key: cord-299967-90aknp7c authors: Terry, Rachael L; Getts, Daniel R; Deffrasnes, Celine; van Vreden, Caryn; Campbell, Iain L; King, Nicholas JC title: Inflammatory monocytes and the pathogenesis of viral encephalitis date: 2012-12-17 journal: J Neuroinflammation DOI: 10.1186/1742-2094-9-270 sha: doc_id: 299967 cord_uid: 90aknp7c Monocytes are a heterogeneous population of bone marrow-derived cells that are recruited to sites of infection and inflammation in many models of human diseases, including those of the central nervous system (CNS). Ly6C(hi)/CCR2(hi) inflammatory monocytes have been identified as the circulating precursors of brain macrophages, dendritic cells and arguably microglia in experimental autoimmune encephalomyelitis; Alzheimer’s disease; stroke; and more recently in CNS infection caused by Herpes simplex virus, murine hepatitis virus, Theiler’s murine encephalomyelitis virus, Japanese encephalitis virus and West Nile virus. The precise differentiation pathways and functions of inflammatory monocyte-derived populations in the inflamed CNS remains a contentious issue, especially in regard to the existence of monocyte-derived microglia. Furthermore, the contributions of monocyte-derived subsets to viral clearance and immunopathology are not well-defined. Thus, understanding the pathways through which inflammatory monocytes migrate to the brain and their functional capacity within the CNS is critical to inform future therapeutic strategies. This review discusses some of the key aspects of inflammatory monocyte trafficking to the brain and addresses the role of these cells in viral encephalitis. Virus infection of the brain can cause severe and lifethreatening disease. Despite this, few therapies beyond intensive supportive care are available to treat patients with encephalitis [1, 2] . Anti-viral drugs have been developed for some viruses that can infect the brain, such as Herpes simplex virus (HSV)-1 and 2, and human immunodeficiency virus (HIV), but even with these treatments outcomes remain relatively poor [2] [3] [4] [5] . Many patients succumb to disease, and survivors often suffer permanent neurological sequelae [6] [7] [8] [9] . While the development and clinical implementation of novel anti-viral drugs may improve patient outcomes, it is becoming increasingly clear that therapies targeting pathogenic elements of the host immune response may be critical for successful intervention during infection [10] [11] [12] [13] [14] . Monocyte infiltration is a hallmark of central nervous system (CNS) inflammation, including viral infection. These cells migrate into the infected brain, where they differentiate into dendritic cell (DC), macrophage and, arguably, microglial populations. Once differentiated, these cells engage in a number of potent effector functions including antigen presentation and T cell stimulation, the production and secretion of numerous pro-inflammatory mediators as well as reactive oxygen species (ROS), all of which are focused on viral containment and clearance (Table 1) . However, unbalanced and poorly controlled migration and effector functions of these cells may result in immune-mediated pathology, resulting in tissue damage and destruction during some infections (Table 1) . Therefore, it is of high importance to understand the processes driving monocyte development, recruitment, differentiation and function, to aid in the development of novel therapeutics that inhibit immunopathological responses. Monocytes are derived from hematopoietic stem cells (HSC) in the bone marrow (BM) (Figure 1 ). The earliest defined precursor is the common myeloid precursor (CMP), distinguished from HSC by the expression of CD34 but not SCA-1 [39] [40] [41] [42] (Figure 1 ). These cells give rise to a pool of precursors called granulocyte/macrophage precursors (GMPs), which express CD16/32 [39] . Included within this subset is the recently defined macrophage/DC precursor (MDP), which specifically expresses high levels of the PU.1-controlled chemokine receptor CD115 (CSF-1R/ M-CSFR), chemokine receptor CX 3 CR 1 (fractalkine receptor), and Flt-3 (CD135/Flk2) [43] [44] [45] [46] [47] [48] (Figure 1 ). The MDP gives rise to CD11b + , CD115 + , F4/80 + , CD11c -, Ly6Gmonocytes, that can be isolated from the BM and [29] Inhibition of NOS2 prolonged survival of WNVinfected animals [30] Reviewed in [31] Proteases MMP ↑ breakdown of the BBB MMP-9 −/− mice show reduced viral loads and increased survival during WNV encephalitis [32] ↑ neuronal damage/ death ↑ demyelination ↑ pro-inflammatory cytokines Reviewed in [33, 34] Neurotransmitters Glutamate ↑ neuronal misfiring/ seizures Competitive and non-competitive glutamate receptor antagonists promote survival during neurovirulent Sindbis virus encephalitis [35, 36] and improved outcomes during coronavirus encephalitis [37] ↑ neuronal damage/ death ↑ production of NO/ ROS Reviewed in [38] BBB blood brain barrier; CNS central nervous system; HSV herpes simplex virus; MDP macrophage/dendritic cell precursor; MHV murine hepatitis