Tumour-Associated Macrophages
Toby Lawrence • Thorsten Hagemann Editors
Tumour-Associated Macrophages
Editors Toby Lawrence Centre d’Immunologie Marseille Luminy Inserm-CNRS-Universitie de Mediteranee Inflammation Biology Group Parc Scientifique de Luminy, Case 906 Marseille Cedex 9, France
[email protected]
Thorsten Hagemann Centre for Cancer and Inflammation Barts Cancer Institute, Barts and the London School of Medicine and Dentistry Queen Mary University of London London, EC1M 6BQ, UK
[email protected]
ISBN 978-1-4614-0661-7 e-ISBN 978-1-4614-0662-4 DOI 10.1007/978-1-4614-0662-4 Springer New York Dordrecht Heidelberg London Library of Congress Control Number: 2011937444 © Springer Science+Business Media, LLC 2012 All rights reserved. This work may not be translated or copied in whole or in part without the written permission of the publisher (Springer Science+Business Media, LLC, 233 Spring Street, New York, NY 10013, USA), except for brief excerpts in connection with reviews or scholarly analysis. Use in connection with any form of information storage and retrieval, electronic adaptation, computer software, or by similar or dissimilar methodology now known or hereafter developed is forbidden. The use in this publication of trade names, trademarks, service marks, and similar terms, even if they are not identified as such, is not to be taken as an expression of opinion as to whether or not they are subject to proprietary rights. Printed on acid-free paper Springer is part of Springer Science+Business Media (www.springer.com)
Preface
Macrophages are tissue resident phagocytes derived from blood monocytes; they have diverse functions in development and immunity and display enormous phenotypic heterogeneity. Macrophages in different tissues have specialized and specific functions that support organ development and physiology, for example, kupffer cells in the liver filter debris from the blood and aid liver regeneration after injury, Langerhans cells in the skin are important immune sentinel cells and mediate immune surveillance, osteoclasts mediate bone morphogenesis, and microglia in the brain support the development and maintenance of neuronal networks. In response to inflammation or injury, monocytes are recruited into tissue and differentiate locally into macrophages and depending on the nature of the insult or injury these macrophages may acquire distinct phenotypes. Tumours are frequently infiltrated by large number of macrophages and in most cases this is linked with tumour progression and poor prognosis. Macrophage polarization is a poorly defined phenomenon; the mediators and mechanisms that maintain the phenotype of distinct macrophage subsets in both physiology and disease remain to be described. Based primarily on in vitro studies, two particular macrophage phenotypes have been described: “classically” activated or M1 macrophages are characterized by the production of pro-inflammatory cytokines and increased microbicidal or even tumouricidal activity. The second, “alternatively” activated or M2 macrophages, in contrast produce anti-inflammatory cytokines and are linked with angiogenesis, tissue repair, and remodeling. These polarized phenotypes have been described based on in vitro stimulation of macrophages with either interferon (IFN) g, in the case of M1 macrophages, or interleukin (IL)-4 for M2 macrophages. It still is not clear what correlates these populations have in vivo and their physiological relevance remains ambiguous. While these classifications have been useful in that they allow the functional grouping of different macrophage phenotypes, M1 macrophages being proinflammatory cells and M2 macrophages linked with trophic functions and wound healing, there are undoubtedly several intermediates between these polarized phenotypes. However, this classification is too restrictive and it is clear that the functional diversity macrophages in vivo may not be associated with these distinct phenotypic subsets. In fact, the question remains in the context of inflammation and v
vi
Preface
tumours if “the macrophage” merely displays functional plasticity within tissue responding to environmental cues, or distinct stable subsets of macrophages exist with specialized functions. This issue is particularly pertinent in the case of TAM; these cells often display an M2-like phenotype associated with trophic functions promoting tumour angiogenesis, invasion, and metastasis. However, TAM also often produce pro-inflammatory cytokines and have been associated with the promotion of inflammation-associated cancer. This volume provides an overview of current research on the form and function of TAM, highlighting both the mechanistic roles they play in carcinogenesis and tumour progression as well as the molecular mechanisms that control their phenotype and function. Marseille, France London, UK
Toby Lawrence Thorsten Hagemann
Contents
Part I
Form and Function
1
Macrophage Phenotype in Tumours ..................................................... Hsi-Hsien Lin and Siamon Gordon
2
Role of Tumour-Associated Macrophages in the Regulation of Angiogenesis ........................................................................................ Russell Hughes, Hsin-Yu Fang, Munitta Muthana, and Claire E. Lewis
3
The Role of Tumour-Associated Macrophages in Malignant Invasion ............................................................................. Claudia Binder
3
17
31
4
Tumour-Induced Immune Suppression by Myeloid Cells................... Serena Zilio, Giacomo Desantis, Mariacristina Chioda, and Vincenzo Bronte
49
5
TAM: A Moving Clinical Target ............................................................ Simon Hallam and Thorsten Hagemann
63
Part II
Mechanisms of Action
6
Arginine Metabolism and Tumour-Associated Macrophages............. Melissa Phillips and Peter W. Szlosarek
7
Indoleamine 2,3-Dioxygenase Amino Acid Metabolism and Tumour-Associated Macrophages: Regulation in Cancer-Associated Inflammation and Immune Escape .................. George C. Prendergast, Richard Metz, Mee Young Chang, Courtney Smith, Alexander J. Muller, and Suzanne Ostrand-Rosenberg
77
91
vii
viii
Contents
8
Vascular Endothelial Growth Factor and Tumour-Associated Macrophages............................................................................................ 105 Christian Stockmann and Randall S. Johnson
Part III
Molecular Regulation
9
TLR Signaling and Tumour-Associated Macrophages........................ 119 Oscar R. Colegio and Ruslan Medzhitov
10
SHIP and Tumour-Associated Macrophages ....................................... 135 Victor W. Ho, Melisa J. Hamilton, Etsushi Kuroda, Jens Ruschmann, Frann Antignano, Vivian Lam, and Gerald Krystal
11
NF-KappaB-Mediated Regulation of Tumour-Associated Macrophages: Mechanisms and Significance ....................................... 153 Antonio Sica and Alberto Mantovani
12
Role of Hypoxia-Inducible Transcription Factors in TAM Function ..................................................................................... 167 Nadine Rohwer and Thorsten Cramer
Index ................................................................................................................. 183
Contributors
