It is the cache of ${baseHref}. It is a snapshot of the page. The current page could have changed in the meantime.
Tip: To quickly find your search term on this page, press Ctrl+F or ⌘-F (Mac) and use the find bar.

ASN NEURO (2011) 3, e00064 - Neoplastic macrophages/microglia in GBM - L C Huysentruyt, Z Akgoc and T N Seyfried
About   Submit   Librarians   Authors   Readers   Help
AMERICAN SOCIETY FOR NEUROCHEMISTRY The Latest in Molecular and Cellular Neurobiology
 
Editorial Board
Editor-in-Chief
Monica Carson
   Riverside, U.S.A.

Deputy Editor-in-Chief
Christopher S. Colwell
   Los Angeles, U.S.A.

Reviews Editors
Scott T. Brady
   Chicago, U.S.A.
Wilma J. Friedman
   Newark, U.S.A.

Senior Editors
Susan Y. Bookheimer
   Los Angeles, U.S.A.
Carol A. Colton
   Durham, U.S.A.
Iryna M. Ethell
   Riverside, U.S.A.
Kazuhiro Ikenaka
   Osaka, Japan
Wendy Macklin
   Denver, U.S.A.
Mary C. McKenna
   Baltimore, U.S.A.
Vladimir Parpura
   Birmingham, U.S.A.
Thomas Seyfried
   Chestnut Hill, U.S.A.

Founding Editor
Anthony T. Campagnoni
   Los Angeles, U.S.A.
ISSN: 1759-0914
Published by Portland Press Limited on behalf of the American Society for Neurochemistry
*Publication time subject to receipt of payment
†2012 Journal Citation Reports® (Thomson Reuters, 2013)
The author has paid for access to this article to be freely availble
ASN NEURO (2011) 3(4):art:e00064.doi:10.1042/AN20110011
Review
Hypothesis: are neoplastic macrophages/microglia present in glioblastoma multiforme?
Leanne C Huysentruyt*, Zeynep Akgoc† and Thomas N Seyfried† 1
*Department of Medicine, Hematology and Oncology, University of California, San Francisco, CA, U.S.A.
†Department of Biology, Boston College, Chestnut Hill, MA, U.S.A.


Cite this article as:  Leanne C Huysentruyt, Zeynep Akgoc and Thomas N Seyfried  (2011)  Hypothesis: are neoplastic macrophages/microglia present in glioblastoma multiforme?. ASN NEURO 3(4):art:e00064.doi:10.1042/AN20110011

Most malignant brain tumours contain various numbers of cells with characteristics of activated or dysmorphic macrophages/microglia. These cells are generally considered part of the tumour stroma and are often described as TAM (tumour-associated macrophages). These types of cells are thought to either enhance or inhibit brain tumour progression. Recent evidence indicates that neoplastic cells with macrophage characteristics are found in numerous metastatic cancers of non-CNS (central nervous system) origin. Evidence is presented here suggesting that subpopulations of cells within human gliomas, specifically GBM (glioblastoma multiforme), are neoplastic macrophages/microglia. These cells are thought to arise following mitochondrial damage in fusion hybrids between neoplastic stem cells and macrophages/microglia.


Key words: fusion, glioblastoma multiforme, glioma, macrophage, microglia, phagocytosis

Abbreviations: AIF1, allograft inflammatory factor 1, CNS, central nervous system, GFAP, glial fibrillary acidic protein, GBM, glioblastoma multiforme, IL, interleukin, MDSC, myeloid-derived suppressor cell, MNGC, multinucleated giant cell, PXA, pleomorphic xanthoastrocytoma, ROS, reactive oxygen species, RTG, retrograde, TAM, tumour-associated macrophage, VM, vasculogenic mimicry

1To whom correspondence should be addressed (email Thomas.seyfried@bc.edu).


INTRODUCTION

It has recently been suggested that many types of human cancers (lung, breast, colon, kidney, etc.) contain neoplastic cells with mesenchymal/macrophage properties (Huysentruyt and Seyfried, 2010). These neoplastic macrophages are often the most highly invasive and metastatic cells within the tumour. It is not clear if similar kinds of cells are part of the malignant-cell population in human GBM (glioblastoma multiforme). GBM is the most common form of primary brain cancer in adults and represents ∼65% of all newly diagnosed malignant gliomas (Ohgaki and Kleihues, 2005; Stupp et al., 2009). GBM portends an extremely poor outcome with only approx. 10% of patients surviving 5 years after diagnosis, and a median survival of ∼12 months (Krex et al., 2007; Preusser et al., 2011). The poor prognosis is due largely to the highly invasive nature of this tumour. GBM invades throughout the brain and often produces multi-centric secondary legions at sites distant from the primary tumour (Scherer, 1940; Rubinstein, 1972; Laws et al., 1993). Complete surgical resection of GBM is extremely rare. Radiation therapy, which enhances the necrotic microenvironment, often results in further tissue damage and more aggressive tumours (Lakka and Rao, 2008; Kargiotis et al., 2010; Seyfried et al., 2010, 2011). Thus, effective therapeutic options are desperately needed for GBM patients.

GBM is classified as either primary or secondary. Primary GBM arise de novo without any prior evidence of a low-grade tumour, whereas secondary GBM arises from malignant progression of a lower grade glioma (Ohgaki and Kleihues, 2009). A defining characteristic of GBM is the ‘secondary structures of Scherer’ which include diffuse parenchymal invasion, perivascular growth, subpial surface growth and invasion along white matter tracks (Scherer, 1940; Shelton et al., 2010b). While systemic metastasis of GBM is uncommon due to early patient death, GBM can be extremely metastatic especially if the neoplastic cells gain access to extraneural sites (Hoffman and Duffner, 1985; Rubinstein, 1972; Ng et al., 2005; Taha et al., 2005; Frank et al., 2009; Zhen et al., 2010; Gotway et al., 2011). These findings indicate that GBM is not only highly invasive within the CNS (central nervous system), but can also invade outside the CNS.

GBM generally contain multiple morphologically diverse cell types that express neural, glial and myeloid markers (Rubinstein, 1972; Wood and Morantz, 1979; Seyfried, 2001; Yuan et al., 2004; Huysentruyt et al., 2008). In fact, mesenchymal cells with characteristics of TAM (tumour-associated macrophages) and/or microglia can comprise up to 70% of some GBM (Morantz et al., 1979). It has been difficult to determine with certainty, however, the origin of all the mesenchymal and macrophage-like cells that appear within the GBM (Tso et al., 2006; Ricci-Vitiani et al., 2010). Based on numerous similarities between macrophages and invasive neoplastic cells in non-CNS tumours (Huysentruyt and Seyfried, 2010), we propose that some neoplastic cells within the GBM arise from transformed macrophage/microglia. As cells of the myeloid/macrophage lineage naturally embody the capacity to invade (Mallat et al., 2005; Banaei-Bouchareb et al., 2006), we suggest that some of the highly invasive mesenchymal-type cells within GBM may arise from resident or infiltrating myeloid cells of the tumour stroma that then become neoplastic during disease progression (Huysentruyt and Seyfried, 2010).

Our hypothesis comes from information on the VM (vasculogenic mimicry) mouse model of GBM which consists of highly invasive tumours (Shelton et al., 2010b). The VM tumours arise spontaneously in the brains of inbred VM mice (Fraser, 1971, 1986; Shelton et al., 2010b). The neoplastic cells in the invasive VM brain tumours express multiple properties of macrophages and microglial cells and the VM-M3 and VM-M2 tumours have been established as a model system for human GBM (Huysentruyt et al., 2008; Huysentruyt et al., 2010; Huysentruyt and Seyfried, 2010; Shelton et al., 2010b). We recently reviewed evidence showing that many invasive and metastatic human cancers also express multiple properties of mesenchymal myeloid cells (Huysentruyt and Seyfried, 2010). If many non-neural metastatic tumours arise from mesenchymal myeloid type cells, what about malignant tumours of the CNS? The goal of this commentary is to highlight similarities between myeloid cells and the invasive-cell populations of GBM, and to discuss possible mechanisms by which neoplastic cells could express these properties.

MYELOID CELLS AND INVASIVE CANCER

Myeloid cells have long been considered the origin of human metastatic cancers; however, this hypothesis has received little attention in the cancer field (Munzarova and Kovarik, 1987; Rachkovsky et al., 1998; Huysentruyt and Seyfried, 2010; Pawelek, 2000; Vignery, 2005; Pawelek and Chakraborty, 2008). Although large numbers of cells with macrophage and myeloid properties are found in most malignant cancers including GBM, these cells are generally considered part of the tumour stroma and not part of the neoplastic population (Seyfried, 2001; Mantovani et al., 2002). These cells are often referred to as TAM, and are thought to facilitate tumour development and malignant progression (Seyfried, 2001; Bingle et al., 2002; Lewis and Pollard, 2006; Talmadge et al., 2007; Pollard, 2008). The tumour stroma is a complex microenvironment that generally comprises malignant tumour cells and host infiltrating cells that include immune and epithelial cells (Bissell and Hines, 2011). The most commonly accepted view is that tumour-derived chemoattractants signal monocytes to extravasate out of the bloodstream and infiltrate the tumour mass (Bottazzi et al., 1983; Murdoch et al., 2004). Once in the tumour microenvironment, enhanced inflammation and angiogenesis establish the pre-metastatic niche, thus contributing to tumour progression (Seyfried, 2001; Bingle et al., 2002; Lewis and Pollard, 2006; Talmadge et al., 2007; Pollard, 2008). However, little consideration has been given to the possibility that microglial or TAM subsets might actually be part of the neoplastic cell population within GBM (Graeber et al., 2002; Watters et al., 2005). As cells of macrophage/microglial origin can express multiple mesenchymal morphologies depending on the microenvironment, how is it possible to be sure that none of these stromal cells are part of the neoplastic cell population?

