Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2007 Jul 30.
Published in final edited form as: Free Radic Biol Med. 2006 Sep 16;41(11):1621–1628. doi: 10.1016/j.freeradbiomed.2006.08.026

Tumor Macrophage Redox and Effector Mechanisms Associated with Hypoxia

Michael Graham Espey 1
PMCID: PMC1934898  NIHMSID: NIHMS13701  PMID: 17145549

Abstract

Monocytes are recruited from the circulation into solid tumors where they differentiate into macrophages with unique phenotypes. While macrophages utilize oxygen in a broad range of immune effector functions, the generation of reactive oxygen and nitrogen oxide species is less clear in the setting of hypoxia, which can be a prominent feature of solid tumors. The relationships between innate immunity, redox systems and the plasticity of phenotypic changes tumor-associated macrophages undergo in conjunction with tumor hypoxia will be examined.

Keywords: Tumor associated macrophage, Innate, iNOS, Hypoxia, Nitric oxide, Oxygen, NOX-2, antimicrobial peptide, M2

Introduction

A hallmark of most solid tumors is the accumulation of macrophages, which are often the most abundant of infiltrating leukocytes [1-5]. Tumor-associated macrophages (TAM) are bone marrow-derived leukocytes solicited and directed by cancer cells to adopt unique phenotypes that can facilitate tumor growth and survival. While the formation of innate redox effectors is thought to contribute to genetic instability and carcinogenic transformation of pre-neoplasia, less well characterized is the role redox species play in the maintenance and progression of tumors once they are established. Because oxygen is inherently tied to the generation of superoxide (O2-), hydrogen peroxide (H2O2) and nitrogen oxide species, the participation of redox effectors in cancer is further complicated by the situation of hypoxia that develops regionally in most types of solid tumors. In this review, we will highlight the role oxygen tension can play in the redox biology and heterogeneity of TAM.

Heterogeneity of TAM immuno-phenotypes

Immunologists often use the so-called M1 to M2 classification scheme to phenotypically subdivide macrophages along a spectrum with cells displaying distinct patterns of function in association with lymphocytic T-helper1 versus T-helper2 driven responses. The TAM phenotype has been described as skewed along the M2 axis [6-11]. Prototypic M2-polarizing agents are interleukin (IL)-4, IL-10, IL-13, TGF-β1, steroids and prostaglandin-E2; each agent capable of rendering distinct phenotypic changes. M2-oriented macrophages generally express high levels of galactose, mannose, and scavenger-type A receptors and have IL-12low, IL-10high or IL4/13high cytokine phenotypes. In addition to solid tumors, macrophage responses skewed toward M2 are associated with certain parasitic infections, asthma and wound healing.

In contrast to M2, macrophages exposed to microbial products and interferon-γ can be activated toward the M1 phenotype. M1-biased macrophages have prototypic IL-12high, IL-10low or IL4/13low cytokine phenotypes and are enriched in their responses to opsonized ligands and toll-like receptor engagement. M1 macrophage populations are generally recognized for an enhanced capacity to form reactive oxygen and nitrogen oxide species, which they utilize for bactericidal and tumoricidal activities.

A third type of TAM known as myeloid suppressor cells (MSC) can be defined by the Gr-1high,CD11bhigh, F4/80low pattern of cell surface receptors and are represented by a heterogeneous myeloid progenitor population of macrophages, granulocytes and dendritic cells originating in bone marrow [12]. MSC in the peripheral lymphoid organs facilitate tolerance in adaptive immune responses. MSC are often enriched in the lymph nodes and spleen of cancer patients where they are effective suppressors of tumoricidal CD4+ and CD8+ T lymphocytes [12-15]. While the majority of TAM are Gr-1low, F4/80high, MSC can also infiltrate tumors comprising approximately 5% of total cells [15]. One report suggests that MSC in particular may have the capacity to acquire endothelial cell properties at perivascular sites [15]. Hypoxia can promote selective myeloid infiltration to these positions [16]. MSC are emblematic of the problem in distinctly ascribing M1/M2 classifications for TAM as they employ generation of reactive oxygen and nitrogen species, a feature of M1, and foster immunosuppression, a feature of M2.

It should be stressed that the M1/M2 axis is a conceptual framework to assist in categorizing macrophage mediation of immune responses. An overwhelming percentage of either M1- or M2-biased TAM within a tumor is usually not evident unless a model system specifically devised to drive either phenotype is used. In vitro studies have shown that the sequence in which polarized macrophages are exposed to cytokines can redistribute them into a variety of functional phenotypes [17, 18]. These studies suggested that macrophage M1/M2 polarization was not static; rather a pliable spectrum of responsiveness exists that permits macrophage adaptation to multiple microenvironment cues. Distinct differences in the transcriptome of MSC derived from peripheral sites versus TAM isolated from tumor were found following IL-4 stimulation [19]. This finding reinforces the viewpoint that tissue environment clearly influences the functional phenotype of TAM subpopulations. In addition to the cytokine milieu, oxygenation status can also serve a significant determinant cue in directing macrophage phenotypes [20]. This makes evolutionary sense when considering the advantage such flexibility provides in macrophage acute immune responses and subsequent resolution of inflammation where changes in oxygen tension play a prominent role; for instance, our response to both infectious facultative anaerobes and wounds, which occurs on a regular basis relative to cancer. In contrast to acute inflammation, resolution in cancer often is not forthcoming without aggressive therapeutic intervention. The combination of longevity of response, tumor cell signaling and changes associated with oxygen tension divert inflammatory macrophage immunity toward the unique characteristics of TAM.

Solid malignancies on the micro-scale also undergo constant change, with outgrowth of subclonal tumor cell populations, parenchymal remodeling and neovascularization in a recapitulation of morphogenesis. Hypoxia is a salient feature of solid tumors and the levels of tissue oxygenation have a profound influence on the biology of TAM [2, 4, 5, 9-11, 19, 20-26]. The need for the tumor to maintain adequate nutrient and oxygenation status can drive invasive growth, angiogenesis, metastatic intravasation and development of necrotic pockets. The aggregate effect of the continuous cycle of change on TAM is to become usurped by the tumor into numerous supporting rather than tumoricidal roles. TAM differentiation is determined by proximal cues from tumor cells, which vary dependent upon their location within the tumor mass. TAM phenotype can be quite heterogeneous and specialized for a particular micro-region of the tumor. Oxygen tension can be a powerful determinant in the paracrine relationships between TAM and tumor cells. Despite the opinion that the M2-type bias of TAM often denudes the involvement of redox effector species, we will review findings that suggest redox regulation for TAM exists at numerous levels in tumor biology and merits a second look.