Frann Antignano The Terry Fox Laboratory, B.C. Cancer Agency, Vancouver, BC V5Z 1L3, Canada Claudia Binder Department of Hematology/Oncology, University of Göttingen, Göttingen 37099, Germany Vincenzo Bronte Verona University Hospital and Department of Pathology, Immunology Section, Piazzale L.A. Scuro 10, 37134 Verona, Italy Mee Young Chang Lankenau Institute for Medical Research, Wynnewood, PA, USA Mariacristina Chioda Istituto Oncologico Veneto (IOV), IRCCS, Via Gattamelata 64, 35128 Padova, Italy Oscar R. Colegio Department of Immunobiology, Howard Hughes Medical Institute, Yale University, New Haven, CT, USA Thorsten Cramer Medizinische Klinik mit Schwerpunkt Hepatologie und Gastroenterologie and Molekulares Krebsforschungszentrum, Charité – Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany Giacomo Desantis Department of Oncology and Surgical Sciences, University of Padova, Via Gattamelata 64, 35128 Padova, Italy Hsin-Yu Fang Academic Unit of Inflammation & Tumour Targeting, University of Sheffield Medical School, Beech Hill Road, Sheffield S10 2RX, UK Siamon Gordon Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
ix
x
Contributors
Thorsten Hagemann Centre for Cancer and Inflammation, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK Simon Hallam Centre for Cancer and Inflammation, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK Melisa J. Hamilton The Terry Fox Laboratory, B.C. Cancer Agency, Vancouver, BC V5Z 1L3, Canada Victor W. Ho The Terry Fox Laboratory, B.C. Cancer Agency, Vancouver, BC V5Z 1L3, Canada Russell Hughes Academic Unit of Inflammation & Tumour Targeting, University of Sheffield Medical School, Beech Hill Road, Sheffield S10 2RX, UK Randall S. Johnson Molecular Biology Section, Division of Biology, University of California, San Diego, CA 92093, USA Gerald Krystal The Terry Fox Laboratory, B.C. Cancer Agency, Vancouver, BC V5Z 1L3, Canada Etsushi Kuroda The Terry Fox Laboratory, B.C. Cancer Agency, Vancouver, BC V5Z 1L3, Canada Vivian Lam The Terry Fox Laboratory, B.C. Cancer Agency, Vancouver, BC V5Z 1L3, Canada Claire E. Lewis Academic Unit of Inflammation & Tumour Targeting, University of Sheffield Medical School, Beech Hill Road, Sheffield S10 2RX, UK Hsi-Hsien Lin Department of Microbiology and Immunology, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, Taiwan Alberto Mantovani Istituto Clinico Humanitas IRCCS, via Manzoni 56, 20089 Rozzano, Italy Department of Translational Medicine, University of Milan, Milan, Italy Ruslan Medzhitov Department of Immunobiology, Howard Hughes Medical Institute, Yale University, New Haven, CT, USA Richard Metz New Link Genetics Corporation, Ames, IA, USA Alexander J. Muller Lankenau Institute for Medical Research, Wynnewood, PA, USA Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA Munitta Muthana Academic Unit of Inflammation & Tumour Targeting, University of Sheffield Medical School, Beech Hill Road, Sheffield S10 2RX, UK
Contributors
xi
Suzanne Ostrand-Rosenberg Department of Biological Sciences, University of Maryland at Baltimore, Baltimore, MD, USA Melissa Phillips Centre for Molecular Oncology and Imaging, Institute of Cancer, Barts and The London School of Medicine, Queen Mary College, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK George C. Prendergast Lankenau Institute for Medical Research, Wynnewood, PA, USA Department of Pathology, Anatomy & Cell Biology, Jefferson Medical School, Philadelphia, PA, USA Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA Nadine Rohwer Medizinische Klinik mit Schwerpunkt Hepatologie und Gastroenterologie and Molekulares Krebsforschungszentrum, Charité – Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany Jens Ruschmann The Terry Fox Laboratory, B.C. Cancer Agency, Vancouver, BC V5Z 1L3, Canada Antonio Sica Istituto Clinico Humanitas IRCCS, via Manzoni 56, 20089 Rozzano, Italy DiSCAFF, University of Piemonte Orientale A. Avogadro, 28100 Novara, Italy Courtney Smith Lankenau Institute for Medical Research, Wynnewood, PA, USA Christian Stockmann Molecular Biology Section, Division of Biology, University of California, San Diego, CA 92093, USA Institut für Physiologie, University of Duisburg-Essen, Duisburg-Essen, Germany Peter W. Szlosarek Centre for Molecular Oncology and Imaging, Institute of Cancer, Barts and The London School of Medicine, Queen Mary College, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK Serena Zilio Department of Oncology and Surgical Sciences, University of Padova, Via Gattamelata 64, 35128 Padova, Italy Istituto Oncologico Veneto (IOV), IRCCS, Via Gattamelata 64, 35128 Padova, Italy
Part I
Form and Function
Chapter 1
Macrophage Phenotype in Tumours Hsi-Hsien Lin and Siamon Gordon
Introduction Monocytes and macrophages are a prominent component of the host response to, and manipulation by, tumour cells (Gordon and Martinez 2010; Mantovani et al. 2008). Together with other myeloid and lymphoid cells, they influence tumour development, both positively and negatively. Although the factors that determine outcome of the host–tumour relationship are not well understood, many tumours recruit immature myelomonocytic cells, block their differentiation, subvert their cytotoxicity, suppress lymphoid effector cells, and induce peripheral tolerance. In addition, they mimic and utilise macrophage functions to enhance growth, produce a stroma and promote angiogenesis, local invasion of their micro-environment and metastasis (Qian and Pollard 2010). In particular, the uptake of apoptotic tumour cells can suppress anti-tumour inflammatory responses by TGF-beta and prostaglandins. The macrophage growth factor CSF-1 stimulates macrophage recruitment and modulates its phenotype, limiting the activation of cytotoxic effector functions; Interleukin-4 and -13, acting through common and specific receptors, induce a trophic, alternative M2 activation phenotype, distinct from cytotoxic M1, classically activated (Interferon-gamma-dependent) macrophages (Reviewed by Gordon and Martinez 2010). Interleukin-10 is a potent deactivator of macrophage inflammatory properties, whereas TGF-beta, another deactivator, promotes fibrosis and vascular remodelling.
H.-H. Lin Department of Microbiology and Immunology, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, Taiwan S. Gordon (*) Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK e-mail:
[email protected] T. Lawrence and T. Hagemann (eds.), Tumour-Associated Macrophages, DOI 10.1007/978-1-4614-0662-4_1, © Springer Science+Business Media, LLC 2012
3
4
H.-H. Lin and S. Gordon
A wide range of chemokines such as MCP-1, often produced by tumour cells, attract mononuclear and myeloid cells. TNF-alpha has also been implicated in tumourigenesis (Mantovani et al. 2008). Monocyte-macrophages express a wide range of plasma membrane receptors which govern their response to chemokines, cytokines, growth factors and other tumour- and host-derived ligands (Taylor et al. 2005). Other membrane molecules regulate cellular responses to diverse agonists, inhibiting or enhancing macrophage effector mechanisms. These molecules provide useful markers for the presence, characterisation and possible functions of tumour-associated monocyte/macrophages, and targets for therapeutic intervention. In this review, we present a range of possible molecular markers for in situ characterisation, with special reference to the EGF-TM7 family of myeloid G protein-coupled receptors (GPCRs) with large extracellular domains. Their potential is reviewed in the context of macrophage heterogeneity and plasticity (Auffray et al. 2009; Gordon and Taylor 2005) and the experimental analysis of macrophage phenotype in tumours.