Macrophages and microglia are among the most versatile cells of the body with respect to their ability to migrate, to change shape and to secrete growth factors and cytokines (Gordon, 1999; Stossel, 1999; Burke and Lewis, 2002; Huysentruyt et al., 2008). Macrophages fluctuate between two activation states depending on the microenvironment. Macrophages can become classically activated (M1 activation) in response to pathogens and pro-inflammatory molecules, resulting in the generation of NO and ROS (reactive oxygen species) (Sica et al., 2002, 2006; Biswas et al., 2008; Mantovani and Sica, 2010; Qian and Pollard, 2010). In contrast, macrophages can become alternatively activated (M2 activation) in response to anti-inflammatory molecules such as IL-4 (interleukin-4) and IL-10 and are the major wound-healing cells (Gordon, 2003; Biswas et al., 2008). Interestingly, the two macrophage subpopulations play separate roles in tumorigenic processes (Biswas et al., 2008). M1 macrophages, located at sites of chronic inflammation, contribute to neoplastic transformation through the release of cytotoxic and DNA damaging pro-inflammatory molecules (Biswas et al., 2008). Macrophages with an M2 phenotype contribute to angiogenesis and metastasis within the established tumour (Mantovani et al., 2002; Sica et al., 2006; Biswas et al., 2008; Huysentruyt and Seyfried, 2010; Mantovani and Sica, 2010). However, recent evidence suggests that TAMs may be a more complex subtype that can share features of both activation states (Ojalvo et al., 2009, 2010; Qian and Pollard, 2010).

Ineffective anti-tumour immunity resulting from reduced T-cell function is a hallmark of many cancers including GBM (Waziri, 2010; Raychaudhuri et al., 2011). Recently, it has been suggested that another myeloid cell type, MDSCs (myeloid-derived suppressor cells) are partially responsible for GBM-related immune suppression (Raychaudhuri et al., 2011). MDSCs are a heterogeneous population of immature myeloid cells that accumulate in tumour-bearing hosts where they suppress T-cell function, resulting in ineffective anti-tumour immunity (Gabrilovich and Nagaraj, 2009; Ostrand-Rosenberg, 2010; Raychaudhuri et al., 2011). This cell type accumulates in the blood, lymph nodes and spleen in patients with solid tumours and has been reported to represent approx. 5% of the total tumour mass in various experimental tumour-model systems (Yang et al., 2004). Raychaudhuri et al. demonstrated that patients with GBM have increased MDSC counts in their blood when compared with healthy controls and these cells likely promote T-cell immune suppression in this patient population (Raychaudhuri et al., 2011). While the phenotype and function of MDSCs are believed to be distinct from those of TAMs, MDSCs can differentiate into TAMs within the tumour microenvironment thus further contributing to tumour progression (reviewed in Gabrilovich and Nagaraj, 2009). To our knowledge, it has not been demonstrated if MDSCs contribute to GBM tumour mass.

PHAGOCYTOSIS: A DEFINING BEHAVIOUR OF MACROPHAGES AND INVASIVE CELLS

Phagocytosis, involving engulfment and ingestion of extracellular material, is a defining property of macrophages/microglia and of other professional phagocytes (Burke and Lewis, 2002). In order to maintain tissue homoeostasis, macrophages phagocytose cellular debris, apoptotic cells and pathogens (Burke and Lewis, 2002). Phagocytosis is also expressed in numerous invasive and metastatic cancers including breast, lung, liver, pancreatic, skin and brain (reviewed in Huysentruyt and Seyfried, 2010). It has been suggested that invasive tumour cells use phagocytosis to facilitate migration through the extracellular matrix and into surrounding tissues (Bjerknes et al., 1987; Coopman et al., 1998).

The phagocytic/cannibalistic behaviour of tumour cells was first described over a century ago when foreign cell bodies were identified within human cancer cells (reviewed in Steinhaus, 1981). These cells were commonly described as ‘signet-ring’ and ‘birds-eye’ cells due to the peripheral displacement of their nuclei from engulfed materials (Fais, 2007). Although numerous micro-organisms use cannibalism as a feeding mechanism, cellular cannibalism is an exclusive property of malignant tumour cells (Fais, 2007). Both human and murine cancers have been shown to phagocytose tumour cells, erythrocytes, leucocytes, platelets, apoptotic cells and extracellular particles (reviewed in Huysentruyt and Seyfried, 2010). Interestingly, phagocytic tumour cells are observed in malignant gliomas, especially in GBM (Figure 1) (Youness et al., 1980; Bjerknes et al., 1987; Nitta et al., 1992; Zimmer et al., 1995; Chang et al., 2000; Persson and Englund, 2009; van Landeghem et al., 2009).

We previously identified two spontaneous brain tumours (VM-M2 and VM-M3) in the inbred VM mouse strain that are highly invasive in brain (Huysentruyt et al., 2008; Shelton et al., 2010b). These VM brain tumours express multiple properties of macrophages including phagocytic activity and are a model for human GBM (Huysentruyt et al., 2008; Shelton et al., 2010b). The phagocytic phenotype in these cells is remarkably similar to that of observed in the RAW 264.7 macrophage cell line (Figure 1A) (Huysentruyt et al., 2008). In a similar study to ours that examined the phagocytic activity of four rat glioma cell lines with varying degrees of in vivo invasiveness, Bjerknes et al. demonstrated that the invasive glioma cells phagocytosed bacteria, red blood cells, zymosan particles and glia cell fragments (Bjerknes et al., 1987). Furthermore, the two most invasive rat glioma cell lines displayed the highest level of phagocytosis. The authors noted that the phagocytic behaviour correlated with the amount of rat brain destruction during tumour cell invasion, suggesting that phagocytic activity may be connected to the excretion of lysosomal enzymes (Bjerknes et al., 1987). Lysosomal enzyme secretion and phagocytosis are macrophage-specific behaviours. Invasive glioma cell lines that display macrophage/microglia-like behaviours suggest a myeloid origin. Chang et al. (2000) also demonstrated that the glioma cell lines U87, U251 and SF268 exhibit phagocytic behaviour against apoptotic glioma cells. These findings further demonstrate that invasive glioma cells can exhibit macrophage-like phagocytic behaviour.

Although astrocytes have been described as semi-professional phagocytes, their phagocytic ability is limited. It could be suggested that the phagocytic behaviour observed in the GBM might arise from transformed astrocytes. However, the recent findings of Persson and Englund (2009) suggest that the phagocytic activity observed in human GBM are properties of malignant macrophages/microglia, rather than properties of malignant astrocytes. Utilizing paraffin-embedded GBM material, they identified numerous CD68-positive phagocytic macrophage/microglia throughout brain tumour specimens. Double-immunostaining with CD68 and the tumour cell marker hTERT (human telomerase reverse transcriptase) was used to verify that the CD68-positive cells were in fact part of the malignant GBM cell population (Persson and Englund, 2009). These findings support the hypothesis that neoplastic macrophages/microglia can exist within GBM.

In addition to GBM, other invasive CNS tumours contain neoplastic phagocytic cells. For example, phagocytic cells were observed in the bone marrow of a patient with a highly invasive and metastatic medulloblastoma, which involved invasion and metastasis to brainstem, bone marrow, lymph nodes and spinal cord (Youness et al., 1980). After extensive characterization of the phagocytic cells via histopatholgy, electron microscopy and cytogenetics, the authors stated that the phagocytic cells were unquestionably metastatic medulloblastoma cells. Interestingly, this tumour metastasized outside the CNS after excision of the primary tumour from the right cerebellar region (Youness et al., 1980). While extracranial metastasis of CNS tumours is uncommon, invasive CNS tumours can metastasize throughout the body if they gain access to extraneural sites (Rubinstein, 1972; Hoffman and Duffner, 1985; Vural et al., 1996; Taha et al., 2005). In fact, several reports show that GBM can be highly metastatic (Hoffman and Duffner, 1985; Ng et al., 2005; Taha et al., 2005; Frank et al., 2009; Zhen et al., 2010; Gotway et al., 2011). It therefore appears that this case of metastatic medulloblastoma exhibited the macrophage/microglia characteristics seen in some GBM.