Oxygen and TAM localization

TAM are thought to extravasate from the blood circulating pool of monocytes [20, 28 ]in response to a variety of chemokine and cytokine substances. TAM accumulation often occurs in margination zones that surround islands of burgeoning tumor cells and in association with the tumor vasculature [1, 11, 18, 19, 28-31]. TAM can also be prominent in hypoxic regions proximal to sites of necrosis and poor vascularization. The steps that determine TAM differentiation into distinct local subpopulations within tumors and the degree to which the nature of tumor hypoxia influences TAM position cueing remain key issues to resolve.

The presence of blood vessels does not necessarily ensure that a region of tumor is receiving appropriate perfusion and oxygenation. Likewise, hypoxia is not an absolute condition [28-33]. Tumor blood flow may be transiently compromised in regions where microvessels are immature, occluded or supplying deoxygenated blood [24, 68, 69]. Over time, TAM subpopulations near vessels may be exposed to sporadic cycles of hypoxia. In contrast to intermittent hypoxia, chronic tumor hypoxia develops in areas where cells are beyond the diffusion distance of oxygen from microvessels; importantly, a gradient for oxygen along the radial distance from the vessel(s) exists. Therefore, TAM in chronically hypoxic regions can experience a range of abnormally low oxygen tensions dependent on their position relative to microvessels.

The dynamic nature of oxygen in tumors can influence TAM trafficking through several redox-related mechanisms. Hypoxic regulation of TAM motility is consistent with macrophage function as an innate responder to areas of necrosis during infection, wound damage and repair. Although amazing improvements have been made in techniques to visualize TAM traffic in situ [34], questions on the influence of oxygen tension remain open. Two-photon microscopy studies show that tumor cells and TAM utilize type-1 collagen fibers for movement within tumors [34]. The degree of TAM mobilization into and egress from chronically hypoxic compartments is currently unclear.

Emphasis to date has been on TAM recruitment into tumors. In vitro studies have shown that hypoxia can cause TAM migratory arrest. Hypoxic conditions (1% O2) decreased murine macrophage mRNA expression and protein secretion of monocyte chemoattractant protein-1 (CCL-2) and CCR5, the receptor for macrophage inflammatory protein-1α and monocyte chemoattractant protein-2 [35, 36]. Studies with human peripheral blood-derived macrophages and THP-1 cells suggested that TAM chemoreceptor signaling (e.g., CCR2) through p42/p44 mitogen activated protein kinases (MAPK) can be rapidly attenuated by hypoxia-induced activation of MAPK phosphatase-1 [37]. These experiments suggest that hypoxia diminishes facets of paracrine signaling amongst TAM and between TAM and tumor cells resulting in weakened chemokine responsiveness.

Two-way trafficking, however, implies a need for both stop and start signals under tumor hypoxia. Matrilysin matrix metalloproteinase (MMP-7) is illustrative of how differential redox regulation may govern aspects of itinerant TAM that may enable transit both into and out of hypoxic regions. Hypoxia strongly induces MMP-7 expression in TAM [21], however, the presence of oxygen is necessary for pro-enzyme processing and activity via cysteinyl oxygenation in the enzyme pro-domain [38]. In this manner, TAM movement from hypoxic regions via MMP-7 may be forwardly directed along gradients of increasing oxygen concentration. Interestingly, the basement membrane degrading activity of mature MMP-7 becomes inhibited through cross-linking of a tryptophanyl residue in markedly oxidative environments [39]. Hypoxic induction, oxygen dependent processing and oxidative stress inhibition for MMP-7 taken together show a specialized adaptation to changing redox microenvironments. Selective impairment of chemoattractant production by hypoxic conditions [35-37] may create a gradient well for these agents that strengthens chemotaxis-driven emigration of cancer cells and allied TAM chaperones adapted to move out of chronically hypoxic regions. Initiation of metastatic disease therefore may be inherently tied to oxygen tension and redox regulation of cell motility.

Evolution, oxygen and innate immunity

It is somehow fitting that the first description of macrophages and the concept of innate immunity was born from experimental observations by Metchnikoff of a relatively simple invertebrate, a starfish larva, which he had irritated with a rose thorn [40, 41]. The origins of macrophages in irradicating pathogenic invaders are indeed primordial, stretching backwards to our earliest metazoan ancestors. The usage of oxygen as a substrate for specialized immune functions through NADPH oxidase (NOX-2) and nitric oxide synthase (NOS-2 or i nducible NOS) occurred relatively late in evolution. The pattern of molecular and phylogenetic changes in both enzymes suggests that they were the products of gene duplication, perhaps developing in chordates with the separation of mammals and fishes [42-46].

Features of TAM biology under low oxygen tensions may be rooted in our innate immune responses to facultative anaerobes. With the emergence of NOX-2 and iNOS enzymes, a new dimension was added to the more ancient antimicrobial peptide defense network. Secreted chiefly by phagocytes and epithelial cells, defensins and cathelicidins are small peptides typically organized into highly cationic and hydrophobic domains that directly promote membrane lysis when in contact with bacteria [47-49]. Members of this diverse family participate in macrophage chemoattraction [50] and promote angiogenesis [51, 52]. Antimicrobial peptides can act as opsonizing agents to enhance superoxide formation by NOX-2 [53]. Pro-α-defensin is a substrate for MMP-7 [54], which is activated by low concentrations of myeloperoxidase-derived HOCl [38]. The proline-arginine-rich antimicrobial PR-39 was found to directly inhibit NOX-2 through direct binding of p47phox subunit [55], while serving as an inducer of iNOS expression and activity [56].

Taken together, these observations highlight a complex level of cross regulation between different members of our innate effector systems that are predisposed to act under varied oxygen tensions during infection. Knockout studies showed that expression of antimicrobial peptides in macrophages was coordinated by the action of hypoxia-inducible factor-1 (HIF-1) [57]. Therefore, HIF-1 represents an important link between our inherent innate response strategy to infectious agents and a key transcription factor involved in both TAM and tumor cell biology [58]. The contribution of antimicrobial peptides in tumor biology and TAM phenotypes may be most relevant in tumors of epithelial origin where production of these agents by malignant cells and TAM could be activated by dysregulation of mucosal immunity processes, necrosis and hypoxia.

Hypoxic iNOS and NOX-2

The functional affects of TAM catalyzing the generation of reactive species (e.g., O2-, H2O2, NO) derived from oxygen will be strongly influenced by local tumor oxygen gradients. The macrophage iNOS Michaelis-Menten constant (Km) for oxygen has been studied in detail [59, 60]. Kinetic models and experimental data show that the Km for oxygen shifts from 2.5 μM in the absence of arginine to 7.5 μM (or 0.85%) in the presence of arginine due to NO intrinsically (near-geminate NO) binding within the iNOS heme pocket as the enzyme is in the ferric state. Subsequent to heme reduction to the ferrous form, the iNOS apparent Km for oxygen increases (25 to 130 μM) as a function of NO concentration in the surrounding milieu, which can enter the enzyme to form ferrousnitrosyl complexes that rapidly consume oxygen in a futile cycle to produce alternate products (e.g., nitrate, NO3-) in lieu of NO. From a kinetic viewpoint, NO formation from iNOS at relatively low oxygen tensions would be compromised by scant oxygen availability for arginine (and N-hydroxyarginine) oxidation. On the other hand, decreased NO output would render iNOS less prone to NO feedback inhibition lowering the apparent Km for oxygen. The net effect for hypoxic TAM would be an iNOS equilibrium optimized for a minimal oxygen Km and maximal productive NO biosynthesis. Further complicating this scheme is the possibility for the formation of either O2- or H2O2 from iNOS via uncoupled electron flow [61]; however, this alternate enzymology may be related primarily to the neuronal and endothelial NOS isoforms [62, 63].