Macrophage Heterogeneity in Tumours Some of the earliest studies on the presence and possible role of macrophages in tumours were undertaken by Evans and Alexander, Mantovani, Pollard and their collaborators (Mantovani et al. 2008; Qian and Pollard 2010). The topic received renewed impetus in recent years with the work of Bronte (Peranzoni et al. 2010) and Gabrilovitch (Gabrilovich and Nagaraj 2009) and their groups. Important contributions came from Balkwill (Mantovani et al. 2008), Lewis (Coffelt et al. 2009), Karin (Grivennikov et al. 2010) and Coussens (Coussens and Werb 2002), Rosenberg (Domachowske et al. 2000) and Joyce (Joyce and Pollard 2009). A great deal of confusion has resulted from myeloid cell heterogeneity and terms such as TAMs (tumour-associated macrophages) and MDSC (myeloid-derived suppessor cells) are currently in wide use. The former embraces cells with macrophagerestricted markers such as F4/80 and alternative activation markers such as Arginase-1 (Gordon and Martinez 2010); the latter term includes cells with immature monocytic phenotype (Gr-1 low) and granulocyte characteristics (Gr-1 high). Mononuclear phagocyte heterogeneity associated with stages of differentiation and activation status gives rise to considerable plasticity within and among cell populations. Studies by Geissmann (Geissmann et al. 2010) and Jung (Varol et al. 2009) have utilised the fractalkine receptor, in combination with other chemokine receptors, to define precursors of tissue macrophages during development, adult life, physiologically and in various inflammatory and pathologic states. Studies by Nussenzweig (Dudziak et al. 2007), Merad (Merad 2010; Merad and Manz 2009) and their colleagues have helped to clarify the origins and population dynamics of myeloid dendritic cells, vis-à-vis monocyte/macrophages. Their fluorescence and transgenic methods will be useful to trace precursors of myelomonocytic cells in mouse tumours.
1
Macrophage Phenotype in Tumours
5
Tumours are obviously heterogeneous themselves, not only in their ability to invade (benign or malignant), but also in their micro-environment (lung, liver, bone and lymph nodes), origin (epithelial, mesenchymal and haemopoietic), vascularisation, within individual tumours as well as among different primary or secondary tumour populations. Other differences pertain as tumours induce matrix synthesis and catabolism, undergo hypoxia, apoptosis and necrosis. The concomitant presence of CD4+, CD8+ lymphocytes, FoxP3 positive suppressor cells, as well as innate lymphoid cells (NKT and NK cells) modulates myeloid cells, reciprocally. Tumour cells themselves often express characteristic properties of leukocytes that can contribute to their migration and invasion. Macrophages can also be tolerogenic and contribute to lymphocyte suppression by cell contact or secretory products. Dendritic cell maturation and antigen presentation can also be subverted by tumour- or other myeloid-derived products. Apart from the above considerations, many difficulties hinder experimental analysis of macrophage phenotype in tumours. Ideally, one should study naturally occurring tumours in situ, rather than transplantable models. Isolation of myeloid cells, especially macrophages, is difficult and prone to artefact, particularly if FACS analysis is not combined with immunocytochemistry in situ. The use of oncogenic transgenes, e.g. by Hanahan and colleagues (Hanahan 1989) made it possible to synchronise defined stages of experimental tumours. Mouse models do not necessarily replicate human tumours, often studied at late stages, or after chemotherapy and irradiation. Finally, macrophage markers used in the mouse and human may differ markedly between species. The interactions between macrophages and tumour cells result in novel gene expression profiles in both cell types, only partially reproduced during co-cultivation in vitro. Microarray and proteomic analyses, while powerful indicators of signatures, e.g. of type 1 interferon activation pathways, need refinement. The traditional methods of morphologic, diagnostic pathology are undergoing rapid advances, but have not yet progressed to interpret function at the single-cell level sufficiently.
Membrane Markers for Macrophages in Tumours Given the above caveats, we present a list of validated and candidate antigen markers to define macrophage heterogeneity in tumours (Table 1.1). We feel that the present focus reported in the literature is too narrow, that FACS analysis of isolated macrophages is insufficient and that whilst antigens are reasonably well-defined in the mouse, markers for human antigens are limited and not sufficiently characterised. Monoclonal and polyclonal antibodies for FACS and western blotting are not necessarily suitable for immunocytochemistry. Tissue preservation, antigen stability and antibody staining need to be optimised for each epitope. Table 1.1 includes members of a range of molecular families, varying in cell specificity. Markers include opsonic and non-opsonic phagocytic receptors, lectins and scavenger receptors, as well as cytokine receptors and other differentiation antigens, with some functional correlates.
6
H.-H. Lin and S. Gordon
Table 1.1 Selected membrane markers for macrophages in tumours Molecule Property Comment F4/80 EGF-TM7/adhesion-GPCR Peripheral tolerance, MI subpopulation CD97 EGF-TM7/adhesion-GPCR Myeloid, other cells EMR2 EGF-TM7/adhesion-GPCR Human, not mouse, aberrant in breast cancers CD68 LAMP family Pan-MI and DC, some tumours Gr-1 Ly-6 family PMN, immature monocytes Polymorphic, PMN, immature monocytes 7/4 Ly-6 family (Rosas et al. 2010) Siglec-1 IgSF Sialyl-ligand, e.g. Muc-1 CD163 SRCR family Glucocorticoid, IL-10 induced CD200/CD200R IgSF Receptor/ligand pair, negative regulator FcR IgSF Activatory/inhibitory CR3 Beta-2 integrin Opsonic and non-opsonic phagocytosis Adhesion SR-A SRCR family Clearance apoptotic cells, CSF-1 upregn MARCO SRCR family Adhesion, induced via TLRs CD36 Bispanner SR-B Ox-LDL, Apoptotic, Thrombospondin R MR C-type lectin Alternative activation marker Dectin-1 C-type lectin-like Beta-glucan R, ITAM-like domain Dectin-2 C-type lectin-like Subset macrophages, Mannose-ligand TLRs Leucine-rich repeat Sensor exogenous, host ligands IL-4/13 R Cytokine R Common, specific R, alternative activation CSF-1R Receptor tyrosine kinase fms GM-CSF R Haemopoietic R Fc-GMCSF chimeric ligand (Rosas et al. 2007) CX3CR1 GPCR Membrane bound fractalkine R CCR2 GPCR MCP-1 ligand
Curiously, in some cases, e.g. CD68, non-haemopoietic tumour cells are able to express leukocyte markers ectopically. Giant cells and hybrids arising from fusion of tumour cells and macrophages provide another mechanism for aberrant marker expression. Some of these markers have been utilised in inflammatory and infectious models in the mouse but not in tumours. The need for co-localisation and double/multiple labelling, so useful in FACS, is more difficult to achieve in immunohistochemistry, which also lacks quantitation. Laser capture microscopy and tissue arrays may overcome some of these difficulties.
EGF-TM7 Receptors and Tumour-Associated Macrophages The mouse differentiation antigen F4/80 is a well-characterised marker for mouse macrophages and has been implicated in peripheral tolerance in a non-tumour model (see below for references). The mouse and human antigen CD97 is not only associated
1
Macrophage Phenotype in Tumours
7
with myeloid cell differentiation and activation, but has also been studied in a variety of tumour settings in vivo. The closely related antigen EMR2 provides a sensitive marker for macrophage identification in human tissues. We review the common and selective characteristics of these molecules in detail, in relation to tissue specificity and as potential markers of macrophage heterogeneity and function in tumour–host interactions.