While it might be difficult to demonstrate a myeloid origin of invasive GBM, the evidence reviewed here suggests that subpopulations of neoplastic GBM cells display the phagocytic behaviour of macrophages/microglia. As microglia are the resident macrophages of the brain, we propose that subpopulations of the malignant GBM cells could arise from microglia/macrophages (Morantz et al., 1979; Seyfried, 2001). Human GBM contain mixtures of numerous neoplastic cell types, many of which have mesenchymal properties, do not express GFAP (glial fibrillary acidic protein) and are of unknown origin (Duffy, 1983; Ohgaki and Kleihues, 2009; Han et al., 2010). Indeed, the original 19th century observations of Virchow (1863/1865) described glioblastomas as gliosarcomas of mesenchymal origin (Scherer, 1940; Zagzag et al., 2008). While numerous mesenchymal cells are frequently seen in GBM, the specific classification of all tumour cell types within human GBM remains ambiguous at best (Yates, 1992; Tso et al., 2006; Fan et al., 2007). According to our hypothesis, some of these neoplastic mesenchymal cells could arise from transformed macrophages/microglia.

Novel therapeutic approaches to GBM management become possible if the phagocytic behaviour can be targeted. Gollapudi and co-workers demonstrated that tumour cells undergo apoptosis after ‘feeding’ tumour cells with anti-tumour agents (Ghoneum and Gollapudi, 2004; Ghoneum et al., 2005, 2007, 2008). They showed that various tumour cells, grown either in vitro or in vivo, underwent a yeast-induced apoptosis after engulfing Saccharomyces cerevisiae. Recent studies indicate that similar approaches could have therapeutic efficacy in GBM. Glioblastoma patients underwent treatment with a magnetic nanoparticle-based immunotherapy where nanoparticles were engulfed by cells within the GBM (van Landeghem et al., 2009). To identify the phagocytic cells within the tumour, these investigators stained post-mortem fixed GBM sections with either the macrophage marker, CD68, or with the astrocyte/CNS stem cell marker, GFAP (van Landeghem et al., 2009). The authors reported that most of the cells engulfing the magnetic nanoparticles were CD68-positive. It was not possible from their study, however, to determine if the CD68-positive cells were also neoplastic since CD68 would stain both transformed macrophages/microglia as well as non-neoplastic TAM.

FUSOGENICITY

Fusogenicity involves the merging of two distinct plasma membranes and is a defining characteristic of macrophages and microglia (Vignery, 2000; Duelli and Lazebnik, 2003; Huysentruyt and Seyfried, 2010). Cell fusion is a highly regulated process that is essential for fertilization, skeletal muscle and placenta formation (Duelli and Lazebnik, 2003). Outside of normal developmental processes, fusion events are normally restricted to cells of myeloid origin (reviewed in Duelli and Lazebnik, 2003). Macrophages often fuse during osteoclast formation and wound healing in order increase cell volume to facilitate engulfment of large extracellular materials (Vignery, 2000, 2005). Macrophages also undergo heterotypic fusions with numerous cell types. It is currently believed that macrophage heterotypic fusion is a mechanism by which macrophages can ‘heal’ a defective cell or tissue (Vignery, 2000; Camargo et al., 2004a, 2004b; Vignery, 2005; Powell et al., 2011). There is also considerable evidence demonstrating that macrophages fuse with tumour cells (reviewed in Huysentruyt and Seyfried, 2010). These properties would also be expected in microglia since microglia are the resident macrophages of the CNS.

Nearly a century ago, Aichel first suggested that fusions between somatic cells and leucocytes could result in aneuploidy and malignant hybrids (reviewed in Rachkovsky et al., 1998). Sixty years later, Mekler and Warner proposed that fusions between myeloid cells and tumour cells would produce daughter cells endowed with the invasive properties of the myeloid cell as well as the unlimited proliferative potential of the tumour cell (reviewed in Rachkovsky et al., 1998). The fusion hybrid hypothesis has emerged as a credible alternative to the epithelial mesenchymal transition for the origin of invasive and metastatic cancers (Huysentruyt and Seyfried, 2010; Pawelek and Chakraborty, 2008). Indeed, Pawelek and co-workers showed that fusions between non-metastatic cells and macrophages result in cells with the ability to invade and metastasize (Rachkovsky et al., 1998; Rachkovsky and Pawelek, 1999; Chakraborty et al., 2000, 2001, 2004; Pawelek, 2000, 2005; Handerson et al., 2005; Yilmaz et al., 2005; Pawelek and Chakraborty, 2008). Additionally, they provided evidence demonstrating that these fusion events occur spontaneously both in vitro and in vivo in a variety of animal models and in human cancers (Rachkovsky et al., 1998; Chakraborty et al., 2000, 2004; Yilmaz et al., 2005). Fusions between tumour cells and macrophage/microglia, with subsequent nuclear fusion, could produce novel phenotypes in the absence of new mutations since the tumour-macrophage hybrids express genetic and functional traits of both parental cells (Figure 2A) (Rachkovsky et al., 1998; Powell et al., 2011). Tumour–macrophage hybrids could also account in part for immune surveillance evasion seen in GBM and other metastatic cancers (Huysentruyt and Seyfried, 2010).

GBM is composed of neoplastic cells with features that vary from small round cells to MNGCs (multinucleated giant cells) (Homma et al., 2006). MNGCs arise from the fusion of two or more macrophages (Vignery, 2000, 2005). Recent reports suggest that MNGCs in GBM could arise through fusions of peripheral macrophages or microglia with glioma cells (Alvarez-Dolado et al., 2003). A recent study demonstrated that CD98, a protein that regulates monocyte fusion events, was expressed on the surface of MNGCs in 15/16 GBMs (Figure 2B) (Takeuchi et al., 2008). These findings suggest that MNGC formation in GBM is similar to what occurs during normal monocyte/macrophage fusion events (Takeuchi et al., 2008). Microglia and bone-marrow derived cells can fuse with CNS cells resulting in MNGCs (Alvarez-Dolado et al., 2003; Ackman et al., 2006). The MNGCs in GBM often express GFAP, a protein expressed on CNS stem cells and astrocytes, suggesting that this cell population contains macrophage/microglia and CNS stem cell/astrocyte hybrids (Takeuchi et al., 2008). Patient prognosis is generally worse when GBMs contain large numbers of MNGCs and other macrophage-like cells when compared with GBMs containing fewer of these cells (Deininger et al., 2003). This situation is similar to that seen in non-CNS tumours in that prognosis is generally worse for malignant tumours with higher rather than lower numbers of macrophages (Leek et al., 1996; Homma et al., 2006; Shabo et al., 2009; Huysentruyt and Seyfried, 2010).

The large size and unique phenotype of MNGCs make them easily detectable in brain tumour biopsy specimens (Figure 1B) (Rubinstein, 1972). However, it can be difficult to detect fusion events among tumour cells and myeloid cells with subsequent nuclear fusions. Utilizing a double immunostaining technique, Deininger et al. (2000) identified a subset of glioma cells that were positive for both GFAP and AIF1 (allograft inflammatory factor 1), a microglial marker also referred to as Iba1. GFAP+/AIF1+ cells were localized near areas of tumour growth and were indicative of spontaneous in vivo fusion events (Deininger et al., 2000). It is well documented that TAM aid tumour progression by promoting angiogenesis, invasion and metastasis (reviewed in Seyfried, 2001; Lewis and Pollard, 2006). However, fusion events between tumour cells and tissue macrophage/microglia would also accelerate tumour progression since resulting daughter cells would inherit the invasive/migratory potential of microglia and unlimited proliferative potential of the tumour cell. Hence, a microglial/glioma cell hybrid could rapidly acquire an invasive phenotype in the absence of novel mutations.

The macrophage-like characteristics and invasive properties of the murine VM-M2 and VM-M3 brain tumours are more similar to human GBM than to most previously described experimental mouse brain tumour models (Shelton et al., 2010b). If fusion hybridization of neoplastic cells with macrophages/microglia could be the origin of invasive cells within human GBM, why are these cell types rarely found in rodent brain tumour transplant models? Most macrophage/microglia seen in chemically induced brain tumours are derived from resident microglia and TAM (Ecsedy et al., 1998). We suggest that the mouse or the rat brain would respond to a tumour implant as if it were an acute infection. This would involve invasion of TAM and activation of local microglia. This is not what usually occurs in the development of a human GBM, where neoplastic transformation is a protracted process. For example, GBM can arise many years after head wound trauma (Gruss et al., 1993; Sabel et al., 1999). It is therefore possible that conditions for macrophage/microglia fusogenicity are greater in the microenvironment of human brain than in the microenvironment of the rodent brain during malignant transformation.