In addition to genes involved in glycolysis and angiogenesis [58], HIF has an association with iNOS in TAM. Murine B16 melanoma tumors engineered to constitutively express GM-CSF resulted a large TAM infiltrate that showed high immunoreactivity for both HIF-1 and iNOS [64]. However, clinical studies suggest that TAM expressing iNOS and/or HIF in human tumors represent more select subpopulations of TAM that vary widely depending on the tumor type [e.g., 65-68,]. Interestingly, human tumors with a relatively lower numbers of TAM were observed to have a higher proportion of TAM with HIF-1 immunoreactivity [68].

These studies suggest that the relationship between HIF-1 and iNOS may serve a variety of specialized functions in TAM depending on their tissue location and nadir of hypoxia. HIF-1 can drive the expression of iNOS [69, 70] and NO product can differentially regulate HIF protein stability dependent upon oxygen tension, redox tone and the absolute level of NO exposure [71-74]. This delicate system of feedback regulation operates at several levels. Moderate steady state levels of NO under aerobic conditions were shown to stabilize HIF protein [71, 74]. In contrast, HIF degradation is prompted by NO exposure in cells under hypoxia [72, 73]. One hypothesis proposes that by virtue of NO inhibition of mitochondrial respiration, micro gradients of increased oxygen availability are created, which in turn is permissive for prolyl hydroxylase mediated destabilization of HIF [72]. In the context of TAM, glucose metabolism under hypoxia is shifted from oxidative phosphorylation to glycolysis [20]. Reactive oxygen species produced during intermittent hypoxia and/or within the penumbra of chronically hypoxic regions of tumor could also shift the balance of nitrosylative chemistry in such cells toward an more oxidative character that results in HIF destabilization [73, 74]. The aggregate effect of this system is one of feedforward where either NO or other agents promote HIF stabilization, which mediates transactivation of iNOS gene expression and NO generation to strengthen HIF stabilization. Negative feedback regulation under hypoxia can occur from by NO chemistry scaled to the concentration of oxygen and presence of reactive oxygen species within the immediate microenvironment resulting in HIF destabilization.

In contrast to iNOS, NOX-2 in TAM biology and hypoxia is relatively unexplored. Studies on NOX-2 and hypoxia have focused on this isozyme in the context of neutrophils and innate host defense against infectious agents. The NOX-2 Michaelis-Menten constant (Km) value for oxygen in neutrophils was estimated to be approximately 5 to 10 μM, with an affinity increase of 2 to 3-fold following activation [75, 76]. Therefore, formation of reactive oxygen species via NOX-2 in activated TAM would be plausible even at very low oxygen tensions; however, net output would still be low dependent on the absolute availability of oxygen. This raises the issue that reactive oxygen species generated at both high concentrations and sufficient duration are generally required to elicit cytotoxic effects, while low levels may contribute pro-growth signals to tumor cells. In this manner, hypoxia could convert TAM reactive oxygen-based cytolytic mechanisms into one that fosters tumor cell survival. Always inherently tied into this dynamic is the relative concentration of NO, which serves to modulate the balance in O2- and H2O2 actions toward either cytotoxic or pro-growth [71, 73, 74, 77].

Interestingly, MSC isolated from tumor bearing mice were found to have a tonic elevation in H2O2>production relative to those obtained from naïve and immunized mice that was insensitive to superoxide dismutase treatment [78]. In addition to peripheral lymphoid tissues, the potential generation of H2O2>by MSC within tumors may suppress cytotoxic effector function of tumor infiltrating lymphocytes. TAM extrinsic sources of reactive oxygen species derived from tumor and endothelial cells certainly contribute to the overall redox microenvironment. Indeed, other NOX isoforms can be overexpressed in many types of cancer cells [79]. While formation of reactive oxygen species plays a major role in cytotoxicity associated with immunosurveillance and carcinogenesis, generation of these agents in established tumors is weighted toward immunosuppression and growth progression affects.

Significant formation of reactive oxygen species in tumors would be most involved in zones of intermittent hypoxia and reperfusion. The anionic character of O2- limits it diffusion through membranes; therefore, H2O2 may be considered as the traveler among reactive oxygen species thereby affecting areas more distal to its point of formation through peroxidase-driven chemistry. In the presence of peroxidases or metal catalysts, H2O2 in combination with NO autoxidation product nitrite (NO2-) generates another relatively motile species NO2 [80], which is the chief agent responsible for formation of nitrated moieties such as 3-nitrotyrosyl residues on proteins [81]. An exciting area of investigation is the reductive conversion of nitrite (NO2-) to NO under conditions of tumor hypoxia, which would relieve the need for O2 consumption to activate soluble guanylyl cyclase (unpublished observations).

Wound repair, hypoxic tumors and arginine metabolism

In step with the view that aspects of tumors are similar to wounds that do not heal [82], insight from the relationship between oxygen and macrophage arginine metabolism during wound repair can be gained [83]. Rat macrophages derived from hypoxic wounds displayed marked shift in arginine catalysis from iNOS to that mediated via the polyamine enzyme arginase-1 at oxygen tensions of less than 5% (45 μM) [84]. Upon reoxygenation of rat wound derived macrophages, the absolute level of arginine catabolism was augmented due to increased expression of both arginase-1 and iNOS. At the substrate level, arginase-1 may loose out in competition with iNOS due to a relatively higher Km for arginine and the potent competitive inhibition of arginase-1 by N-hydroxyarginine produced during iNOS catalysis [84, 85]. Likewise, arginase activity is independent of oxygen tension.

These data suggest that hypoxia can elicit induction of both arms of the arginine axis (oxidation and hydrolysis) in macrophages with catabolic fate toward either NO or ornithine biosynthesis dictated by oxygen tension. In the rat wound model, temporal segregation of arginine metabolism was observed consistent with an inflammation-wound repair sequence [83]. Both HIF-1 induction and iNOS activity were evident primarily in the initial 48-72 hours of wound formation, which preceded development of wound hypoxia and arginase activity occurring only after 4 to 5 days of injury in this model [86].