Common Characteristics of the EGF-TM7 Receptors F4/80, EMR2 and CD97 all belong to the group of EGF-TM7 molecules that make up the second largest GPCR sub-family in man, the adhesion-GPCRs (Fig. 1.1) (McKnight and Gordon 1996; McKnight and Gordon 1998; Stacey et al. 2000; Yona et al. 2008a; Bjarnadottir et al. 2004; Bjarnadottir et al. 2007). The EGF-TM7 receptors share many common characteristics in protein structure, cellular function
E E E E E
E
E
E E
E E
E
E
E
E
E E
F4/80
Expression
EGF-like domains GPS proteolysis Ligand(s)
Function(s)
EMR2
Monocytes Resident tissue macrophages (Kupffer cells, microglia, etc.) Immature dendritic cells Eosinophils TAMs 7
Monocytes Macrophages Dendritic cells Neutrophils
5
No Unknown
Yes Chondroitin sulfate
Induction of Peripheral tolerance
Priming of neutrophils
CD97
Lymphocytes Monocytes Macrophages Dendritic cells Neutrophils Smooth muscle cells Tumour cells 5 Yes CD55, Chondroitin sulfate Integrins (A5B1/AvB3 ) Migration and homeostasis of PMNs, HSC mobilization, CD4+ T cell co-stimulation, Angiogenesis, Tumour invasion
Fig. 1.1 Characteristics of F4/80, EMR2 and CD97. The three receptors are represented schematically. The EGF-like (E) motifs are shown as triangles, the GPS motif as a triangle with two disulfide bonds and the 7TM domain is represented by seven cylinders
8
H.-H. Lin and S. Gordon
and expression patterns (Stacey et al. 2000; Yona et al. 2008a). As suggested by the nomenclature, EGF-TM7 molecules are chimeric proteins composed of two major protein modules, namely the epidermal growth factor (EGF)-like motifs and the seven-span transmembrane (7TM) domains. A typical EGF-TM7 molecule contains tandem repeats of EGF-like motifs at the N terminus followed by a stalk region, which connects to a 7TM domain (McKnight and Gordon 1996; McKnight and Gordon 1998). Most of the EGF-like motifs of the EGF-TM7 molecules belong to the Ca2+binding subtype usually found in extracellular matrix proteins. Thus, Ca2+ binding is important for the cellular function of these receptors (Lin et al. 2001). The 7TM domains of the EGF-TM7 molecules share strong sequence similarity to the class B or secretin-like GPCRs. However, recent phylogenetic analyses have suggested an independent evolutionary lineage and group them with others to form the adhesionGPCR sub-family (Bjarnadottir et al. 2004; Bjarnadottir et al. 2007; Fredriksson et al. 2003). The stalk region between the EGF-like motifs and the 7TM domain usually contains numerous Ser and Thr residues and N-link glycosylation sites. Therefore, it is believed to be heavily decorated with O- and N-link glycans and is thought as a rigid structure. In addition, a highly conserved Cys-rich motif of ~50 residues located immediately upstream of the first TM region has been identified. A posttranslational auto-proteolytic reaction at this Cys-rich motif would cleave the receptor molecule into an extracellular- and 7TM-subunits (Lin et al. 2004). The specific proteolytic cleavage site is, therefore, named the GPCR proteolysis site (GPS) (Krasnoperov et al. 1999). It is thought that the EGF-TM7 receptors would bind to specific cellular ligands through the extracellular domain, especially by the EGF-like motifs, which in turn activate the 7TM domain to transmit intracellular signals. The majority of the EGF-TM7 receptors are restrictedly expressed in myeloid cells, including monocytes, macrophages, polymorphonuclear cells and dendritic cells (Stacey et al. 2000; Yona et al. 2008a). Thus, a role in innate as well as adaptive immune functions was predicted for these molecules.
F4/80 The F4/80 molecule is a ~160 kDa cell surface glycoprotein (Austyn and Gordon 1981). However, the full-length F4/80 cDNA predicted a mature protein of 904 amino acid residues with an estimated molecular weight of 99 kDa (Lin et al. 1997; McKnight et al. 1996). It was shown that the ~60 kDa difference in size was due to heavy glycosylation of the molecule as predicted by the multiple O- and N-linked glycosylation sites in the stalk region. In addition, a total of 7 EGF-like motifs, one glycosaminoglycan attachment site and an Arg-Gly-Asp (RGD) motif, were also identified in the extracellular domain of F4/80 (McKnight and Gordon 1996; McKnight and Gordon 1998; McKnight et al. 1996). Despite the presence of a typical
1
Macrophage Phenotype in Tumours
9
GPS motif, no GPS proteolytic cleavage was identified in F4/80 because of the lack of a consensus cleavage site. Hence, unlike typical adhesion-GPCRs, the F4/80 receptor is a single-chain polypeptide. The main cellular functions of F4/80 was found to be involved in the generation of Ag-specific efferent CD8+ regulatory T (Treg) cells responsible for peripheral immune tolerance (Lin et al. 2005). During the induction of peripheral immune tolerance, it is thought that F4/80 is required for the cellular interactions among Ag-presenting cells (APC) and other immune effector cells that lead to the generation of efferent CD8+ Treg cells. In addition, it was also suggested that F4/80 is involved in macrophage-NK interaction in a Listeria monocytogenes infection model (Warschkau and Kiderlen 1999). Overall, F4/80 is believed to play an immuneregulatory role through the interaction with an unidentified cellular ligand expressed on other immune effector cells (Lin et al. 2005; van den Berg and Kraal 2005). F4/80 is one of the best surface markers for the majority of mouse tissue macrophages (Taylor et al. 2005; Gordon and Taylor 2005; Austyn and Gordon 1981). The F4/80 Ag was detected strongly in many tissue macrophage populations, including Kupffer cells in liver, red pulp macrophages in spleen, microglia cells in brain as well as other resident macrophages in bone marrow stroma, gut lamina propria, testis, kidney, lymph nodes and peritoneum. On the other hand, macrophages within T-cell areas of lymph nodes (paracortex), spleen (white pulp) and Peyer’s patches are usually negative for F4/80. Very low levels of F4/80 were expressed in some other resident tissue macrophages such as alveolar macrophages, marginal zone and subcapsular sinus macrophages in the spleen and lymph nodes (Taylor et al. 2005; McKnight and Gordon 1998). Blood monocytes, the precursors of tissue macrophages, express less F4/80 than their tissue counterparts, suggesting that the expression of F4/80 is regulated during differentiation. Furthermore, it was also noted that F4/80 expression is modulated according to the activation status of macrophages. With regards to cells of other hematopoietic origins, no reactivity was observed in lymphoid cells, neutrophils and monocyte-derived osteoclasts. Nevertheless, eosinophils were shown to also express the F4/80 Ag. Likewise, Langerhans cells, a type of dendritic cells (DC) in the epidermis, were found to be F4/80 positive, but the Ag is down-regulated upon subsequent DC maturation and migration to the draining lymph nodes (Taylor et al. 2005; McKnight and Gordon 1998). In mice, F4/80-expressing TAMs have been documented for many types of tumours, either occurred spontaneously or induced experimentally (Qian & Pollard 2010; Mantovani et al. 2002; Rabinovich et al. 2007; Umemura et al. 2008). Therefore, the presence or absence of the F4/80 reactivity has been used to determine the efficiency of macrophage ablation experiments in tumour studies. In addition, F4/80 was also detected in tumour-infiltrating MDSC. The Ag has been useful to define the cellular origin of MDSC in tumours as MDSC was initially thought to represent immature myelomonocytic cells with common monocytic and granulocytic phenotypes. It is now generally accepted that TAMs and MDSC include cells with similar phenotypes to alternatively activated macrophages, which all express F4/80 (Gordon and Martinez 2010; Umemura et al. 2008; Martinez et al. 2009; Sinha et al. 2005).