GLIOMA CELL EXPRESSION OF MYELOID ANTIGENS

Macrophages and microglia express unique antigens that can help identify these cells in various tissues (Roggendorf et al., 1996; Guillemin and Brew, 2004). A list of some of these antigens is shown in Table 1. Numerous studies have reported high numbers of cells in GBM that express antigens seen only in macrophages/microglia (Rossi et al., 1987; Roggendorf et al., 1996; Leung et al., 1997; Badie and Schartner, 2000; Graeber et al., 2002). As mentioned above, most cells within GBM that express macrophage antigens are generally considered host-infiltrating TAM, and are not considered part of the neoplastic tumour cell population (Roggendorf et al., 1996; Seyfried, 2001). However, several reports show that macrophage/microglial antigens are expressed on neoplastic cells within GBM (Figure 3) (Rossi et al., 1987; Leenstra et al., 1995; Graeber et al., 2002; Deininger et al., 2003; Strojnik et al., 2006). These findings in GBM are consistent with findings reported for other human cancer types including breast, lung, skin, ovarian, pancreatic, rectal and renal (Rossi et al., 1987; Leenstra et al., 1995; Deininger et al., 2003; Matyja et al., 2003; Strojnik et al., 2006, 2009; Colman et al., 2010) and reviewed in Huysentruyt and Seyfried (2010). The expression of macrophage antigens in neoplastic cell subpopulations within GBM would support our hypothesis that malignant microglia are a component of human GBM.


Table 1 Malignant glioma cell expression of myeloid antigens

Myeloid antigen References
CD98 Takeuchi et al. (2008)
AIF-1 (Iba-1) Deininger et al. (2000) and Huysentruyt et al. (2008)
CD14 Deininger et al. (2003)
HLA-class II Rossi et al. (1987) and Matyja et al. (2003)
CD68 Leenstra et al. (1995), Matyja et al. (2003), Strojnik et al. (2006), Huysentruyt et al. (2008) and Strojnik et al. (2009)
CD11b Leenstra et al. (1995) and Huysentruyt et al. (2008)
HAM56 Leenstra et al. (1995)



Deininger et al. (2003) utilized double labelling experiments to demonstrate that neoplastic cells located in regions of infiltrative glioma growth were positive for both GFAP and the macrophage marker CD14. It is currently thought that TAM, localized at the leading edge of the tumour, facilitate local tumour cell invasion (Lewis and Pollard, 2006). However, the invading cells were CD14+/GFAP+ suggesting that these cells were part of the invading tumour cell population and not TAM. It is possible that these cells arise through fusion of tumour cells with macrophages/microglia, thus representing a neoplastic hybrid-cell population. In contrast to the Deininger et al.'s study (Deininger et al., 2003), most studies of macrophages in brain tumours have not used double labelling to distinguish neoplastic cells from non-neoplastic cells (Rossi et al., 1987; Shinonaga et al., 1988; Morimura et al., 1990; van Landeghem et al., 2009). Consequently, it is often difficult to determine with certainty the origin of all cell types that express macrophage/microglia antigens in GBM. However, the human glioblastoma cell line U138 expressed the macrophage marker Ki-M1P (CD68) in an in vitro environment where there were no contaminating TAMs (Figure 3A) (Paulus et al., 1992). This finding is similar to what we showed for the murine VM-M2 and VM-M3 cell lines (Huysentruyt et al., 2008). A few investigators have entertained the possibility that some macrophage/microglial-type cells in GBM might be part of the neoplastic cell population. Several previous studies show, however, that subpopulations of neoplastic macrophage-like cells are present in most human cancers (reviewed in Huysentruyt and Seyfried, 2010). It would therefore be important to consider the possibility that some GBM cell subpopulations, previously identified as TAM, are in fact neoplastic cells.

Like GBM, PXA (pleomorphic xanthoastrocytoma) contains cells of multiple morphologies (Matyja et al., 2003). As in GBM, MNGC are also a characteristic cell type in PXA (Matyja et al., 2003). In an attempt to further characterize this cell subtype, Matyja et al. (2003) evaluated the co-expression of glial and macrophage markers in the various tumour cell populations in eight cases of PXA. The authors identified a population of neoplastic cells that expressed GFAP and either HLA-class II or CD68 within the cytoplasm (Matyja et al., 2003). Many of these neoplastic cells were large and lipid-laden suggesting a mesenchymal origin. According to our hypothesis, neoplastic microglia/macrophages could give rise to PXA following fusion hybridizations.

Expression of macrophage markers has been reported in high-grade gliomas including GBM (Rossi et al., 1987; Leenstra et al., 1995; Strojnik et al., 2006). Leenstra et al. (1995) examined macrophage characteristics in six malignant glioma cell lines. All the cell lines examined were classified as malignant gliomas based upon DNA flow cytometry and GFAP expression. Interestingly, all six glioma lines co-expressed macrophage markers and GFAP. In contrast, macrophage antigens were not found in cultured astrocytes isolated from healthy brain tissue, suggesting that the macrophage markers expressed by malignant astrocytes were not artefacts of an in vitro culture environment (Leenstra et al., 1995). In a separate study, immunohistochemical analysis revealed that 40% of grade III astrocytomas and GBM tumour specimens contained neoplastic cells with macrophage markers (Rossi et al., 1987). The U87 GBM model has also been shown to express both CD68 (Figure 3B) and glia markers when grown either in vitro or in vivo (Strojnik et al., 2006). Strojnik et al. (2009) evaluated the prognostic significance of CD68 expression in malignant human gliomas. It was clear form their study that the presence of high levels of CD68-positive tumour cells was predictive of reduced survival. CD68 was expressed by both microglia and tumour cells (Strojnik et al., 2009). The macrophage/microglia marker, Iba1, is also expressed by the VM-M2 mouse GBM tumour cells (Figure 3C) (Huysentruyt et al., 2008). Taken together, these findings support our hypothesis and indicate that some neoplastic GBM cells could be of macrophage/microglial origin.

NUMEROUS HUMAN CANCERS EXPRESS MACROPHAGE PROPERTIES

Evidence presented above suggests that subpopulations of the invasive GBM tumour cells include neoplastic cells with multiple characteristics of macrophages/microglia. Recent studies show that macrophage properties and antigens are expressed in the neoplastic cells of numerous human metastatic cancers including bladder, brain, breast, carcinoma of unknown primary, endometrial, fibrosarcoma, gall bladder, liver, lung, lymphoma, melanoma, multiple myeloma, ovarian, pancreatic, rectal, rhabdomyosarcoma and renal cancers (reviewed in Huysentruyt and Seyfried, 2010). Commonly, these macrophage characteristics are observed in the invasive and metastatic cell populations, further highlighting the possibility that the highly-invasive tumour cells may be of mesenchymal origin. Indeed, these observations led to our recent prediction that the metastatic cells in most human cancers are of myeloid origin (Huysentruyt and Seyfried, 2010). As microglia are mesenchymal cells of myeloid origin, we suggest that neoplastic microglia can represent an invasive cell population in GBM. This shared property among numerous invasive/metastatic tumours could have broad therapeutic implications. For example, any therapy that is able to reduce invasion and metastasis of one tumour type would likely be effective in treating tumours with macrophage characteristics.

POSSIBLE MECHANISMS

How could resident microglia or TAM become part of the neoplastic cell population in GBM? We recently reviewed evidence indicating that all cancer, regardless of tissue or cellular origin, is primarily a disease of impaired cellular energy metabolism (Seyfried and Shelton, 2010). Otto Warburg first proposed that all types of cancers arise from irreversible damage to cellular respiration (Warburg, 1931, 1956). Persistent injury to oxidative phosphorylation will require compensatory non-oxidative energy metabolism to maintain viability (Seyfried and Shelton, 2010; Seyfried et al., 2010; Shelton et al., 2010b). The mitochondrial stress response or RTG (retrograde) signalling from the mitochondria to the nucleus is required to up-regulate the oncogenes needed to sustain glycolysis and non-oxidative mitochondrial energy production (Seyfried and Shelton, 2010; Seyfried et al., 2010). However, persistent RTG activation leads to eventual nuclear genomic instability and other recognized hallmarks of cancer (Butow and Avadhani, 2004; Singh et al., 2005; Seyfried and Shelton, 2010). Mitochondria energy production is often damaged as a consequence of mutagens, hypoxia, inflammation, ROS or inherited mutations (Kiebish et al., 2008). We recently showed that mitochondria isolated from murine gliomas contain numerous lipid defects supporting previous findings that respiratory energy production is dysfunctional in tumour mitochondria (Kiebish et al., 2008, 2009; Ordys et al., 2010; Seyfried and Shelton, 2010). Macrophages/microglia home to mitochondria-damaging environments in response to inflammation, infection, wound repair and tumorigenesis (Giulian et al., 1989; Chettibi, 1999; Bingle et al., 2002; Graeber et al., 2002; Lewis and Murdoch, 2005; Martin and Leibovich, 2005; Lewis and Pollard, 2006). Inflammation and hypoxia in the tumour's microenvironment can damage macrophage mitrochondria (Frost et al., 2005; Navarro and Boveris, 2005). It is therefore possible that mitochondria could become dysfunctional when macrophages/microglia respond to various chronic tissue injuries, resulting in a persistent RTG response with eventual malignant transformation (Seyfried and Shelton, 2010). Hence, we suggest that some invasive GBM cells could arise from resident tissue microglia or TAM that have suffered mitochondrial damage during tumour initiation or progression.