Notable differences between wound macrophages and TAM with respect to arginine metabolism exist. The prototypic M1 interferon-γ activated transcription factor STAT-1 was found to be crucial for expression of both iNOS and arginase-1 genes in freshly isolated MSC from murine tumor, but not spleen of afflicted mice [87]. This finding again highlights that the M1/M2 scheme cannot be applied broadly to selective TAM subpopulations [19, 87]. This and other studies amply demonstrate that arginine catabolism and phenotypic behavior of TAM can be varied and change dependent on their tissue of origin (e.g., blood, spleen, tumor) and length of time cultured ex vivo; debate continues on interspecies differences in macrophage arginine catabolism [83-92] showing the circumstance of TAM arginine catabolism in human tumors and its relationship to hypoxia to be an important subject of investigation.

Summary

Hypoxia, intermittent hypoxia and associated reperfusion oxidative stress differentially influences TAM behavior. It is currently not known whether TAM diversity is generated as a result of distinct defined lineages, tumor-directed prompting or weighted by physical factors such as oxygen tension. In vitro studies show that the sequence in which macrophages are exposed to cytokines can sway them to different functional phenotypes, suggesting that the M1/M2 polarization is not static, rather it is pliable dependent on multiple microenvironment cues [17, 18]. The context of oxygen tension within the tumor cytokine/chemokine milieu is an important consideration for studies ascribing mechanism to TAM phenotypic responses.

Innate immunity between mammalian species can be similar, yet distinct in specific ways. The repertoire of antimicrobial peptides in rats and mice is quite different from that in humans [47-49]. A comparison of pulmonary granulomas induced by infection with M. tuberculosis in mice and humans found select differences in the spatial organization of macrophages to other leukocytes as well as marked differences in relative oxygen tension [93]. Interspecies comparisons of TAM and their adaptations to hypoxia involving redox mechanisms should be evaluated with caution.

The redox behavior of TAM is a key area of study from the perspective of understanding how to improve treatment strategies and alleviate suffering of patients with solid tumors. The predilection for tumor recruitment and differentiation of peripheral blood monocytes to hypoxic TAM opens the door for Trojan horse tactics that employs manipulation of TAM gene expression and metabolism to enhance tumoricidal effects of current treatment modalities [4, 21, 22, 68]. Itinerant TAM can improve disbursement of therapeutic cargo in areas that are inaccessible to delivery of chemotherapeutics through the vasculature or reticent to radiation treatment due to hypoxia. Future developments in the use of cancer patient autologous monocytes for TAM-based therapy will require an increased appreciation for redox regulation of their trafficking and effector functions.

Acknowledgments

We kindly thank Drs. Leto (NIAID, NIH) and Lambeth (Emory) for helpful discussions, Dr. Stuehr (Cleveland Clinic) for providing the manuscript music, Dr. Mitchell (NCI, NIH) for helpful discussions and Dr. Wink (NCI, NIH) for his continued support. This research was supported by the Intramural Research Program of the NIH, National Cancer Institute, Center for Cancer Research.

Abbreviations

HIF

hypoxia-inducible transcription factor

iNOS

inducible NOS nitric oxide synthase

IL

interleukin

GM-CSF

granulocyte macrophage colony stimulating factor

MMP

matrix metalloproteinase

MAPK

mitogen activated protein kinase

NOX-2

NADPH oxidase

TAM

tumor-associated macrophages

Biography

Brief Biography

Dr. Espey is a native of Clinton, Iowa and received his B.S. in Medical Technology at the University of Iowa in 1987. He is Board certified in Clinical Pathology, Histocompatibility and Immunogenetics. After working in immunology, organ transplant and infectious disease laboratories at Georgetown Hospital and NIH Clinical Center, he obtained a Ph.D. with distinction for his thesis dissertation on the biochemistry and immunology of tryptophan from Georgetown University in 1995. Dr. Espey’s postdoctoral research on retrovirus-induced neurodegeneration was in the NIDDK at NIH. He is currently a Staff Scientist in the David Wink group at NCI, NIH where his research focus is on the detection and imaging of redox metabolism as it relates to cancer biology. A goal is to understand the biochemistry of reactive nitrogen and oxygen species and their relationship to signals that govern tumor growth and response to interventions such as irradiation.