10
H.-H. Lin and S. Gordon
CD97 The full-length CD97 molecule contains a total of five EGF-like motifs. However, as a result of RNA alternative splicing, three major isoforms containing different combinations of EGF-like motifs were predicted. These include CD97(1, 2, 5), CD97(1, 2, 3, 5) and CD97(1, 2, 3, 4, 5) (Gray et al. 1996; Hamann et al. 1995). Apart from multiple O- and N-linked glycosylation sites, one RGD motif and a complete GPS motif were present in the extracellular region. Hence, CD97 was cleaved at the predicted GPS site into two subunits (Gray et al. 1996). CD97 was found to interact through its extracellular region with several cellular ligands, including CD55 (DAF) (Hamann et al. 1998; Hamann et al. 1996), dermatan sulphate (Kwakkenbos et al. 2005; Stacey et al. 2003) and D5E1/DvE3 integrins (Wang et al. 2005). Interestingly, the CD97-ligand interaction is mostly isoform-specific such that CD55 binds better to CD97(1, 2, 5), while dermatan sulphate only interacts with CD97(1, 2, 3, 4, 5) (Lin et al. 2001; Stacey et al. 2003). CD97–CD55 interaction is species-specific so that human CD97 only reacts with human CD55 but not murine CD55 (Lin et al. 2001). Through the utilisation of specific mAbs, soluble CD97 proteins, and the generation and analysis of knock-out animals, CD97 has been implicated in the cellular migration and homeostasis of polymorphonuclear cells (Kop et al. 2006; Leemans et al. 2004; Wang et al. 2007), hematopoietic stem cell/progenitor cell mobilisation (van Pel et al. 2008a; van Pel et al. 2008b), co-stimulation of CD4+ T cells (Abbott et al. 2007; Capasso et al. 2006) and angiogenesis (Wang et al. 2005). CD97 was also shown to be potentially involved in the pathogenesis of arthritis (Kop et al. 2006). CD97 was first identified as an early activation marker for T and B lymphocytes, which express low levels of CD97 in resting conditions (Gray et al. 1996; Hamann et al. 1995). On the other hand, CD97 is constitutively expressed on granulocytes and monocytes/macrophages (Jaspars et al. 2001). Immunohistochemistry staining of normal human tissues in situ has shown that CD97 is abundantly expressed in resident macrophages of most tissues (Jaspars et al. 2001). These include liver (Kupffer cells and periportal histiocytes), lung (alveolar macrophages), skin (including Langerhans cells), brain (perivascular macrophages but not microglia), kidney (mesangial cells of the glomeruli) and secondary lymphoid organs such as lymph nodes, spleen (red pulp and white pulp macrophages), tonsil and mucosa-associated lymphoid tissues (MALT). Dendritic cells in most of the lymphoid tissues also express CD97, whereas CD97-expressing lymphocytes are mostly restricted in intraepithelial and sub-epithelial locations. Lymphocytes in paracortical areas, follicles and germinal centres are mostly CD97 weak or negative. Outside lymphoid tissues, smooth muscle cells also express CD97. In addition, CD97 was often detected in many types of tumours, including thyroid, gastric, pancreatic, esophageal and colorectal carcinomas (Aust et al. 1997; Aust et al. 2002; Steinert et al. 2002). Most interestingly, stronger CD97 expression levels were usually detected at the invasion fronts of tumours, suggesting a role in tumour cell migration/invasion (Steinert et al. 2002; Galle et al. 2006; Wobus et al. 2006).
1
Macrophage Phenotype in Tumours
11
Specifically, in colorectal and gastric carcinomas, the strongest CD97 staining was identified in disseminated or scattered tumour cells at the invasion front of the tumour. Furthermore, a poorer clinical stage and increased lymph vessel invasion were found to be correlated positively in tumour patients with more CD97-high scattered tumour cells. In the cell level, higher CD97 expression stimulated singlecell motility, enhanced the proteolytic activity of matrix metalloproteinases and secretion of chemokines. Tumour growth in scid mice was enhanced in cells overexpressing CD97 (Steinert et al. 2002; Galle et al. 2006; Wobus et al. 2006). Moreover, CD97 was shown recently to stimulate angiogenesis through binding to D5E1 integrin on endothelial cells (Wang et al. 2005). Briefly, soluble CD97 was tested in a quantitative, directed in vivo angiogenesis assay (DIVAA). It was found that CD97 at 25 and 100 ng/ml is as efficient as bFGF in inducing blood vessel development in vivo. Furthermore, developing tumours derived from CD97expressing cells display a significantly greater vessel density than those from control CD97-negative tumour cells. The angiogenic response of CD97 is most likely mediated by the RGD motif in the stalk as it interacts with the D5E1 and DvE3 integrins. Through these interactions, CD97 promotes the adhesion, migration and invasion of human umbilical vein endothelial cells (HUVECs). Nevertheless, another ligand of CD97, chondroitin sulphate, seems to also contribute to these effects (Wang et al. 2005). A more recent study using CD97 transgenic mice, however, indicates that the role of CD97 in tumourigenesis might be more complicated than we thought earlier (Becker et al. 2010). To investigate the involvement of CD97 in colorectal carcinogenesis, Becker et al. generated transgenic mice that overexpress CD97 specifically in enterocytes (Becker et al. 2010). These animals were then subjected to azoxymethane (AOM)/dextran sodium sulphate (DSS)-induced colitis-associated tumourigenesis. Interestingly, DSS-induced colitis was reduced in transgenic mice when compared with the wild-type control. This reduction was dependent on the copy number of the CD97 transgene. Through ultrastructural and biochemical analyses, it was concluded that CD97 over-expression can enhance the structural integrity of enterocytic adherens junctions, which in turn enforce intestinal epithelial strength leading to the attenuation of experimental colitis.
EMR2 Highly similar to CD97, the full-length EMR2 molecule also contains a total of five EGF-like motifs. A total of four major isoforms, namely EMR2(1, 2), EMR2 (1, 2, 5), EMR2(1, 2, 3, 5) and EMR2(1, 2, 3, 4, 5), were predicted from spliced mRNA sequences (Lin et al. 2000). EMR2 shares ~97% sequence identity in the EGF-like domains with CD97. Interestingly, however, a highly restricted ligand binding specificity was identified for these EGF-TM7 molecules. Thus, EMR2 (1, 2, 5) was shown to bind CD55 with a tenfold weaker affinity than CD97(1, 2, 5), even though the two molecules only differ in three residues (Lin et al. 2001).