Focal hypoxia combined with persistent inflammation and stem cell proliferation could produce macrophage–microglia or macrophage–stem cell fusions. Respiratory damage could either proceed or follow fusion events thus initiating the path to frank neoplasia. Additionally, damaged mitochondria could also be passed from one cell to another via cytoplasmic inheritance during cellular-fusion events (Seyfried and Shelton, 2010). Although normal mitochondria are known to suppress tumorigenesis in fusion hybrids (Seyfried and Shelton, 2010), persistent inflammation in the tumour microenvironment could contribute to continued mitochondrial damage in the fused hybrids. This would enhance the Warburg effect, unbridled proliferation and genomic instability. As microglia naturally embody the ability to migrate throughout the brain (Graeber and Streit, 1990; Amat et al., 1996), we suggest that transformed cells of macrophage/microglia origin would possess invasive potential.

Currently, the cause of macrophage and tumour cell hybridization is unknown. Various stages of the macrophage response to tumour development could increase the probability for cell-fusion events. The host immune system treats tumours as unhealed wounds (Dvorak, 1986; Seyfried, 2001; Bissell and Hines, 2011). Macrophages home to hypoxic tumour areas in the process of wound healing. The failure of macrophages to completely digest apoptotic cells could result in macrophage×tumour hybrid cells (Pawelek, 2000). It is therefore possible that macrophages fuse with tumour cells in an attempt to ‘heal’ the tumour, as macrophages are known to fuse with non-myeloid cells during tissue repair. Radiation is commonly used as a primary therapy for most brain tumours. However, radiation therapy enhances the incidence of fusion hybrid formation and could result in a more aggressive and invasive tumours (Shabo et al., 2009; Seyfried et al., 2011). This could account in part for the exacerbating effects of radiation therapy on GBM progression (Seyfried et al., 2010).

The macrophage fusion hybrid hypothesis could potentially explain many of the distinguishing characteristics seen in invasive and metastatic GBM tumour cells. These macrophage-reprogramming strategies may account for some of the cellular heterogeneity seen in human GBM since macrophage×CNS cell hybrids would likely retain the histology of the CNS fusion partner. Tumour hybrids could also account for the cancer cell aneuploidy and chromosomal abnormalities often seen in tumour cells (Steinhaus, 1981; Pawelek, 2000). Further studies will be needed to determine the extent to which fusion hybridization might contribute to the invasive properties seen in GBM.

CONCLUDING REMARKS

GBM is a highly complex and lethal tumour. Unfortunately, the current therapies available to GBM patients are largely ineffective and often have a negative impact on the patient's quality of life. In this commentary, we aim to highlight the possibility that the most aggressive and invasive cells in human GBMs are neoplastic macrophages/microglia. While further experimentation is needed to confirm the gene expression and immunohistochemical data reviewed here, we believe that novel therapeutic approaches could be developed targeting the macrophage characteristics of invasive GBM cells. It would be of particular interest to determine if the CD68+ cell population of human GBM has neoplastic properties.

ACKNOWLEDGEMENTS

We thank Dr Annette Persson for providing Figure 1(b) and for invaluable discussions. We also thank Dr Werner Paulus for providing the high-resolution image for Figure 3(a).

Funding

This work was supported, in part, by the National Institutes of Health [grant numbers HD -39722, NS - 55195 and CA -102135], a grant from the American Institute of Cancer Research and the Boston College Expense Fund .

REFERENCES

Ackman JB, Siddiqi F, Walikonis RS, LoTurco JJ (2006) Fusion of microglia with pyramidal neurons after retroviral infection. J Neurosci 26:11413-11422 .
Crossref   Medline   1st Citation  

Alvarez-Dolado M, Pardal R, Garcia-Verdugo JM, Fike JR, Lee HO, Pfeffer K, Lois C, Morrison SJ, Alvarez-Buylla A (2003) Fusion of bone-marrow-derived cells with Purkinje neurons, cardiomyocytes and hepatocytes. Nature 425:968-973 .
Crossref   Medline   1st Citation   2nd  

Amat JA, Ishiguro H, Nakamura K, Norton WT (1996) Phenotypic diversity and kinetics of proliferating microglia and astrocytes following cortical stab wounds. Glia 16:368-382 .
Crossref   Medline   1st Citation  

Badie B, Schartner JM (2000) Flow cytometric characterization of tumor-associated macrophages in experimental gliomas. Neurosurgery 46:957-961 .
Medline   1st Citation  

Banaei-Bouchareb L, Peuchmaur M, Czernichow P, Polak M (2006) A transient microenvironment loaded mainly with macrophages in the early developing human pancreas. J Endocrinol 188:467-480 .
Crossref   Medline   1st Citation  

Bingle L, Brown NJ, Lewis CE (2002) The role of tumour-associated macrophages in tumour progression: implications for new anticancer therapies. J Pathol 196:254-265 .
Crossref   Medline   1st Citation   2nd   3rd  

Bissell MJ, Hines WC (2011) Why don't we get more cancer? A proposed role of the microenvironment in restraining cancer progression. Nat Med 17:320-329 .
Crossref   Medline   1st Citation   2nd  

Biswas SK, Sica A, Lewis CE (2008) Plasticity of macrophage function during tumor progression: regulation by distinct molecular mechanisms. J Immunol 180:2011-2017 .
Medline   1st Citation   2nd   3rd   4th   5th  

Bjerknes R, Bjerkvig R, Laerum OD (1987) Phagocytic capacity of normal and malignant rat glial cells in culture. J Natl Cancer Inst 78:279-288 .
Medline   1st Citation   2nd   3rd   4th  

Bottazzi B, Polentarutti N, Acero R, Balsari A, Boraschi D, Ghezzi P, Salmona M, Mantovani A (1983) Regulation of the macrophage content of neoplasms by chemoattractants. Science 220:210-212 .
Crossref   Medline   1st Citation  

Burke B, Lewis CE (2002) The Macrophage., New York: Oxford University Press.
1st Citation   2nd   3rd  

Butow RA, Avadhani NG (2004) Mitochondrial signaling: the retrograde response. Mol Cell 14:1-15 .
Crossref   Medline   1st Citation  

Camargo FD, Chambers SM, Goodell MA (2004a) Stem cell plasticity: from transdifferentiation to macrophage fusion. Cell Prolif 37:55-65 .
Crossref   Medline   1st Citation  

Camargo FD, Finegold M, Goodell MA (2004b) Hematopoietic myelomonocytic cells are the major source of hepatocyte fusion partners. J Clin Invest 113:1266-1270 .
Medline   1st Citation  

Chakraborty A, Lazova R, Davies S, Backvall H, Ponten F, Brash D, Pawelek J (2004) Donor DNA in a renal cell carcinoma metastasis from a bone marrow transplant recipient. Bone Marrow Transplant 34:183-186 .
Crossref   Medline   1st Citation   2nd  

Chakraborty AK, de Freitas Sousa J, Espreafico EM, Pawelek JM (2001) Human monocyte x mouse melanoma fusion hybrids express human gene. Gene 275:103-106 .
Crossref   Medline   1st Citation  

Chakraborty AK, Sodi S, Rachkovsky M, Kolesnikova N, Platt JT, Bolognia JL, Pawelek JM (2000) A spontaneous murine melanoma lung metastasis comprised of host×tumor hybrids. Cancer Res 60:2512-2519 .
Medline   1st Citation   2nd  

Chang GH, Barbaro NM, Pieper RO (2000) Phosphatidylserine-dependent phagocytosis of apoptotic glioma cells by normal human microglia, astrocytes, glioma cells. Neuro Oncol 2:174-183 .
Medline   1st Citation   2nd  

Chettibi S, MWJ Ferguson (1999) Wound repair: an overview. In: In Inflammation: Basic Principals and Clinical Correlates (Snyderman J, ed), pp 865-881., New York: Lippincott Williams and Wilkins.
1st Citation  

Colman H, Zhang L, Sulman EP, McDonald JM, Shooshtari NL, Rivera A, Popoff S, Nutt CL, Louis DN, Cairncross JG, Gilbert MR, Phillips HS, Mehta MP, Chakravarti A, Pelloski CE, Bhat K, Feuerstein BG, Jenkins RB, Aldape K (2010) A multigene predictor of outcome in glioblastoma. Neuro Oncol 12:49-57 .
Medline   1st Citation  

Coopman PJ, Do MT, Thompson EW, Mueller SC (1998) Phagocytosis of cross-linked gelatin matrix by human breast carcinoma cells correlates with their invasive capacity. Clin Cancer Res 4:507-515 .
Medline   1st Citation  

Deininger MH, Meyermann R, Schluesener HJ (2003) Expression and release of CD14 in astrocytic brain tumors. Acta Neuropathol (Berl) 106:271-277 .
Crossref   1st Citation   2nd   3rd   4th   5th   6th  

Deininger MH, Seid K, Engel S, Meyermann R, Schluesener HJ (2000) Allograft inflammatory factor-1 defines a distinct subset of infiltrating macrophages/microglial cells in rat and human gliomas. Acta Neuropathol (Berl) 100:673-680 .
Crossref   1st Citation   2nd   3rd  

Duelli D, Lazebnik Y (2003) Cell fusion: a hidden enemy? Cancer Cell 3:445-448 .
Crossref   Medline   1st Citation   2nd   3rd  

Duffy PE (1983) Astrocytes: Normal, Reactive, and Neoplastic., p 224., New York: Raven Press.
1st Citation  