References

  • [1].Virchow R. Die Krankhaften Geschwulste. Verlag von August Hirschwald; Berlin: 1863. Aetologie der neoplastichen Geschwulste/Pathogenie der neoplastischen Geschwulste; pp. 57–101. [Google Scholar]
  • [2].Kreutz M, Fritsche J, Andreesen R. Macrophages in Tumor Biology. In: Burke B, Lewis CE, editors. The Macrophage. Oxford University Press; 2002. pp. 457–489. [Google Scholar]
  • [3].Wood GW, Gollahon KA. Detection and quantitation of macrophage infiltration into primary human tumors with the use of cell-surface markers. J Natl Cancer Inst. 1977;59:1081–1087. doi: 10.1093/jnci/59.4.1081. [DOI] [PubMed] [Google Scholar]
  • [4].Milas L, Wike J, Hunter N, Volpe J, Basic I. Macrophage content of murine sarcomas and carcinomas: associations with tumor growth parameters and tumor radiocurability. Cancer Res. 1987;47:1069–1075. [PubMed] [Google Scholar]
  • [5].Mantovani A, Bottazzi B, Colotta F, Sozzani S, Ruco L. The origin and function of tumor-associated macrophages. Immunol Today. 1992;13:265–270. doi: 10.1016/0167-5699(92)90008-U. [DOI] [PubMed] [Google Scholar]
  • [6].Goerdt S, Politz O, Schledzewski K, Birk R, Gratchev A, Guillot P, Hakiy N, Klemke CD, Dippel E, Kodelja V, Orfanos CE. Alternative versus classical activation of macrophages. Pathobiology. 1999;67:222–226. doi: 10.1159/000028096. [DOI] [PubMed] [Google Scholar]
  • [7].Elgert KD, Alleva DG;, Mullins DW. Tumor-induced immune dysfunction: the macrophage connection. J Leukoc Biol. 1998;64:275–290. doi: 10.1002/jlb.64.3.275. [DOI] [PubMed] [Google Scholar]
  • [8].Gordon S. Alternative activation of macrophages. Nat Rev Immunol. 2003;3:23–35. doi: 10.1038/nri978. [DOI] [PubMed] [Google Scholar]
  • [9].Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer. 2004;4:71–78. doi: 10.1038/nrc1256. [DOI] [PubMed] [Google Scholar]
  • [10].Sica A, Schioppa T, Mantovani A, Allavena P. Tumour-associated macrophages are a distinct M2 polarised population promoting tumour progression: Potential targets of anti-cancer therapy. Eur J Cancer. 2006;42:717–27. doi: 10.1016/j.ejca.2006.01.003. [DOI] [PubMed] [Google Scholar]
  • [11].Lewis CE, Pollard JW. Distinct role of macrophages in different tumor microenvironments. Cancer Res. 2006;66:605–612. doi: 10.1158/0008-5472.CAN-05-4005. [DOI] [PubMed] [Google Scholar]
  • [12].Serafini P, De Santo C, Marigo I, Cingarlini S, Dolcetti L, Gallina G, Zanovello P, Bronte V. Derangement of immune responses by myeloid suppressor cells. Cancer Immunol Immunother. 2004;53:64–72. doi: 10.1007/s00262-003-0443-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Young MR, Lathers DM. Myeloid progenitor cells mediate immune suppression in patients with head and neck cancers. IntJImmunopharmacol. 1999;21:241–252. doi: 10.1016/s0192-0561(99)00008-9. [DOI] [PubMed] [Google Scholar]
  • [14].Almand B, Clark JI, Nikitina E, van Beynen J, English NR, Knight SC, Carbone DP, Gabrilovich DI. Increased production of immature myeloid cells in cancer patients: a mechanism of immunosuppression in cancer. J Immunol. 2001;166:678–689. doi: 10.4049/jimmunol.166.1.678. [DOI] [PubMed] [Google Scholar]
  • [15].Yang L, DeBusk LM, Fukuda K, Fingleton B, Green-Jarvis B, Shyr Y, Matrisian LM, Carbone DP, Lin PC. Expansion of myeloid immune suppressor Gr+CD11b+ cells in tumor-bearing host directly promotes tumor angiogenesis. Cancer Cell. 2004;6:409–421. doi: 10.1016/j.ccr.2004.08.031. [DOI] [PubMed] [Google Scholar]
  • [16].O’Neill TJ, 4th, Wamhoff BR, Owens GK, Skalak TC. Mobilization of bone marrow-derived cells enhances the angiogenic response to hypoxia without transdifferentiation into endothelial cells. Circ Res. 2005;97:1027–35. doi: 10.1161/01.RES.0000189259.69645.25. [DOI] [PubMed] [Google Scholar]
  • [17].Stout RD, Jiang C, Matta B, Tietzel I, Watkins SK, Suttles J. Macrophages sequentially change their functional phenotype in response to changes in microenvironmental influences. J Immunol. 2005;175:342–9. doi: 10.4049/jimmunol.175.1.342. [DOI] [PubMed] [Google Scholar]
  • [18].Mills CD, Kincaid K, Alt JM, Heilman MJ, Hill AM. M-1/M-2 macrophages and the Th1/Th2 paradigm. J Immunol. 2000;164:6166–6173. doi: 10.4049/jimmunol.1701141. [DOI] [PubMed] [Google Scholar]
  • [19].Biswas SK, Gangi L, Paul S, Schioppa T, Saccani A, Sironi M, Bottazzi B, Doni A, Vincenzo B, Pasqualini F, Vago L, Nebuloni M, Mantovani A, Sica A. A distinct and unique transcriptional program expressed by tumor-associated macrophages (defective NF-B and enhanced IRF-3/STAT1 activation) Blood. 2006;107::2112–2122. doi: 10.1182/blood-2005-01-0428. [DOI] [PubMed] [Google Scholar]
  • [20].Lewis JS, Lee JA, Underwood JC, Harris AL, Lewis CE. Macrophage responses to hypoxia: relevance to disease mechanisms. J Leukoc Biol. 1999;66:889–900. doi: 10.1002/jlb.66.6.889. [DOI] [PubMed] [Google Scholar]
  • [21].Bingle L, Brown NJ, Lewis CE. The role of tumour-associated macrophages in tumour progression: implications for new anticancer therapies. J Pathol. 2002;196:254–265. doi: 10.1002/path.1027. [DOI] [PubMed] [Google Scholar]
  • [22].Burke B, Giannoudis A, Corke KP, Gill D, Wells M, Ziegler-Heitbrock L, Lewis CE. Hypoxia-induced gene expression in human macrophages: implications for ischemic tissues and hypoxia-regulated gene therapy. Am J Pathol. 2003;163:1233–1243. doi: 10.1016/S0002-9440(10)63483-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Coleman CN. Hypoxia in tumors: a paradigm for the approach to biochemical and physiologic heterogeneity. J Natl Cancer Inst. 1988;80:310–317. doi: 10.1093/jnci/80.5.310. [DOI] [PubMed] [Google Scholar]
  • [24].Brown JM, Giaccia AJ. The unique physiology of solid tumors: opportunities (and problems) for cancer therapy. Cancer Res. 1998;58:1408–1416. [PubMed] [Google Scholar]
  • [25].Raleigh JA, Chou SC, Bono EL, Thrall DE, Varia MA. Semiquantitative immunohistochemical analysis for hypoxia in human tumors. Int J Radiat Oncol Biol Phys. 2001;49:569–574. doi: 10.1016/s0360-3016(00)01505-4. [DOI] [PubMed] [Google Scholar]