virus; MMP matrix metalloproteinases; NO nitric oxide; NOS2 nitric oxide synthase-2; ROS reactive oxygen species; TMEV Theiler's murine encephalomyelitis virus; WNV West Nile virus. blood [49] [50] [51] [52] (Figure 1 ). The spleen has also been identified as an important reservoir of undifferentiated monocytes that are rapidly deployed to sites of inflammation, including the ischemic heart and brain [53] [54] [55] . Furthermore, a recent study has shown that cardiac infarction triggers a significant increase in numbers of MDPs in the spleen, which supply monocytes throughout the duration of acute inflammation [56] . Whether the spleen is a significant source of monocytes during CNS infection is yet to be determined, but presents a critical area of future investigation. It is likely that both the BM and spleen are critical for supplying monocytes to the infected CNS, particularly in cases of acute and severe infection, in which large numbers of these cells are rapidly deployed and recruited to the brain. Monocytes are classified into two phenotypically and functionally distinct subsets The MDPs give rise to two phenotypically and functionally distinct subsets of monocytes [50, 57] . Ly6C hi monocytes are characterized by high expression of the chemokine receptor CCR2, adhesion molecule CD62L and low expression of the fractalkine receptor CX 3 CR 1 [48, 51, 58] . These cells have been termed 'inflammatory' because they are selectively recruited to sites of inflammation and infection in many models of disease, including atherosclerosis [59] [60] [61] [62] ; rheumatoid arthritis [63] ; experimental colitis [64] ; cardiac infarction [65] ; and CNS infections including experimental autoimmune encephalomyelitis (EAE) [66, 67] , amyotrophic lateral sclerosis [68] , and stroke [53] . Recent studies have shown that these cells are also recruited to the virus-infected brain in animal models of HSV, HIV, murine hepatitis virus (MHV), Theiler's murine encephalomyelitis virus (TMEV) and a number of flaviviral encephalitides, where they give rise to macrophage, DC and, arguably, to microglial populations [11, 13, 14, 69] . Conversely, Ly6C lo/monocytes are smaller in size than their Ly6C hi counterparts and express low levels of CCR2 and CD62L and high levels of CX 3 CR 1 [48, 51, 58] (Figure 1 ). Development of monocytes in the bone marrow and recruitment to the virus-infected brain. Monocytes are generated from hematopoietic precursors in the bone marrow (BM). Sca-1 + Lin -HSC (a) give rise to CD34 + , Sca-1 -CMP (b). These cells in turn give rise to a pool of precursors known as granulocyte/macrophage precursors (GMPs), which express CD34 and CD16/32 (c). A fraction of these progenitors also express CD115 and CX 3 CR1 and are known as macrophage/dendritic cell precursor (MDP) (d). MDPs are the direct precursors of Ly6C hi inflammatory monocytes (e). MDPs also give rise to circulating Ly6C lo/monocytes directly, or via a Ly6C hi monocyte intermediate (f). During viral encephalitis, large quantities of the chemokine CCL2 is produced by infected astrocytes, macrophages/microglia and/or neurons (g). CCL2 binds the chemokine receptor CCR2, expressed at high levels by Ly6C hi inflammatory monocytes, which promotes the egress of these cells from the BM (h) into the blood, and thus recruitment from the blood into the infected central nervous system (CNS) (i). Here, these cells can give rise to CD45 hi Ly6C hi macrophages (j) and/or CD45 int Ly6C int immigrant microglia (k), although it is unclear whether Ly6C int immigrant microglia are derived from a Ly6C hi macrophage intermediate or directly differentiate from Ly6C hi monocytes. Furthermore, it is unclear whether recruited macrophages and immigrant microglia give rise to CD45 lo Ly6C lo/resident microglia (l) if/when virus is cleared from the CNS. In some models of viral encephalitis, Ly6C hi inflammatory monocytes can also give rise to Ly6C hi /CD11c + DC in the brain (m). Several studies have shown that Ly6C hi monocytes can give rise to circulating Ly6C lo/monocytes [58, [70] [71] [72] . Interest in this subset has increased substantially in the past few years [72, 73] . Recent studies have described the patrolling behavior of these cells in the vasculature [73] , and have shown that in some models of disease they rapidly enter inflamed tissue and can contribute to early inflammatory responses before domination by Ly6C hi monocytes [73] . In the resolution phase of some diseases, Ly6C lo/monocytes are critical for wound healing and angiogenesis [50] . While apparently important in the periphery, the role of Ly6C lo/monocytes during CNS infection remains poorly defined, with little evidence supporting their migration into the brain during inflammation [74] . The importance of monocyte-derived cells in the pathogenesis of brain infection highlights the importance of understanding the pathway(s) through which monocytes migrate from the periphery into the brain. It is apparent that this process is regulated by cytokine/chemokine and