Charlestown, Massachusetts
| | - Xiaogang Pan
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University Medical Center, Columbus, Ohio
| | - Yanhui Lu
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University Medical Center, Columbus, Ohio
| | - Claire S. Kaufman
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Balveen Kaur
- Department of Neurological Surgery, Dardinger Laboratory for Neuro-oncology and Neurosciences, James Cancer Hospital/Solove Research Institute, Columbus, Ohio
| | - Sean E. Lawler
- Department of Neurological Surgery, Dardinger Laboratory for Neuro-oncology and Neurosciences, James Cancer Hospital/Solove Research Institute, Columbus, Ohio
| | - Robert J. Lee
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University Medical Center, Columbus, Ohio
| | - Clay B. Marsh
- Department of Internal Medicine, The Ohio State University Medical Center, Columbus, Ohio
| | - Daniel J. Brat
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Nico van Rooijen
- Department of Cell Biology and Immunology, Vrije Universiteit, Amsterdam, the Netherlands
| | - Anat Stemmer Rachamimov
- Molecular Neuro-oncology Laboratories, Neurosurgery Service, and Center for Molecular Imaging, Massachusetts General Hospital-East Building, Charlestown, Massachusetts
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Fred H. Hochberg
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts
| | - Ralph Weissleder
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Robert L. Martuza
- Brain Tumor Research Center, Neurosurgery Service, Simches Research Building, Boston, Massachusetts
| | - E. Antonio Chiocca
- Molecular Neuro-oncology Laboratories, Neurosurgery Service, and Center for Molecular Imaging, Massachusetts General Hospital-East Building, Charlestown, Massachusetts
- Department of Neurological Surgery, Dardinger Laboratory for Neuro-oncology and Neurosciences, James Cancer Hospital/Solove Research Institute, Columbus, Ohio
| |
Collapse
|
222
|
Hoelzinger DB, Demuth T, Berens ME. Autocrine factors that sustain glioma invasion and paracrine biology in the brain microenvironment. J Natl Cancer Inst 2007; 99:1583-93. [PMID: 17971532 DOI: 10.1093/jnci/djm187] [Citation(s) in RCA: 279] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Invasion is a defining hallmark of glioblastoma multiforme, just as metastasis characterizes other high-grade tumors. Glial tumors invariably recur due to the regrowth of invasive cells, which are unaffected by standard treatment modalities. Drivers of glioma invasion include autocrine signals propagated by secreted factors that signal through receptors on the tumor. These secreted factors are able to diffuse through the peritumoral stroma, thereby influencing parenchymal cells that surround the tumor mass. Here we describe various autocrine motility factors that are expressed by invasive glioma cells and explore the effects that they may have on normal cells present in the path of invasion. Conversely, normal brain parenchymal cells secrete ligands that can stimulate receptors on invasive glioma cells and potentially facilitate glioma invasion or create a permissive microenvironment for malignant progression. Parallel observations have been made for solid tumors of epithelial origin, in which parenchymal and stromal cells either support or suppress tumor invasion. Most autocrine and paracrine interactions involved in glioma invasion constitute known signaling systems in stages of central nervous system development that involve the migration of precursor cells that populate the developing brain. Key paracrine interactions between glioma cells and the brain microenvironment can influence glioma pathobiology and therefore contribute to its poor prognosis. Current therapies for glioma that could have an impact on paracrine communication between tumors and normal cells are discussed. We suggest that cells in the normal brain parenchyma be considered as potential targets for adjuvant therapies to control glioma growth because such cells are less likely to develop resistance than glioma cells.
Collapse
Affiliation(s)
- Dominique B Hoelzinger
- Cancer and Cell Biology Division, Translational Genomics Research Institute, 445 North Fifth Street, Phoenix, AZ 85004, USA
| | | | | |
Collapse
|
223
|
Wesolowska A, Kwiatkowska A, Slomnicki L, Dembinski M, Master A, Sliwa M, Franciszkiewicz K, Chouaib S, Kaminska B. Microglia-derived TGF-β as an important regulator of glioblastoma invasion—an inhibition of TGF-β-dependent effects by shRNA against human TGF-β type II receptor. Oncogene 2007; 27:918-30. [PMID: 17684491 DOI: 10.1038/sj.onc.1210683] [Citation(s) in RCA: 186] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The invasion of tumor cells into brain tissue is a pathologic hallmark of malignant gliomas and contributes to treatment failures. Diffuse glioblastomas contain numerous microglial cells, which enhance the progression of gliomas; however, factors responsible for invasion-promoting role of microglia are unknown. Transforming growth factor-beta (TGF-beta) can enhance tumor growth, invasion, angiogenesis and immunosuppression. Antagonizing TGF-beta activity has been shown to inhibit tumor invasion in vitro and tumorigenicity, but a systemic inhibition or lack of TGF-beta signaling results in acute inflammation and disruption of immune system homeostasis. We developed plasmid-transcribed small hairpin RNAs (shRNAs) to downregulate the TGF-beta type II receptor (TbetaIIR) expression, which effectively inhibited cytokine-induced signaling pathways and transcriptional responses in transiently transfected human glioblastoma cells. Silencing of TbetaIIR abolished TGF-beta-induced glioblastoma invasiveness and migratory responses in vitro. Moreover, tumorigenicity of glioblastoma cells stably expressing TbetaIIR shRNAs in nude mice was reduced by 50%. Microglia strongly enhanced glioma invasiveness in the co-culture system, but this invasion-promoting activity was lost in glioma cells stably expressing shTbetaRII, indicating a crucial role of microglia-derived TGF-beta in tumor-host interactions. Our results demonstrate a successful targeting of TGF-beta-dependent invasiveness and tumorigenicity of glioblastoma cells by RNAi-mediated gene silencing.