12
H.-H. Lin and S. Gordon
Nevertheless, EMR2(1, 2, 3, 4, 5) and CD97(1, 2, 3, 4, 5) were found to bind dermatan sulphate equally well (Stacey et al. 2003). These results suggest potential overlapping and also unique functions for EMR2 and CD97. Phylogenetic analysis has revealed an interesting evolutionary relationship between EMR2 and CD97; although both genes are highly homologous in the human genome, the EMR2 gene is lost in rodents, while the CD97 gene is present in man and mouse. In fact, it seems that EMR2 evolved through gene conversion and duplication by combining part of CD97 and another EGF-TM7 receptor, EMR3. Thus, EMR2 is a chimeric gene evolved relatively recently, probably to meet the need of the immune system in primates (Kwakkenbos et al. 2004; Kwakkenbos et al. 2006). A role for EMR2 in regulating neutrophil activation was found recently (Yona et al. 2008b). Binding of EMR2 receptor by a specific mAb was shown to strongly enhance the inflammatory responses of neutrophils to a panel of stimuli. Interestingly, mAb treatment alone did not activate neutrophils. Hence, EMR2 receptor activation seems to have a priming effect on neutrophil activation. Expression of EMR2 is more restricted than CD97 to myeloid cells (Lin et al. 2000; Chang et al. 2007; Kwakkenbos et al. 2002). Monocytes, macrophages, neutrophils and dendritic cells have all been shown to express EMR2. Specifically, the strongest EMR2 signal was detected on CD16+ blood monocytes, macrophages and BDCA-3+ myeloid dendritic cells. No expression was ever detected in resting or activated lymphocytes. Interestingly, it was found that EMR2 is up-regulated during the differentiation and maturation of macrophages in vitro, but is downregulated during dendritic cell maturation (Chang et al. 2007). EMR2 expression in monocytes and macrophages can be up-regulated by LPS and IL-10 in an IL-10dependent manner. In some limited tissues surveyed, EMR2 was detected in certain macrophage sub-populations of skin, spleen, lung, placenta, and tonsil. Macrophages of liver and kidney do not seem to express EMR2. In inflamed tissues, EMR2 was found in sub-populations of macrophages and neutrophils. Recently, it was shown that foamy macrophages in atherosclerotic vessels and Gaucher cells in spleen express high levels of EMR2. In contrast, multiple sclerosis brain foam cells expressed little if any EMR2, but strong CD97 (van Eijk et al. 2010). Unlike CD97, EMR2 expression in tumours has not been observed much. In fact, in thyroid, gastric, pancreatic, and esophageal carcinomas, many tumour cells are CD97 positive but EMR2 negative (Aust et al. 2002). In some colorectal tumour cell lines and adenocarcinomas, alternatively spliced EMR2 mRNA transcripts were identified, but EMR2 protein expression remained low in the tumour cells (Aust et al. 2003). Within the carcinomas, certain tumour-infiltrating macrophages expressed strong surface EMR2. Interestingly, by staining breast cancer tissue sections, we recently identify strong EMR2 reactivity within tumour cells (Davies et al. unpublished results). The significance of this finding awaits further investigation, but suggests that aberrantly expressed EMR2 could be linked to certain tumour types. In conclusion, members of the myeloid-restricted adhesion-GPCRs, the EGF-TM7 receptors, could be considered as important new molecules associated with tumourigenesis. Based on the structural, functional and expressional characteristics, the
1
Macrophage Phenotype in Tumours
13
EGF-TM7 proteins could serve as the cell surface marker of tumour-infiltrating or tumour-associated myeloid cells (macrophages and neutrophils). These receptors can also be expressed by tumour cells to facilitate the interaction with the tumour microenvironment including cells and extracellular matrix. By doing so, the EGF-TM7 receptors might promote angiogenesis and the migration and invasion of tumour cells. Finally, a potential role of the EGF-TM7 receptors in immunosuppression might provide tumour cells an opportunity to evade immune surveillance.
Conclusion A larger range of molecular markers is available than currently in use, to characterise macrophages within and isolated from tumours. The leap from macrophage marker to function, taking into account the relevant tumour subpopulation actually responsible for tumour progression, is considerable. A great deal of further characterisation of promising markers is needed, especially in humans, who provide a large pool of material of natural history and diversity. Studies on individual macrophage markers, membrane and otherwise, combined with biomarkers in whole blood and tissues, should aid diagnosis and possible targeting of macrophage functions in the future.
References Abbott RJ, Spendlove I, Roversi P, Fitzgibbon H, Knott V, Teriete P, McDonnell JM, Handford PA, Lea SM (2007) Structural and functional characterization of a novel T cell receptor co-regulatory protein complex, CD97–CD55. J Biol Chem 282(30):22023–22032 Auffray C, Sieweke MH, Geissmann F (2009) Blood monocytes: development, heterogeneity, and relationship with dendritic cells. Annu Rev Immunol 27:669–692 Aust G, Eichler W, Laue S, Lehmann I, Heldin NE, Lotz O, Scherbaum WA, Dralle H, Hoang-Vu C (1997) CD97: a dedifferentiation marker in human thyroid carcinomas. Cancer Res 57(9): 1798–1806 Aust G, Steinert M, Schutz A, Boltze C, Wahlbuhl M, Hamann J, Wobus M (2002) CD97, but not its closely related EGF-TM7 family member EMR2, is expressed on gastric, pancreatic, and esophageal carcinomas. Am J Clin Pathol 118:699–707 Aust G, Hamann J, Schilling N, Wobus M (2003) Detection of alternatively spliced EMR2 mRNAs in colorectal tumor cell lines but rare expression of the molecule in colorectal adenocarcinomas. Virchows Arch 443(1):32–37 Austyn JM, Gordon S (1981) F4/80, a monoclonal antibody directed specifically against the mouse macrophage. Eur J Immunol 11(10):805–85 Becker S, Wandel E, Wobus M, Schneider R, Amasheh S, Sittig D, Kerner C, Naumann R, Hamann J, Aust G (2010) Overexpression of CD97 in intestinal epithelial cells of transgenic mice attenuates colitis by strengthening adherens junctions. PLoS One 5(1):e8507 Bjarnadottir TK, Fredriksson R, Hoglund PJ, Gloriam DE, Lagerstrom MC, Schioth HB (2004) The human and mouse repertoire of the adhesion family of G-protein-coupled receptors. Genomics 84(1):23–33 Bjarnadottir TK, Fredriksson R, Schioth HB (2007) The adhesion GPCRs: a unique family of G protein-coupled receptors with important roles in both central and peripheral tissues. Cell Mol Life Sci 64(16):2104–2119
14
H.-H. Lin and S. Gordon