Dvorak HF (1986) Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med 315:1650-1659 .
Crossref   Medline   1st Citation  

Ecsedy JA, Yohe HC, Bergeron AJ, Seyfried TN (1998) Tumor-infiltrating macrophages contribute to the glycosphinglipid composition of brain tumors. J Lipid Res 39:2218-2227 .
Medline   1st Citation  

Fais S (2007) Cannibalism: a way to feed on metastatic tumors. Cancer Lett 258:155-164 .
Crossref   Medline   1st Citation   2nd  

Fan X, Salford LG, Widegren B (2007) Glioma stem cells: evidence and limitation. Semin Cancer Biol 17:214-218 .
Crossref   Medline   1st Citation  

Frank S, Kuhn SA, Brodhun M, Mueller U, Romeike B, Kosmehl H, Regenbrecht CR, Ewald C, Reichart R, Kalff R (2009) Metastatic glioblastoma cells use common pathways via blood and lymphatic vessels. Neurol Neurochir Pol 43:183-190 .
Medline   1st Citation   2nd  

Fraser H (1971) Astrocytomas in an inbred mouse strain. J Pathol 103:266-270 .
Crossref   Medline   1st Citation  

Fraser H (1986) Brain tumours in mice, with particular reference to astrocytoma. Food Chem Toxicol 24:105-111 .
Crossref   Medline   1st Citation  

Frost MT, Wang Q, Moncada S, Singer M (2005) Hypoxia accelerates nitric oxide-dependent inhibition of mitochondrial complex I in activated macrophages. Am J Physiol Regul Integr Comp Physiol 288:R394-R400 .
Medline   1st Citation  

Gabrilovich DI, Nagaraj S (2009) Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 9:162-174 .
Crossref   Medline   1st Citation   2nd  

Ghoneum M, Gollapudi S (2004) Phagocytosis of Candida albicans by metastatic and non metastatic human breast cancer cell lines in vitro. Cancer Detect Prev 28:17-26 .
Crossref   Medline   1st Citation  

Ghoneum M, Hamilton J, Brown J, Gollapudi S (2005) Human squamous cell carcinoma of the tongue and colon undergoes apoptosis upon phagocytosis of Saccharomyces cerevisiae, the baker's yeast, in vitro. Anticancer Res 25:981-989 .
Medline   1st Citation  

Ghoneum M, Matsuura M, Braga M, Gollapudi S (2008) S. cerevisiae induces apoptosis in human metastatic breast cancer cells by altering intracellular Ca2+ and the ratio of Bax and Bcl-2. Int J Oncol 33:533-539 .
Medline   1st Citation  

Ghoneum M, Wang L, Agrawal S, Gollapudi S (2007) Yeast therapy for the treatment of breast cancer: a nude mice model study. In Vivo 21:251-258 .
Medline   1st Citation  

Giulian D, Chen J, Ingeman JE, George JK, Noponen M (1989) The role of mononuclear phagocytes in wound healing after traumatic injury to adult mammalian brain. J Neurosci 9:4416-4429 .
Medline   1st Citation  

Gordon S (1999) Development and distribution of mononuclear phagocytes: revelance to inflammation. In: In Inflammation: Basic Principles and Clinical Correlates (Gallin J, Snyderman, R, eds), pp 35-48., New York: Lippincott Williams and Wilkins.
1st Citation  

Gordon S (2003) Alternative activation of macrophages. Nat Rev Immunol 3:23-35 .
Crossref   Medline   1st Citation  

Gotway MB, Conomos PJ, Bremner RM (2011) Pleural metastatic disease from glioblastoma multiforme. J Thorac Imaging 26:W54-W58 .
Crossref   Medline   1st Citation   2nd  

Graeber MB, Scheithauer BW, Kreutzberg GW (2002) Microglia in brain tumors. Glia 40:252-259 .
Crossref   Medline   1st Citation   2nd   3rd   4th  

Graeber MB, Streit WJ (1990) Microglia: immune network in the CNS. Brain Pathol 1:2-5 .
Crossref   Medline   1st Citation  

Gruss P, Spohr A, Leiber A, Tasler J, Menzl H, Hofstadter F (1993) The traumatic origin of a glioblastoma. Zentralbl Neurochir 54:186-189 .
Medline   1st Citation  

Guillemin GJ, Brew BJ (2004) Microglia, macrophages, perivascular macrophages, and pericytes: a review of function and identification. J Leukoc Biol 75:388-397 .
Medline   1st Citation  

Han SJ, Yang I, Otero JJ, Ahn BJ, Tihan BJ, McDermott MW, Berger MS, Chang SM, Parsa AT (2010) Secondary gliosarcoma after diagnosis of glioblastoma: clinical experience with 30 consecutive patients. J Neurosurg 112:990-996 .
Crossref   Medline   1st Citation  

Handerson T, Camp R, Harigopal M, Rimm D, Pawelek J (2005) Beta1,6-branched oligosaccharides are increased in lymph node metastases and predict poor outcome in breast carcinoma. Clin Cancer Res 11:2969-2973 .
Crossref   Medline   1st Citation  

Hoffman HJ, Duffner PK (1985) Extraneural metastases of central nervous system tumors. Cancer 56:1778-1782 .
Crossref   Medline   1st Citation   2nd   3rd  

Homma T, Fukushima T, Vaccarella S, Yonekawa Y, Di Patre PL, Franceschi S, Ohgaki H (2006) Correlation among pathology, genotype, and patient outcomes in glioblastoma. J Neuropathol Exp Neurol 65:846-854 .
Crossref   Medline   1st Citation   2nd  

Huysentruyt LC, Mukherjee P, Banerjee D, Shelton LM, Seyfried TN (2008) Metastatic cancer cells with macrophage properties: evidence from a new murine tumor model. Int J Cancer 123:73-84 .
Crossref   Medline   1st Citation   2nd   3rd   4th   5th   6th   7th   8th   9th   10th   11th   12th   13th  

Huysentruyt LC, Seyfried TN (2010) Perspectives on the mesenchymal origin of metastatic cancer. Cancer Metastasis Rev 29:695-707 .
Crossref   Medline   1st Citation   2nd   3rd   4th   5th   6th   7th   8th   9th   10th   11th   12th   13th   14th   15th   16th   17th   18th   19th  

Huysentruyt LC, Shelton LM, Seyfried TN (2010) Influence of methotrexate and cisplatin on tumor progression and survival in the VM mouse model of systemic metastatic cancer. Int J Cancer 126:65-72 .
Crossref   Medline   

Kargiotis O, Geka A, Rao JS, Kyritsis AP (2010) Effects of irradiation on tumor cell survival, invasion and angiogenesis. J Neurooncol 100:323-338 .
Crossref   Medline   1st Citation  

Kiebish MA, Han X, Cheng H, Chuang JH, Seyfried TN (2008) Cardiolipin and electron transport chain abnormalities in mouse brain tumor mitochondria: lipidomic evidence supporting the Warburg theory of cancer. J Lipid Res 49:2545-2556 .
Crossref   Medline   1st Citation   2nd  

Kiebish MA, Han X, Cheng H, Seyfried TN (2009) In vitro growth environment produces lipidomic and electron transport chain abnormalities in mitochondria from non-tumorigenic astrocytes and brain tumours. ASN NEURO 1:e00011 .
1st Citation  

Krex D, Klink B, Hartmann C, von Deimling A, Pietsch T, Simon M, Sabel M, Steinbach JP, Heese O, Reifenberger G, Weller M, Schackert G (2007) Long-term survival with glioblastoma multiforme. Brain 130:2596-2606 .
Crossref   Medline   1st Citation  

Lakka SS, Rao JS (2008) Antiangiogenic therapy in brain tumors. Expert Rev Neurother 8:1457-1473 .
Crossref   Medline   1st Citation  

Laws Jr ER, Goldberg WJ, Bernstein JJ (1993) Migration of human malignant astrocytoma cells in the mammalian brain: Scherer revisited. Int J Dev Neurosci 11:691-697 .
Crossref   Medline   1st Citation  

Leek RD, Lewis CE, Whitehouse R, Greenall M, Clarke J, Harris AL (1996) Association of macrophage infiltration with angiogenesis and prognosis in invasive breast carcinoma. Cancer Res 56:4625-4629 .
Medline   1st Citation  

Leenstra S, Das PK, Troost D, de Boer OJ, Bosch DA (1995) Human malignant astrocytes express macrophage phenotype. J. Neuroimmunol 56:17-25 .
Medline   1st Citation   2nd   3rd   4th   5th   6th   7th   8th  

Leung SY, Wong MP, Chung LP, Chan AS, Yuen ST (1997) Monocyte chemoattractant protein-1 expression and macrophage infiltration in gliomas. Acta Neuropathol (Berl) 93:518-527 .
Crossref   1st Citation  

Lewis C, Murdoch C (2005) Macrophage responses to hypoxia: implications for tumor progression and anti-cancer therapies. Am J Pathol 167:627-635 .
Crossref   Medline   1st Citation  

Lewis CE, Pollard JW (2006) Distinct role of macrophages in different tumor microenvironments. Cancer Res 66:605-612 .
Crossref   Medline   1st Citation   2nd   3rd   4th   5th  