  • [26].Lanzen J, Braun RD, Klitzman B, Brizel D, Secomb TW, Dewhirst MW. Direct demonstration of instabilities in oxygen concentrations within the extravascular compartment of an experimental tumor. Cancer Res. 2006;66:2219–2223. doi: 10.1158/0008-5472.CAN-03-2958. [DOI] [PubMed] [Google Scholar]
  • [27].Matsumoto K, Subramanian S, Devasahayam N, Aravalluvan T, Murugesan R, Cook JA, Mitchell JB, Krishna MC. Electron paramagnetic resonance imaging of tumor hypoxia: Enhanced spatial and temporal resolution for in vivo pO(2) determination. Magn Reson Med. 2006;55:1157–1163. doi: 10.1002/mrm.20872. [DOI] [PubMed] [Google Scholar]
  • [28].van Furth R. Origin and turnover of monocytes and macrophages. Curr Top Pathol. 1989;79:125–150. [PubMed] [Google Scholar]
  • [29].Leek RD, Lewis CE, Whitehouse R, Greenall M, Clarke J, Harris AL. Association of macrophage infiltration with angiogenesis and prognosis in invasive breast carcinoma. Cancer Res. 1996;56:4625–4629. [PubMed] [Google Scholar]
  • [30].Leek RD, Landers RJ, Harris AL, Lewis CE. Necrosis correlates with high vascular density and focal macrophage infiltration in invasive carcinoma of the breast. Br J Cancer. 1999;79:991–995. doi: 10.1038/sj.bjc.6690158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Murdoch C, Giannoudis A, Lewis CE. Mechanisms regulating the recruitment of macrophages into hypoxic areas of tumors and other ischemic tissues. Blood. 2004;104:2224–2234. doi: 10.1182/blood-2004-03-1109. [DOI] [PubMed] [Google Scholar]
  • [32].Lewis C, Murdoch C. Macrophage responses to hypoxia: implications for tumor progression and anti-cancer therapies. Am J Pathol. 2005;167:627–635. doi: 10.1016/S0002-9440(10)62038-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Pigott KH, Hill SA, Chaplin DJ, Saunders MI. Microregional fluctuations in perfusion within human tumours detected using laser Doppler flowmetry. Radiother Oncol. 1996;40:45–50. doi: 10.1016/0167-8140(96)01730-6. [DOI] [PubMed] [Google Scholar]
  • [34].Chaplin DJ, Hill SA. Temporal heterogeneity in microregional erythrocyte flux in experimental solid tumours. Br J Cancer. 1995;71:1210–1213. doi: 10.1038/bjc.1995.235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Condeelis J, Segall JE. Intravital imaging of cell movement in tumours. Nat Rev Cancer. 2003;3:921–30. doi: 10.1038/nrc1231. [DOI] [PubMed] [Google Scholar]
  • [36].Bosco MC, Puppo M, Pastorino S, Mi Z, Melillo G, Massazza S, Rapisarda A, Varesio L. Hypoxia selectively inhibits monocyte chemoattractant protein-1 production by macrophages. J Immunol. 2004;172:1681–1690. doi: 10.4049/jimmunol.172.3.1681. [DOI] [PubMed] [Google Scholar]
  • [37].Bosco MC, Reffo G, Puppo M, Varesio L. Hypoxia inhibits the expression of the CCR5 chemokine receptor in macrophages. Cell Immunol. 2004;228:1–7. doi: 10.1016/j.cellimm.2004.03.006. [DOI] [PubMed] [Google Scholar]
  • [38].Grimshaw MJ, Balkwill FR. Inhibition of monocyte and macrophage chemotaxis by hypoxia and inflammation—a potential mechanism. Eur J Immunol. 2001;31:480–489. doi: 10.1002/1521-4141(200102)31:2<480::aid-immu480>3.0.co;2-l. [DOI] [PubMed] [Google Scholar]
  • [39].Fu X, Kassim SY, Parks WC, Heinecke JW. Hypochlorous acid oxygenates the cysteine switch domain of pro-matrilysin (MMP-7). A mechanism for matrix metalloproteinase activation and atherosclerotic plaque rupture by myeloperoxidase. J Biol Chem. 2001;276:41279–41287. doi: 10.1074/jbc.M106958200. [DOI] [PubMed] [Google Scholar]
  • [40].Fu X, Kao JL, Bergt C, Kassim SY, Huq NP, d’Avignon A, Parks WC, Mecham RP, Heinecke JW. Oxidative cross-linking of tryptophan to glycine restrains matrix metalloproteinase activity: specific structural motifs control protein oxidation. J Biol Chem. 2004;279:6209–6212. doi: 10.1074/jbc.C300506200. [DOI] [PubMed] [Google Scholar]
  • [41].Metchnikov E. Uber eine sprosspilzkrankheit der daphnien. Beitrag zur lehre uber den kampf der phagocyten gegen krankheitserreger. Virchows Arch. 1884;96:177–195. [Google Scholar]
  • [42].Silverstein AM. History of immunology. Cellular versus humoral immunity: determinants and consequences of an epic 19th century battle. Cell Immunol. 1979;48:208–221. doi: 10.1016/0008-8749(79)90113-8. [DOI] [PubMed] [Google Scholar]
  • [43].Iwabe N, Kuma K, Miyata T. Evolution of gene families and relationship with organismal evolution: rapid divergence of tissue-specific genes in the early evolution of chordates. Mol. Biol. Evol. 1996;13:483–493. doi: 10.1093/oxfordjournals.molbev.a025609. [DOI] [PubMed] [Google Scholar]
  • [44].Cox RL, Mariano T, Heck DE, Laskin JD, Stegeman JJ. Nitric oxide synthase sequences in the marine fish Stenotomus chrysops and the sea urchin Arbacia punctulata, and phylogenetic analysis of nitric oxide synthase calmodulin-binding domains. Comp Biochem Physiol B Biochem Mol Biol. 2001;130:479–491. doi: 10.1016/s1096-4959(01)00446-8. [DOI] [PubMed] [Google Scholar]
  • [45].Chung S, Seacombes CJ. Analysis of events occurring within teleost macrophages during the respiratory burst. Comp Biochem Phys. 1988;89B:539–544. [Google Scholar]
  • [46].Sharp GJE, Secombes CJ. The role of reactive oxygen species in the killing of the bacterial fish pathogen Aeromonas salmonicida by rainbow trout macrophages. Fish Shellfish Immunol. 1993;3:119–129. [Google Scholar]
  • [47].Laing KJ, Grabowski PS, Belosevic M, Secombes CJ. A partial sequence for nitric oxide synthase from a goldfish (Carassius auratus) macrophage cell line. Immunol Cell Biol. 1996;74:374–379. doi: 10.1038/icb.1996.65. [DOI] [PubMed] [Google Scholar]
  • [48].Gallo RL, Nizet V. Endogenous production of antimicrobial peptides in innate immunity and human disease. Curr Allergy Asthma Rep. 2003;3:402–409. doi: 10.1007/s11882-003-0074-x. [DOI] [PubMed] [Google Scholar]
  • [49].Zanetti M. Cathelicidins, multifunctional peptides of the innate immunity. J Leukoc Biol. 2004;75::39–48. doi: 10.1189/jlb.0403147. [DOI] [PubMed] [Google Scholar]