Capasso M, Durrant LG, Stacey M, Gordon S, Ramage J, Spendlove I (2006) Costimulation via CD55 on human CD4+ T cells mediated by CD97. J Immunol 177(2):1070–1077 Chang GW, Davies JQ, Stacey M, Yona S, Bowdish DM, Hamann J, Chen TC, Lin CY, Gordon S, Lin HH (2007) CD312, the human adhesion-GPCR EMR2, is differentially expressed during differentiation, maturation, and activation of myeloid cells. Biochem Biophys Res Commun 353(1):133–138 Coffelt SB, Hughes R, Lewis CE (2009) Tumor-associated macrophages: effectors of angiogenesis and tumor progression. Biochim Biophys Acta 1796(1):11–18 Coussens LM, Werb Z (2002) Inflammation and cancer. Nature 420(6917):860–867 Domachowske JB, Bonville CA, Gao JL, Murphy PM, Easton AJ, Rosenberg HF (2000) The chemokine macrophage-inflammatory protein-1 alpha and its receptor CCR1 control pulmonary inflammation and antiviral host defense in paramyxovirus infection. J Immunol 165(5):2677–2682 Dudziak D, Kamphorst AO, Heidkamp GF, Buchholz VR, Trumpfheller C, Yamazaki S, Cheong C, Liu K, Lee HW, Park CG, Steinman RM, Nussenzweig MC (2007) Differential antigen processing by dendritic cell subsets in vivo. Science (New York, NY) 315(5808): 107–111 Fredriksson R, Lagerstrom MC, Lundin LG, Schioth HB (2003) The G-protein-coupled receptors in the human genome form five main families. Phylogenetic analysis, paralogon groups, and fingerprints. Mol Pharmacol 63(6):1256–1272 Gabrilovich DI, Nagaraj S (2009) Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 9(3):162–174 Galle J, Sittig D, Hanisch I, Wobus M, Wandel E, Loeffler M, Aust G (2006) Individual cell-based models of tumor-environment interactions: Multiple effects of CD97 on tumor invasion. Am J Pathol 169(5):1802–1811 Geissmann F, Manz MG, Jung S, Sieweke MH, Merad M, Ley K (2010) Development of monocytes, macrophages, and dendritic cells. Science (New York, NY) 327(5966):656–661 Gordon S, Martinez FO (2010) Alternative activation of macrophages: mechanism and functions. Immunity 32(5):593–604 Gordon S, Taylor PR (2005) Monocyte and macrophage heterogeneity. Nat Rev Immunol 5(12): 953–964 Gray JX, Haino M, Roth MJ, Maguire JE, Jensen PN, Yarme A, Stetler-Stevenson MA, Siebenlist U, Kelly K (1996) CD97 is a processed, seven-transmembrane, heterodimeric receptor associated with inflammation. J Immunol 157(12):5438–5447 Grivennikov SI, Greten FR, Karin M (2010) Immunity, inflammation, and cancer. Cell 140(6):883–899 Hamann J, Eichler W, Hamann D, Kerstens HM, Poddighe PJ, Hoovers JM, Hartmann E, Strauss M, van Lier RA (1995) Expression cloning and chromosomal mapping of the leukocyte activation antigen CD97, a new seven-span transmembrane molecule of the secretion receptor superfamily with an unusual extracellular domain. J Immunol 155(4):1942–1950 Hamann J, Vogel B, van Schijndel GM, van Lier R (1996) The seven-span transmembrane receptor CD97 has a cellular ligand (CD55, DAF). J Exp Med 184(3):1185–1189 Hamann J, Stortelers C, Kiss-Toth E, Vogel B, Eichler W, van Lier RA (1998) Characterization of the CD55 (DAF)-binding site on the seven-span transmembrane receptor CD97. Eur J Immunol 28(5):1701–1707 Hanahan D (1989) Transgenic mice as probes into complex systems. Science (New York, NY) 246(4935):1265–1275 Jaspars LH, Vos W, Aust G, Van Lier RA, Hamann J (2001) Tissue distribution of the human CD97 EGF-TM7 receptor. Tissue Antigens 57(4):325–331 Joyce JA, Pollard JW (2009) Microenvironmental regulation of metastasis. Nat Rev Cancer 9(4):239–252 Kop EN, Adriaansen J, Smeets TJ, Vervoordeldonk MJ, van Lier RA, Hamann J, Tak PP (2006) CD97 neutralisation increases resistance to collagen-induced arthritis in mice. Arthritis Res Ther 8(5):R155
1
Macrophage Phenotype in Tumours
15
Krasnoperov V, Bittner MA, Holz RW, Chepurny O, Petrenko AG (1999) Structural requirements for alpha-latrotoxin binding and alpha-latrotoxin-stimulated secretion. A study with calciumindependent receptor of alpha-latrotoxin (CIRL) deletion mutants. J Biol Chem 274(6): 3590–3596 Kwakkenbos MJ, Chang GW, Lin HH, Pouwels W, de Jong EC, van Lier RA, Gordon S, Hamann J (2002) The human EGF-TM7 family member EMR2 is a heterodimeric receptor expressed on myeloid cells. J Leukoc Biol 71(5):854–862 Kwakkenbos MJ, Kop EN, Stacey M, Matmati M, Gordon S, Lin HH, Hamann J (2004) The EGF-TM7 family: a postgenomic view. Immunogenetics 55(10):655–666 Kwakkenbos MJ, Pouwels W, Matmati M, Stacey M, Lin HH, Gordon S, van Lier RA, Hamann J (2005) Expression of the largest CD97 and EMR2 isoforms on leukocytes facilitates a specific interaction with chondroitin sulfate on B cells. J Leukoc Biol 77(1):112–119 Kwakkenbos MJ, Matmati M, Madsen O, Pouwels W, Wang Y, Bontrop RE, Heidt PJ, Hoek RM, Hamann J (2006) An unusual mode of concerted evolution of the EGF-TM7 receptor chimera EMR2. FASEB J 20(14):2582–2584 Leemans JC, te Velde AA, Florquin S, Bennink RJ, de Bruin K, van Lier RA, van der Poll T, Hamann J (2004) The epidermal growth factor-seven transmembrane (EGF-TM7) receptor CD97 is required for neutrophil migration and host defense. J Immunol 172(2):1125–1131 Lin HH, Stubbs LJ, Mucenski ML (1997) Identification and characterization of a seven transmembrane hormone receptor using differential display. Genomics 41(3):301–308 Lin HH, Stacey M, Hamann J, Gordon S, McKnight AJ (2000) Human EMR2, a novel EGF-TM7 molecule on chromosome 19p13.1, is closely related to CD97. Genomics 67(2):188–200 Lin HH, Stacey M, Saxby C, Knott V, Chaudhry Y, Evans D, Gordon S, McKnight AJ, Handford P, Lea S (2001) Molecular analysis of the epidermal growth factor-like short consensus repeat domain-mediated protein-protein interactions: dissection of the CD97–CD55 complex. J Biol Chem 276(26):24160–24169 Lin HH, Chang GW, Davies JQ, Stacey M, Harris J, Gordon S (2004) Autocatalytic cleavage of the EMR2 receptor occurs at a conserved G protein-coupled receptor proteolytic site motif. J Biol Chem 279(30):31823–31832 Lin HH, Faunce DE, Stacey M, Terajewicz A, Nakamura T, Zhang-Hoover J, Kerley M, Mucenski ML, Gordon S, Stein-Streilein J (2005) The macrophage F4/80 receptor is required for the induction of antigen-specific efferent regulatory T cells in peripheral tolerance. J Exp Med 201(10):1615–1625 Mantovani A, Sozzani S, Locati M, Allavena P, Sica A (2002) Macrophage polarization: tumorassociated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol 23(11):549–555 Mantovani A, Allavena P, Sica A, Balkwill F (2008) Cancer-related inflammation. Nature 454(7203):436–444 Martinez FO, Helming L, Gordon S (2009) Alternative activation of macrophages: an immunologic functional perspective. Annu Rev Immunol 27:451–483 McKnight AJ, Gordon S (1996) EGF-TM7: a novel subfamily of seven- transmembrane-region leukocyte cell-surface molecules. Immunol Today 17(6):283–287 McKnight AJ, Gordon S (1998) The EGF-TM7 family: unusual structures at the leukocyte surface. J Leukoc Biol 63(3):271–280 McKnight AJ, Macfarlane AJ, Dri P, Turley L, Willis AC, Gordon S (1996) Molecular cloning of F4/80, a murine macrophage-restricted cell surface glycoprotein with homology to the G-protein-linked transmembrane 7 hormone receptor family. J Biol Chem 271(1):486–489 Merad M (2010) PU.1 takes control of the dendritic cell lineage. Immunity 32(5):583–585 Merad M, Manz MG (2009) Dendritic cell homeostasis. Blood 113(15):3418–3427 Peranzoni E, Zilio S, Marigo I, Dolcetti L, Zanovello P, Mandruzzato S, Bronte V (2010) Myeloid-derived suppressor cell heterogeneity and subset definition. Curr Opin Immunol 22(2):238–244 Qian BZ, Pollard JW (2010) Macrophage diversity enhances tumor progression and metastasis. Cell 141(1):39–51