Mallat M, Marin-Teva JL, Cheret C (2005) Phagocytosis in the developing CNS: more than clearing the corpses. Curr Opin Neurobiol 15:101-107 .
Crossref   Medline   1st Citation  

Mantovani A, Sica A (2010) Macrophages, innate immunity and cancer: balance, tolerance, and diversity. Curr Opin Immunol 22:231-237 .
Crossref   Medline   1st Citation   2nd  

Mantovani A, Sozzani S, Locati M, Allavena P, Sica A (2002) Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol 23:549-555 .
Crossref   Medline   1st Citation   2nd  

Martin P, Leibovich SJ (2005) Inflammatory cells during wound repair: the good, the bad and the ugly. Trends Cell Biol 15:599-607 .
Crossref   Medline   1st Citation  

Matyja E, Kroh H, Taraszewska A, Naganska E, Zabek M, Marchel A (2003) Expression of macrophage/histiocytic antigens in pleomorphic xanthoastrocytomas. Folia Neuropathol 41:89-95 .
Medline   1st Citation   2nd   3rd   4th   5th   6th   7th   8th  

Morantz RA, Wood GW, Foster M, Clark M, Gollahon K (1979) Macrophages in experimental and human brain tumors. Part 2: studies of the macrophage content of human brain tumors. J Neurosurg 50:305-311 .
Crossref   Medline   1st Citation   2nd  

Morimura T, Neuchrist C, Kitz K, Budka H, Scheiner O, Kraft D, Lassmann H (1990) Monocyte subpopulations in human gliomas: expression of Fc and complement receptors and correlation with tumor proliferation. Acta Neuropathol (Berl) 80:287-294 .
Crossref   1st Citation  

Munzarova M, Kovarik J (1987) Is cancer a macrophage-mediated autoaggressive disease? Lancet 1:952-954 .
Medline   1st Citation  

Murdoch C, Giannoudis A, Lewis CE (2004) Mechanisms regulating the recruitment of macrophages into hypoxic areas of tumors and other ischemic tissues. Blood 104:2224-2234 .
Crossref   Medline   1st Citation  

Navarro A, Boveris A (2005) Hypoxia exacerbates macrophage mitochondrial damage in endotoxic shock. Am J Physiol Regul Integr Comp Physiol 288:R354-R355 .
Medline   1st Citation  

Ng WH, Yeo TT, Kaye AH (2005) Spinal and extracranial metastatic dissemination of malignant glioma. J Clin Neurosci 12:379-382 .
Crossref   Medline   1st Citation   2nd  

Nitta T, Okumura K, Sato K (1992) Lysosomal enzymic activity of astroglial cells. Pathobiology 60:42-44 .
Crossref   Medline   1st Citation  

Ohgaki H, Kleihues P (2005) Epidemiology and etiology of gliomas. Acta Neuropathol (Berl) 109:93-108 .
Crossref   1st Citation  

Ohgaki H, Kleihues P (2009) Genetic alterations and signaling pathways in the evolution of gliomas. Cancer Sci 100:2235-2241 .
Crossref   Medline   1st Citation   2nd  

Ojalvo LS, King W, Cox D, Pollard JW (2009) High-density gene expression analysis of tumor-associated macrophages from mouse mammary tumors. Am J Pathol 174:1048-1064 .
Crossref   Medline   1st Citation  

Ojalvo LS, Whittaker CA, Condeelis JS, Pollard JW (2010) Gene expression analysis of macrophages that facilitate tumor invasion supports a role for Wnt-signaling in mediating their activity in primary mammary tumors. J Immunol 184:702-712 .
Crossref   Medline   1st Citation  

Ordys BB, Launay S, Deighton RF, McCulloch J, Whittle IR (2010) The role of mitochondria in glioma pathophysiology. Mol Neurobiol 42:64-75 .
Crossref   Medline   1st Citation  

Ostrand-Rosenberg S (2010) Myeloid-derived suppressor cells: more mechanisms for inhibiting antitumor immunity. Cancer Immunol Immunother 59:1593-1600 .
Crossref   Medline   1st Citation  

Paulus W, Roggendorf W, Kirchner T (1992) Ki-M1P as a marker for microglia and brain macrophages in routinely processed human tissues. Acta Neuropathol (Berl) 84:538-544 .
Crossref   1st Citation   2nd  

Pawelek JM (2000) Tumour cell hybridization and metastasis revisited. Melanoma Res 10:507-514 .
Crossref   Medline   1st Citation   2nd   3rd   4th  

Pawelek JM (2005) Tumour-cell fusion as a source of myeloid traits in cancer. Lancet Oncol 6:988-993 .
Crossref   Medline   1st Citation  

Pawelek JM, Chakraborty AK (2008) Fusion of tumour cells with bone marrow-derived cells: a unifying explanation for metastasis. Nat Rev Cancer 8:377-386 .
Crossref   Medline   1st Citation   2nd   3rd  

Persson A, Englund E (2009) The glioma cell edge-winning by engulfing the enemy? Med Hypotheses 73:336-337 .
Crossref   Medline   1st Citation   2nd   3rd  

Persson A, Englund E (2008) The glioma cell lead winning by eating? In 9th European Congress of Neuropathology, p 194., Athens, Greece: Clinical Neuropathology.
1st Citation  

Pollard JW (2008) Macrophages define the invasive microenvironment in breast cancer. J Leukoc Biol 84:623-630 .
Crossref   Medline   1st Citation   2nd  

Powell AE, Anderson EC, Davies PS, Silk AD, Pelz C, Impey S, Wong MH (2011) Fusion between intestinal epithelial cells and macrophages in a cancer context results in nuclear reprogramming. Cancer Res 71:1497-1505 .
Crossref   Medline   1st Citation   2nd  

Preusser M, de Ribaupierre S, Wohrer A, Erridge SC, Hegi M, Weller M, Stupp R (2011) Current concepts and management of glioblastoma. Ann Neurol 70:9-21 .
Crossref   Medline   1st Citation  

Qian BZ, Pollard JW (2010) Macrophage diversity enhances tumor progression and metastasis. Cell 141:39-51 .
Crossref   Medline   1st Citation   2nd  

Rachkovsky M, Pawelek J (1999) Acquired melanocyte stimulating hormone-inducible chemotaxis following macrophage fusion with Cloudman S91 melanoma cells. Cell Growth Differ 10:517-524 .
Medline   1st Citation  

Rachkovsky M, Sodi S, Chakraborty A, Avissar Y, Bolognia J, McNiff JM, Platt J, Bermudes D, Pawelek J (1998) Melanoma×macrophage hybrids with enhanced metastatic potential. Clin Exp Metastasis 16:299-312 .
Medline   1st Citation   2nd   3rd   4th   5th   6th  

Raychaudhuri B, Rayman P, Ireland J, Ko J, Rini B, Borden EC, Garcia J, Vogelbaum MA, Finke J (2011) Myeloid-derived suppressor cell accumulation and function in patients with newly diagnosed glioblastoma. Neuro Oncol 13:591-599 .
Crossref   Medline   1st Citation   2nd   3rd   4th  

Ricci-Vitiani L, Pallini R, Biffoni M, Todaro M, Invernici G, Cenci T, Maira G, Parati EA, Stassi G, Larocca LM, De Maria R (2010) Tumour vascularization via endothelial differentiation of glioblastoma stem-like cells. Nature 468:824-828 .
Crossref   Medline   1st Citation  

Roggendorf W, Strupp S, Paulus W (1996) Distribution and characterization of microglia/macrophages in human brain tumors. Acta Neuropathol (Berl) 92:288-293 .
Crossref   1st Citation   2nd   3rd  

Rossi ML, Hughes JT, Esiri MM, Coakham HB, Brownell DB (1987) Immunohistological study of mononuclear cell infiltrate in malignant gliomas. Acta Neuropathol (Berl) 74:269-277 .
Crossref   1st Citation   2nd   3rd   4th   5th   6th   7th  

Rubinstein LJ (1972) Tumors of the central nervous system., p 400., Washington, DC: Armed Forces Institute of Pathology.
1st Citation   2nd   3rd   4th   5th  

Sabel M, Felsberg J, Messing-Junger M, Neuen-Jacob E, Piek J (1999) Glioblastoma multiforme at the site of metal splinter injury: a coincidence? Case report. J Neurosurg 91:1041-1044 .
1st Citation  

Scherer H (1940) The forms of growth in gliomas and their practical significance. Brain 63:1-34 .
Crossref   1st Citation   2nd   3rd  

Seyfried TN (2001) Perspectives on brain tumor formation involving macrophages, glia, and neural stem cells. Perspect Biol Med 44:263-282 .
Crossref   Medline   1st Citation   2nd   3rd   4th   5th   6th   7th   8th  

Seyfried TN, Kiebish MA, Marsh J, Shelton LM, Huysentruyt LC, Mukherjee P (2011) Metabolic management of brain cancer. Biochim Biophys Acta 1807:577-594 .
Crossref   Medline   1st Citation   2nd  

Seyfried TN, Shelton LM (2010) Cancer as a metabolic disease. Nutr Metab (Lond) 7:7 .
Crossref   Medline   1st Citation   2nd   3rd   4th   5th   6th   7th   8th   9th   10th   11th   12th  

Seyfried TN, Shelton LM, Mukherjee P (2010) Does the existing standard of care increase glioblastoma energy metabolism? Lancet Oncol 11:811-813 .
Crossref   Medline   

Shabo I, Olsson H, Sun XF, Svanvik J (2009) Expression of the macrophage antigen CD163 in rectal cancer cells is associated with early local recurrence and reduced survival time. Int J Cancer 125:1826-1831 .
Crossref   Medline   1st Citation   2nd  

Shelton LM, Huysentruyt LM, Mukherjee P, Seyfried TN (2010a) Calorie restriction as an anti-invasive therapy for malignant brain cancer in the VM mouse. ASN NEURO 2:e00038 .