  • [50].Bowdish DM, Davidson DJ, Scott MG, Hancock RE. Immunomodulatory activities of small host defense peptides. Antimicrob Agents Chemother. 2005;49:1727–1732. doi: 10.1128/AAC.49.5.1727-1732.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Garcia JR, Jaumann F, Schulz S, Krause A, Rodriguez-Jimenez J, Forssmann U, Adermann K, Kluver E, Vogelmeier C, Becker D, Hedrich R, Forssmann WG, Bals R. Identification of a novel, multifunctional beta-defensin (human beta-defensin 3) with specific antimicrobial activity. Its interaction with plasma membranes of Xenopus oocytes and the induction of macrophage chemoattraction. Cell Tissue Res. 2001;306:257–264. doi: 10.1007/s004410100433. [DOI] [PubMed] [Google Scholar]
  • [52].Koczulla R, von Degenfeld G, Kupatt C, Krotz F, Zahler S, Gloe T, Issbrucker K, Unterberger P, Zaiou M, Lebherz C, Karl A, Raake P, Pfosser A, Boekstegers P, Welsch U, Hiemstra PS, Vogelmeier C, Gallo RL, Clauss M, Bals R. An angiogenic role for the human peptide antibiotic LL-37/hCAP-18. J Clin Invest. 2003;111:1665–1672. doi: 10.1172/JCI17545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Li J, Post M, Volk R, Gao Y, Li M, Metais C, Sato K, Tsai J, Aird W, Rosenberg RD, Hampton TG, Sellke F, Carmeliet P, Simons M. PR39, a peptide regulator of angiogenesis. Nat Med. 2000;6:49–55. doi: 10.1038/71527. [DOI] [PubMed] [Google Scholar]
  • [54].Ginsburg I. Cationic polyelectrolytes: potent opsonic agents which activate the respiratory burst in leukocytes. Free Radic Res Commun. 1989;8:11–26. doi: 10.3109/10715768909087968. [DOI] [PubMed] [Google Scholar]
  • [55].Wilson CL, Ouellette AJ, Satchell DP, Ayabe T, Lopez-Boado YS, Stratman JL, Hultgren SJ, Matrisian LM, Parks WC. Regulation of intestinal alpha-defensin activation by the metalloproteinase matrilysin in innate host defense. Science. 1999;286:113–117. doi: 10.1126/science.286.5437.113. [DOI] [PubMed] [Google Scholar]
  • [56].Shi J, Ross CR, Leto TL, Blecha F. PR-39, a proline-rich antibacterial peptide that inhibits phagocyte NADPH oxidase activity by binding to Src homology 3 domains of p47phox. Proc. Natl. Acad. Sci. USA. 1996;93:6014–6018. doi: 10.1073/pnas.93.12.6014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Madhani M, Barchowsky A, Klei L, Ross CR, Jackson SK, Swartz HM, James PE. Antibacterial peptide PR-39 affects local nitric oxide and preserves tissue oxygenation in the liver during septic shock. Biochim Biophys Acta. 2002;1588:232–240. doi: 10.1016/s0925-4439(02)00170-9. [DOI] [PubMed] [Google Scholar]
  • [58].Peyssonnaux C, Datta V, Cramer T, Doedens A, Theodorakis EA, Gallo RL, Hurtado-Ziola N, Nizet V, Johnson RS. HIF-1alpha expression regulates the bactericidal capacity of phagocytes. J Clin Invest. 2005;115:1806–1815. doi: 10.1172/JCI23865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Semenza GL. Hypoxia-inducible factor 1: oxygen homeostasis and disease pathophysiology. Trends Mol Med. 2001;7:345–350. doi: 10.1016/s1471-4914(01)02090-1. [DOI] [PubMed] [Google Scholar]
  • [60].Abu-Soud HM, Ichimori K, Nakazawa H, Stuehr DJ. Regulation of inducible nitric oxide synthase by self-generated NO. Biochemistry. 2001;40:6876–6881. doi: 10.1021/bi010066m. [DOI] [PubMed] [Google Scholar]
  • [61].Santolini J, Meade AL, Stuehr DJ. Differences in three kinetic parameters underpin the unique catalytic profiles of nitric-oxide synthases I, II, and III. J Biol Chem. 2001;276:48887–48898. doi: 10.1074/jbc.M108666200. [DOI] [PubMed] [Google Scholar]
  • [62].Xia Y, Roman LJ, Masters BSS, Zweier JL. Inducible Nitric-oxide Synthase Generates Superoxide from the Reductase Domain. J Biol Chem. 1998;273:22635–22639. doi: 10.1074/jbc.273.35.22635. [DOI] [PubMed] [Google Scholar]
  • [63].Reif A, Shutenko ZV, Feelisch M, Schmidt HH. Superoxide dismutase and catalase are required to detect .-NO from both coupled and uncoupled neuronal no synthase. Free Radic Biol Med. 2004;37:988–997. doi: 10.1016/j.freeradbiomed.2004.07.005. [DOI] [PubMed] [Google Scholar]
  • [64].Sessa WC. eNOS at a glance. J Cell Sci. 2004;117:2427–2429. doi: 10.1242/jcs.01165. [DOI] [PubMed] [Google Scholar]
  • [65].Tendler DS, Bao C, Wang T, Huang EL, Ratovitski EA, Pardoll DA, Lowenstein CJ. Intersection of interferon and hypoxia signal transduction pathways in nitric oxide-induced tumor apoptosis. Cancer Res. 2001;61:3682–3688. [PubMed] [Google Scholar]
  • [66].Klimp AH, Hollema H, Kempinga C, van der Zee AG, de Vries EG, Daemen T. Expression of cyclooxygenase-2 and inducible nitric oxide synthase in human ovarian tumors and tumor-associated macrophages. Cancer Res. 2001;61:7305–7309. [PubMed] [Google Scholar]
  • [67].Onita T, Ji PG, Xuan JW, Sakai H, Kanetake H, Maxwell PH, Fong GH, Gabril MY, Moussa M, Chin JL. Hypoxia-induced, perinecrotic expression of endothelial Per-ARNT-Sim domain protein-1/hypoxia-inducible factor-2alpha correlates with tumor progression, vascularization, and focal macrophage infiltration in bladder cancer. Clin Cancer Res. 2002;8:471–478. [PubMed] [Google Scholar]
  • [68].Burke B, Tang N, Corke KP, Tazzyman D, Ameri K, Wells M, Lewis CE. Expression of HIF-1alpha by human macrophages: implications for the use of macrophages in hypoxia-regulated cancer gene therapy. J Pathol. 2002;196:204–212. doi: 10.1002/path.1029. [DOI] [PubMed] [Google Scholar]
  • [69].Melillo G, Taylor LS, Brooks A, Musso T, Cox GW, Varesio L. Functional requirement of the hypoxia-responsive element in the activation of the inducible nitric oxide synthase promoter by the iron chelator desferrioxamine. J Biol Chem. 1997;272:12236–12243. doi: 10.1074/jbc.272.18.12236. [DOI] [PubMed] [Google Scholar]
  • [70].Angele MK, Schwacha MG, Smail N, Catania RA, Ayala A, Cioffi WG, Chaudry IH. Hypoxemia in the absence of blood loss upregulates iNOS expression and activity in macrophages. Am J Physiol. 1999;276:C285–C290. doi: 10.1152/ajpcell.1999.276.2.C285. [DOI] [PubMed] [Google Scholar]