16
H.-H. Lin and S. Gordon
Rabinovich GA, Gabrilovich D, Sotomayor EM (2007) Immunosuppressive strategies that are mediated by tumor cells. Annu Rev Immunol 25:267–296 Rosas M, Gordon S, Taylor PR (2007) Characterisation of the expression and function of the GM-CSF receptor alpha-chain in mice. Eur J Immunol 37(9):2518–2528 Rosas M, Thomas B, Stacey M, Gordon S, Taylor PR (2010) The myeloid 7/4-antigen defines recently generated inflammatory macrophages and is synonymous with Ly-6B. J Leukoc Biol 88(1):169–180 Sinha P, Clements VK, Ostrand-Rosenberg S (2005) Reduction of myeloid-derived suppressor cells and induction of M1 macrophages facilitate the rejection of established metastatic disease. J Immunol 174(2):636–645 Stacey M, Lin HH, Gordon S, McKnight AJ (2000) LNB-TM7, a group of seven-transmembrane proteins related to family-B G-protein-coupled receptors. Trends Biochem Sci 25(6):284–289 Stacey M, Chang GW, Davies JQ, Kwakkenbos MJ, Sanderson RD, Hamann J, Gordon S, Lin HH (2003) The epidermal growth factor-like domains of the human EMR2 receptor mediate cell attachment through chondroitin sulfate glycosaminoglycans. Blood 102(8):2916–2924 Steinert M, Wobus M, Boltze C, Schutz A, Wahlbuhl M, Hamann J, Aust G (2002) Expression and regulation of CD97 in colorectal carcinoma cell lines and tumor tissues. Am J Pathol 161(5):1657–1667 Taylor PR, Martinez-Pomares L, Stacey M, Lin HH, Brown GD, Gordon S (2005) Macrophage receptors and immune recognition. Annu Rev Immunol 23:901–944 Umemura N, Saio M, Suwa T, Kitoh Y, Bai J, Nonaka K, Ouyang GF, Okada M, Balazs M, Adany R, Shibata T, Takami T (2008) Tumor-infiltrating myeloid-derived suppressor cells are pleiotropic-inflamed monocytes/ macrophages that bear M1- and M2-type characteristics. J Leukoc Biol 83(5):1136–1144 van den Berg TK, Kraal G (2005) A function for the macrophage F4/80 molecule in tolerance induction. Trends Immunol 26(10):506-509 van Eijk M, Aust G, Brouwer MS, van Meurs M, Voerman JS, Dijke IE, Pouwels W, Sandig I, Wandel E, Aerts JM, Boot RG, Laman JD, Hamann J (2010) Differential expression of the EGF-TM7 family members CD97 and EMR2 in lipid-laden macrophages in atherosclerosis, multiple sclerosis and Gaucher disease. Immunol Lett 129(2):64–71 van Pel M, Hagoort H, Hamann J, Fibbe WE (2008a) CD97 is differentially expressed on murine hematopoietic stem-and progenitor-cells. Haematologica 93(8):1137–1144 van Pel M, Hagoort H, Kwakkenbos MJ, Hamann J, Fibbe WE (2008b) Differential role of CD97 in interleukin-8-induced and granulocyte-colony stimulating factor-induced hematopoietic stem and progenitor cell mobilization. Haematologica 93(4):601–604 Varol C, Yona S, Jung S (2009) Origins and tissue-context-dependent fates of blood monocytes. Immunol Cell Biol 87(1):30–38 Wang T, Ward Y, Tian L, Lake R, Guedez L, Stetler-Stevenson WG, Kelly K (2005) CD97, an adhesion receptor on inflammatory cells, stimulates angiogenesis through binding integrin counterreceptors on endothelial cells. Blood 105(7):2836–2844 Wang T, Tian L, Haino M, Gao JL, Lake R, Ward Y, Wang H, Siebenlist U, Murphy PM, Kelly K (2007) Improved antibacterial host defense and altered peripheral granulocyte homeostasis in mice lacking the adhesion class G protein receptor CD97. Infect Immun 75(3):1144–1153 Warschkau H, Kiderlen AF (1999) A monoclonal antibody directed against the murine macrophage surface molecule F4/80 modulates natural immune response to Listeria monocytogenes. J Immunol 163(6):3409–3416 Wobus M, Huber O, Hamann J, Aust G (2006) CD97 overexpression in tumor cells at the invasion front in colorectal cancer (CC) is independently regulated of the canonical Wnt pathway. Mol Carcinog 45(11):881–886 Yona S, Lin HH, Siu WO, Gordon S, Stacey M (2008a) Adhesion-GPCRs: emerging roles for novel receptors. Trends Biochem Sci 33(10):491–500 Yona S, Lin HH, Dri P, Davies JQ, Hayhoe RP, Lewis SM, Heinsbroek SE, Brown KA, Perretti M, Hamann J, Treacher DF, Gordon S, Stacey M (2008b) Ligation of the adhesion-GPCR EMR2 regulates human neutrophil function. FASEB J 22(3):741–751
Chapter 2
Role of Tumour-Associated Macrophages in the Regulation of Angiogenesis Russell Hughes, Hsin-Yu Fang, Munitta Muthana, and Claire E. Lewis
Introduction As mentioned in previous chapters, tumours consist not only of malignant cells but also of various stromal cell types including tumour-associated macrophages (TAMs) (Sica et al. 2008). One early sign that TAMs might influence tumour angiogenesis was the finding that TAM numbers positively correlate with tumour angiogenesis in breast carcinomas (Leek et al. 1996). Several subsequent studies have confirmed such a link in a wide array of tumour types (Aharinejad et al. 2004; Bailey et al. 2007; Koide et al. 2004; Ohta et al. 2003; Saji et al. 2001) and showed that high-TAM numbers are also often linked to poor prognosis (Bingle et al. 2002; Lewis and Pollard 2006). However, definitive evidence for the pro-angiogenic effect of TAMs in tumours was provided using various murine tumour models. First, we demonstrated that macrophage infiltration into small breast tumour nodules (tumour spheroids) markedly enhanced their ability to induce neovascularisation following implantation into window chambers on the flanks of mice (Bingle et al. 2006). Then Lin and colleagues showed that, when MMTV-PyMT mice (which develop mammary tumours) were crossed with transgenic mice carrying a colony stimulating factor-1 (Csf1) null mutation (Csf1op/op), the absence of CSF-1 markedly decreased macrophage infiltration in pre-malignant tumours, which, in turn, resulted in the inhibition of tumour angiogenesis and delayed tumour progression (Lin et al. 2006; Lin et al. 2001). Indeed, they showed that TAMs control the ‘angiogenic switch’ associated with the malignant transition of the spontaneous mammary tumours that form in MMTV-PyMT mice (Lin et al. 2006). Other strategies employing an antibody to CSF-1 (Paulus et al. 2006) or clodronate liposomes (Zeisberger et al. 2006) to deplete TAMs have also resulted in a marked reduction in tumour angiogenesis.
2 (UGHES s ( 9 &ANG