Shelton LM, Mukherjee P, Huysentruyt LC, Urits I, Rosenberg JA, Seyfried TN (2010b) A novel pre-clinical in vivo mouse model for malignant brain tumor growth and invasion. J Neurooncol 99:165-176 .
Crossref   Medline   1st Citation   2nd   3rd   4th   5th   6th   7th   8th  

Shinonaga M, Chang CC, Suzuki N, Sato M, Kuwabara T (1988) Immunohistological evaluation of macrophage infiltrates in brain tumors. Correlation with peritumoral edema. J Neurosurg 68:259-265 .
Crossref   Medline   1st Citation  

Sica A, Saccani A, Mantovani A (2002) Tumor-associated macrophages: a molecular perspective. Int Immunopharmacol 2:1045-1054 .
Crossref   Medline   1st Citation  

Sica A, Schioppa T, Mantovani A, Allavena P (2006) Tumour-associated macrophages are a distinct M2 polarised population promoting tumour progression: potential targets of anti-cancer therapy. Eur J Cancer 42:717-727 .
Crossref   Medline   1st Citation   2nd  

Singh KK, Kulawiec M, Still I, Desouki MM, Geradts J, Matsui S (2005) Inter-genomic cross talk between mitochondria and the nucleus plays an important role in tumorigenesis. Gene 354:140-146 .
Crossref   Medline   1st Citation  

Steinhaus J (1981) Ueber carcinom-einschlusse. Virchows Arch 126:533-535 .
1st Citation   2nd  

Stossel T (1999) Mechanical responses of white blood cells. In: In Inflammation: Basic Principals and Clinical Correlates (Snyderman J, ed), pp 661-679., New York: Lippincott Williams and Wilkins.
1st Citation  

Strojnik T, Kavalar R, Lah TT (2006) Experimental model and immunohistochemical analyses of U87 human glioblastoma cell xenografts in immunosuppressed rat brains. Anticancer Res 26:2887-2900 .
Medline   1st Citation   2nd   3rd   4th   5th  

Strojnik T, Kavalar R, Zajc I, Diamandis EP, Oikonomopoulou K, Lah TT (2009) Prognostic impact of CD68 and kallikrein 6 in human glioma. Anticancer Res 29:3269-3279 .
Medline   1st Citation   2nd   3rd   4th   5th  

Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, Ludwin SK, Allgeier A, Fisher B, Belanger K, Hau P, Brandes AA, Gijtenbeek J, Marosi C, Vecht CJ, Mokhtari K, Wesseling P, Villa S, Eisenhauer E, Gorlia T, Weller M, Lacombe D, Cairncross JG, Mirimanoff RO (2009) Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol 10:459-466 .
Crossref   Medline   1st Citation  

Taha M, Ahmad A, Wharton S, Jellinek D (2005) Extra-cranial metastasis of glioblastoma multiforme presenting as acute parotitis. Br J Neurosurg 19:348-351 .
Crossref   Medline   1st Citation   2nd   3rd  

Takeuchi H, Kubota T, Kitai R, Nakagawa R, Hashimoto N (2008) CD98 immunoreactivity in multinucleated giant cells of glioblastomas: an immunohistochemical double labeling study. Neuropathology 28:127-131 .
Crossref   Medline   1st Citation   2nd   3rd   4th   5th  

Talmadge JE, Donkor M, Scholar E (2007) Inflammatory cell infiltration of tumors: Jekyll or Hyde. Cancer Metastasis Rev 3–4:373-400 .
1st Citation   2nd  

Tso CL, Shintaku P, Chen J, Liu Q, Liu J, Chen Z, Yoshimoto K, Mischel PS, Cloughesy TF, Liau LM, Nelson SF (2006) Primary glioblastomas express mesenchymal stem-like properties. Mol Cancer Res 4:607-619 .
Crossref   Medline   1st Citation   2nd  

van Landeghem FK, Maier-Hauff K, Jordan A, Hoffmann KT, Gneveckow U, Scholz R, Thiesen B, Bruck W, von Deimling A (2009) Post-mortem studies in glioblastoma patients treated with thermotherapy using magnetic nanoparticles. Biomaterials 30:52-57 .
Crossref   Medline   1st Citation   2nd   3rd   4th  

Vignery A (2000) Osteoclasts and giant cells: macrophage–macrophage fusion mechanism. Int J Exp Pathol 81:291-304 .
Crossref   Medline   1st Citation   2nd   3rd   4th  

Vignery A (2005) Macrophage fusion: are somatic and cancer cells possible partners? Trends Cell Biol 15:188-193 .
Crossref   Medline   1st Citation   2nd   3rd   4th  

Vural G, Hagmar B, Walaas L (1996) Extracranial metastasis of glioblastoma multiforme diagnosed by fine-needle aspiration: a report of two cases and a review of the literature. Diagn Cytopathol 15:60-65 .
Crossref   Medline   1st Citation  

Warburg O (1931) The Metabolism of Tumours, p 327., New York: .
1st Citation  

Warburg O (1956) On the origin of cancer cells. Science 123:309-314 .
Crossref   Medline   1st Citation  

Watters JJ, Schartner JM, Badie B (2005) Microglia function in brain tumors. J Neurosci Res 81:447-455 .
Crossref   Medline   1st Citation  

Waziri A (2010) Glioblastoma-derived mechanisms of systemic immunosuppression. Neurosurg Clin N Am 21:31-42 .
Crossref   Medline   1st Citation  

Wood GW, Morantz RA (1979) Immunohistologic evaluation of the lymphoreticular infiltrate of human central nervous system tumors. J Natl Cancer Inst 62:485-491 .
Medline   1st Citation  

Yang L, DeBusk LM, Fukuda K, Fingleton B, Green-Jarvis B, Shyr Y, Matrisian LM, Carbone DP, Lin (2004) Expansion of myeloid immune suppressor Gr+CD11b+ cells in tumor-bearing host directly promotes tumor angiogenesis. Cancer Cell 6:409-421 .
Crossref   Medline   1st Citation  

Yates AJ (1992) An overview of principles for classifying brain tumors. Mol-Chem-Neuropathol 17:103-120 .
Crossref   Medline   1st Citation  

Yilmaz Y, Lazova R, Qumsiyeh M, Cooper D, Pawelek J (2005) Donor Y chromosome in renal carcinoma cells of a female BMT recipient: visualization of putative BMT-tumor hybrids by FISH. Bone Marrow Transplant 35:1021-1024 .
Crossref   Medline   1st Citation   2nd  

Youness E, Barlogie B, Ahearn M, Trujillo JM (1980) Tumor cell phagocytosis. Its occurrence in a patient with medulloblastoma. Arch Pathol Lab Med 104:651-653 .
Medline   1st Citation   2nd   3rd  

Yuan X, Curtin J, Xiong Y, Liu G, Waschsmann-Hogiu S, Farkas DL, Black KL, Yu JS (2004) Isolation of cancer stem cells from adult glioblastoma multiforme. Oncogene 23:9392-9400 .
Crossref   Medline   1st Citation  

Zagzag D, Esencay M, Mendez O, Yee H, Smirnova I, Huang Y, Chiriboga L, Lukyanov E, Liu M, Newcomb EW (2008) Hypoxia- and vascular endothelial growth factor-induced stromal cell-derived factor-1alpha/CXCR4 expression in glioblastomas: one plausible explanation of Scherer's structures. Am J Pathol 173:545-560 .
Crossref   Medline   1st Citation  

Zhen L, Yufeng C, Zhenyu S, Lei X (2010) Multiple extracranial metastases from secondary glioblastoma multiforme: a case report and review of the literature. J Neurooncol 97:451-457 .
Crossref   Medline   1st Citation   2nd  

Zimmer C, Weissleder R, Poss K, Bogdanova A, Wright SC Jr, Enochs WS (1995) MR imaging of phagocytosis in experimental gliomas. Radiology 197:533-538 .
Medline   1st Citation  


Received 22 April 2011/1 August 2011; accepted 12 August 2011

Published as ASN NEURO Immediate Publication 12 August 2011, doi:10.1042/AN20110011


©2011 The Author(s) This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial Licence ( http://creativecommons.org/licenses/by-nc/2.5/ ) which permits unrestricted non-commercial use, distribution and reproduction in any medium, provided the original work is properly cited.