Collapse
Affiliation(s)
- A Wesolowska
- Laboratory of Transcription Regulation, Department of Cell Biology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | | | | | | | | | | | | | | | | |
Collapse
|
224
|
Daginakatte GC, Gutmann DH. Neurofibromatosis-1 (Nf1) heterozygous brain microglia elaborate paracrine factors that promote Nf1-deficient astrocyte and glioma growth. Hum Mol Genet 2007; 16:1098-112. [PMID: 17400655 DOI: 10.1093/hmg/ddm059] [Citation(s) in RCA: 154] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The tumor microenvironment is considered to play an important role in tumor formation and progression by providing both negative and positive signals that influence tumor cell growth. We and others have previously shown that brain tumor (glioma) formation in Nf1 genetically engineered mice requires a microenvironment composed of cells heterozygous for a targeted Nf1 mutation. Using NF1 as a model system to understand the contribution of the tumor microenvironment to glioma formation, we show that Nf1+/- brain microglia produce specific factors that promote Nf1-/- astrocyte growth in vitro and in vivo and identify hyaluronidase as one of these factors in both genetically engineered Nf1 mouse and human NF1-associated optic glioma. We further demonstrate that blocking hyaluronidase ameliorates the ability of Nf1+/- microglia to increase Nf1-/- astrocyte proliferation and that hyaluronidase increases Nf1-/- astrocyte proliferation in an MAPK-dependent fashion. Lastly, inhibiting microglia activation in genetically engineered Nf1 mice significantly reduces mouse optic glioma proliferation in vivo. Collectively, these studies identify Nf1+/- microglia as an important stromal cell type that promotes Nf1-/- astrocyte and optic glioma growth relevant to the pathogenesis of NF1-associated brain tumors and suggest that future brain therapies might be directed against paracrine factors produced by cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Girish C Daginakatte
- Department of Neurology, Washington University School of Medicine, St Louis, MO 63110, USA
| | | |
Collapse
|
225
|
Quiñones-Hinojosa A, Chaichana K. The human subventricular zone: a source of new cells and a potential source of brain tumors. Exp Neurol 2007; 205:313-24. [PMID: 17459377 DOI: 10.1016/j.expneurol.2007.03.016] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2006] [Revised: 02/23/2007] [Accepted: 03/13/2007] [Indexed: 02/04/2023]
Abstract
The mammalian brain has been perceived as a quiescent organ incapable of postnatal neurogenesis for many years. Most recently, several studies have demonstrated that the adult mammalian brain is indeed capable of neurogenesis and that the process is primarily confined to the subventricular zone (SVZ) of the forebrain and the subgranular zone (SGZ) of the hippocampus. Of these regions, the SVZ is the largest niche of neurogenesis in the adult mammalian brain. Within this niche resides a subpopulation of astrocytes with stem cell-like features of self-renewal and multipotentiality. Interestingly, there is also a subpopulation of cells within brain tumors that possess these same characteristics. Based on these findings, the emerging hypothesis is that brain tumor stem cells may be derived from neural stem cells and that both of these populations may originate from the SVZ. This possible connection stresses the importance of studying and understanding the role that the human SVZ plays in not only harboring neural and brain tumor stem cells, but how this microenvironment may support both neurogenesis and tumorigenesis. Furthermore, the obvious differences in the SVZ between humans and other animals make it important to understand the human model when studying human disease. Such an understanding may lead to novel therapeutic strategies for both neurodegenerative diseases and currently intractable brain tumors.
Collapse
Affiliation(s)
- Alfredo Quiñones-Hinojosa
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | |
Collapse
|
226
|
Synowitz M, Glass R, Färber K, Markovic D, Kronenberg G, Herrmann K, Schnermann J, Nolte C, van Rooijen N, Kiwit J, Kettenmann H. A1 Adenosine Receptors in Microglia Control Glioblastoma-Host Interaction. Cancer Res 2006; 66:8550-7. [PMID: 16951167 DOI: 10.1158/0008-5472.can-06-0365] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We report that experimental glioblastoma grow more vigorously in A(1) adenosine receptor (A(1)AR)-deficient mice associated with a strong accumulation of microglial cells at and around the tumors. A(1)ARs were prominently expressed in microglia associated with tumor cells as revealed with immunocytochemistry but low in microglia in the unaffected brain tissue. The A(1)AR could also be detected on microglia from human glioblastoma resections. To study functional interactions between tumor and host cells, we studied glioblastoma growth in organotypical brain slice cultures. A(1)AR agonists suppressed tumor growth. When, however, microglial cells were depleted from the slices, the agonists even stimulated tumor growth. Thus, adenosine attenuates glioblastoma growth acting via A(1)AR in microglia.
Collapse
Affiliation(s)
- Michael Synowitz
- Cellular Neuroscience Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
227
|
|