  • [71].Thomas DD, Espey MG, Ridnour LA, Hofseth LJ, Mancardi D, Harris CC, Wink DA. Hypoxic inducible factor 1alpha, extracellular signal-regulated kinase, and p53 are regulated by distinct threshold concentrations of nitric oxide. Proc Natl Acad Sci USA. 2004;101:8894–8899. doi: 10.1073/pnas.0400453101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Hagen T, Taylor CT, Lam F, Moncada S. Redistribution of intracellular oxygen in hypoxia by nitric oxide: effect on HIF1alpha. Science. 2003;302:1975–1978. doi: 10.1126/science.1088805. [DOI] [PubMed] [Google Scholar]
  • [73].Callapina M, Zhou J, Schmid T, Kohl R, Brune B. NO restores HIF-1alpha hydroxylation during hypoxia: role of reactive oxygen species. Free Radic Biol Med. 2005;39:925–936. doi: 10.1016/j.freeradbiomed.2005.05.009. [DOI] [PubMed] [Google Scholar]
  • [74].Thomas DD, Ridnour LA, Espey MG, Donzelli S, Ambs S, Hussain SP, Harris CC, Degraff W, Roberts DD, Mitchell JB, Wink DA. Superoxide fluxes limit nitric oxide-induced signaling. J Biol Chem. doi: 10.1074/jbc.M602242200. in press. [DOI] [PubMed] [Google Scholar]
  • [75].Gabig TG, Bearman SI, Babior BM. Effects of oxygen tension and pH on the respiratory burst of human neutrophils. Blood. 1979;53:1133–1139. [PubMed] [Google Scholar]
  • [76].Edwards SW, Lloyd D. The relationship between superoxide generation, cytochrome b and oxygen in activated neutrophils. FEBS Lett. 1988;227:39–42. doi: 10.1016/0014-5793(88)81409-1. [DOI] [PubMed] [Google Scholar]
  • [77].Farias-Eisner R, Chaudhuri G, Aeberhard E, Fukuto JM. The chemistry and tumoricidal activity of nitric oxide/hydrogen peroxide and the implications to cell resistance/susceptibility. J Biol Chem. 1996;271:6144–6151. doi: 10.1074/jbc.271.11.6144. [DOI] [PubMed] [Google Scholar]
  • [78].Kusmartsev S, Nefedova Y, Yoder D, Gabrilovich DI. Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. J Immunol. 2004;172:989–999. doi: 10.4049/jimmunol.172.2.989. [DOI] [PubMed] [Google Scholar]
  • [79].Cheng G, Cao Z, Xu X, van Meir EG, Lambeth JD. Homologs of gp91phox: cloning and tissue expression of Nox3, Nox4, and Nox5. Gene. 2001;269:131–140. doi: 10.1016/s0378-1119(01)00449-8. [DOI] [PubMed] [Google Scholar]
  • [80].Thomas DD, Espey MG, Vitek MP, Miranda KM, Wink DA. Protein nitration is mediated by heme and free metals through Fenton-type chemistry: an alternative to the NO/O2-reaction. Proc Natl Acad Sci USA. 2002;99:12691–12696. doi: 10.1073/pnas.202312699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Espey MG, Xavier S, Thomas DD, Miranda KM, Wink DA. Direct real-time evaluation of nitration with green fluorescent protein in solution and within human cells reveals the impact of nitrogen dioxide vs. peroxynitrite mechanisms. Proc Natl Acad Sci USA. 2002;99:3481–3486. doi: 10.1073/pnas.062604199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med. 1986;315:1650–1659. doi: 10.1056/NEJM198612253152606. [DOI] [PubMed] [Google Scholar]
  • [83].Albina JE, Reichner JS. Oxygen and the regulation of gene expression in wounds. Wound Repair Regen. 2003;11:445–451. doi: 10.1046/j.1524-475x.2003.11619.x. [DOI] [PubMed] [Google Scholar]
  • [84].Albina JE, Henry WL, Jr, Mastrofrancesco B, Martin BA, Reichner JS. Macrophage activation by culture in an anoxic environment. J Immunol. 1995;155:4391–4396. [Google Scholar]
  • [85].Morris SW., Jr. Regulation of arginine availability and its impact on NO synthesis. In: Ignarro LJ, editor. Nitric Oxide. Academic Press; San Diego: 2000. pp. 187–197. [Google Scholar]
  • [86].Albina JE, Mastrofrancesco B, Vessella JA, Louis CA, Henry WL, Jr., Reichner JS. HIF-1 expression in healing wounds: HIF-1alpha induction in primary inflammatory cells by TNF-alpha. Am J Physiol Cell Physiol. 2001;281:C1971–1977. doi: 10.1152/ajpcell.2001.281.6.C1971. [DOI] [PubMed] [Google Scholar]
  • [88].Kusmartsev S, Gabrilovich DI. STAT1 signaling regulates tumor-associated macrophage-mediated T cell deletion. J Immunol. 2005;174:4880–4891. doi: 10.4049/jimmunol.174.8.4880. [DOI] [PubMed] [Google Scholar]
  • [89].Fishman M. Functional heterogeneity among peritoneal macrophages. III. No evidence for the role of arginase in the inhibition of tumor cell growth by supernatants from macrophages or macrophage subpopulation cultures. Cell Immunol. 1980;55:174–184. doi: 10.1016/0008-8749(80)90148-3. [DOI] [PubMed] [Google Scholar]
  • [90].Munder M, Eichmann K, Modolell M. Alternative metabolic states in murine macrophages reflected by the nitric oxide synthase/arginase balance: competitive regulation by CD4+ T cells correlates with Th1/Th2 phenotype. J Immunol. 1998;160:5347–5354. [PubMed] [Google Scholar]
  • [91].Munder M, Mollinedo F, Calafat J, Canchado J, Gil-Lamaignere C, Fuentes JM, Luckner C, Doschko G, Soler G, Eichmann K, Muller FM, Ho AD, Goerner M, Modolell M. Arginase I is constitutively expressed in human granulocytes and participates in fungicidal activity. Blood. 2005;105:2549–2556. doi: 10.1182/blood-2004-07-2521. [DOI] [PubMed] [Google Scholar]
  • [91].Ochoa JB, Bernard AC, O’Brien WE, Griffen MM, Maley ME, Rockich AK, Tsuei BJ, Boulanger BR, Kearney PA, Morris SM., Jr. Arginase I expression and activity in human mononuclear cells after injury. Ann Surg. 2001;233:393–399. doi: 10.1097/00000658-200103000-00014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Rouzaut A, Subira ML, de Miguel C, Domingo-de-Miguel E, Gonzalez A, Santiago E, Lopez-Moratalla N. Co-expression of inducible nitric oxide synthase and arginases in different human monocyte subsets. Apoptosis regulated by endogenous NO. Biochim Biophys Acta. 1999;1451:319–333. doi: 10.1016/s0167-4889(99)00106-8. [DOI] [PubMed] [Google Scholar]
  • [92].Rodriguez PC, Quiceno DG, Zabaleta J, Ortiz B, Zea AH, Piazuelo MB, Delgado A, Correa P, Brayer J, Sotomayor EM, Antonia S, Ochoa JB, Ochoa AC. Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res. 2004;64:5839–5849. doi: 10.1158/0008-5472.CAN-04-0465. [DOI] [PubMed] [Google Scholar]
  • [93].Tsai MC, Chakravarty S, Zhu G, Xu J, Tanaka K, Koch C, Tufariello J, Flynn J, Chan J. Characterization of the tuberculous granuloma in murine and human lungs: cellular composition and relative tissue oxygen tension. Cell Microbiol. 2006;8:218–232. doi: 10.1111/j.1462-5822.2005.00612.x. [DOI] [PubMed] [Google Scholar]

RESOURCES