integrin/cellular adhesion molecule interactions that facilitate emigration from the BM into the blood and entry into the CNS. For example, the chemokine receptor CXCR4 and one of its ligands CXCL12 (SDF-1) directly enhance VLA-4-dependent adhesion and thereby aid in retaining immature cells in the BM. Deficiency in either molecule results in impaired myelopoiesis [75] [76] [77] [78] [79] [80] . In addition to CXCR4, CCR2 and its ligands, CCL2 and CCL7 (MCP-3), are a critical requirement for Ly6C hi monocyte egress from the BM into the blood. CCL2/ CCR2 deficiency or blockade with antibody results in monocyte accumulation in the BM in multiple disease models, including EAE, WNV and HSV encephalitides [11, 61, 67, [81] [82] [83] [84] [85] [86] [87] . Monocyte recruitment into the infected brain is dependent on chemokine/chemokine receptor interactions A number of chemokines and their receptors have been implicated in the recruitment of Ly6C hi monocytes from the blood and into the brain. CCR5 is expressed by Ly6C hi monocytes and is important for trafficking to sites of inflammation in some models of disease. In the brain, its ligand CCL5 (RANTES) expression is highly upregulated during infection/inflammation, including WNV, MHV, HSV and tick-borne encephalitis virus encephalitides [88] [89] [90] [91] [92] . Another chemokine of interest that controls the trafficking of monocytes into the brain parenchyma is SDF-1/CXCL12, in conjunction with its receptor CXCR4, expressed by monocytes [93] . In animal models of CNS inflammation including EAE [94] , HIV [95] and WNV [96] , there is significant upregulation of CXCL12. In EAE and WNV, CXCL12 has been shown to play an important role in retaining leukocytes in the perivascular space, thereby inhibiting infiltration into the parenchyma. Loss of this interaction resulted in the loss of perivascular cuffs and uncontrolled infiltration of CXCR4 + leukocytes, including monocytes, into the parenchyma. [94, 96] . While it is clear that there are a multitude of soluble mediators that represent potential targets for future therapies aimed at blocking monocyte migration, the CCR2/ CCL2 axis remains the most potent pathway based on the available literature. Ly6C hi /CCR2 hi monocyte recruitment into the CNS in models of stroke [53] , peripheral inflammation [97] , Alzheimer's disease (AD) [98, 99] and EAE [67, 74, 100, 101] are all dependent on CCR2/CCL2 signaling ( Figure 1 ). In the context of viral encephalitis, the CCL2/CCR2 axis is also very important. The major producers of CCL2 appear to be different depending on the infectious agent, with microglia serving as important sources during HSV infection [16, 102] , neurons in the case of WNV infection [11] and astrocytes in HIV encephalitis [103] . No matter the source of CCL2, the inhibition of CCL2 can significantly reduce the infiltration of inflammatory monocyte-derived macrophages and microglia into the infected brain [11] [12] [13] 69, 88, 102, [104] [105] [106] [107] [108] . The focus in the last decade has been heavily on the chemokines involved in monocyte trafficking, however, cellular adhesion molecules and their integrin ligands are obviously also important. In most models of viral infection, very late antigen-4 (VLA-4) and leukocyte function-associated antigen-1 (LFA-1) are expressed by Ly6C hi monocytes. In addition, their respective binding partner's vascular cell adhesion molecule-1 (VCAM-1) and inter-cellular adhesion molecule-1 (ICAM-1) are usually upregulated on endothelium and other cell types in the inflamed brain [109] [110] [111] [112] [113] [114] [115] . The importance of VLA-4 and VCAM-1 and LFA-1 and ICAM-1 in the recruitment of Ly6C hi monocytes to sites of inflammation is evident in experiments using gene knockout animals or specific blockade of these molecules. VLA-4 and VCAM-1 interactions are critical for monocyte migration to the heart in models of atherosclerosis and arterial injury [116] [117] [118] and the inflamed peritoneum [119] . VLA-4 is also critical for Ly6C hi monocyte infiltration of the CNS in several models of inflammation, including EAE and spinal cord injury [97, 109, 120] . During viral infection of the brain, we have found that recruitment of monocytes to the CNS is also VLA-4-dependent. VLA-4 antibody neutralization significantly impairs the recruitment of Ly6C hi monocytes to the infected brain, in both WNV and JEV infection ( [30] , CvV et al., unpublished observations). LFA-1 and ICAM-1 interactions are also important for monocyte recruitment to atherosclerotic plaques [121, 122] and to the CNS during EAE [110] . We have shown that LFA-1 is also important for recruitment of monocytes to the WNV-infected brain, however blockade resulted in a smaller reduction in monocytes infiltration compared to VLA-4 neutralization, which suggests the differential use of adhesion molecules by Ly6C hi monocyte subsets which enter the WNV-infected brain [30] . In models of CNS diseases, such as EAE and stroke, Ly6C hi monocytes have been shown to primarily differentiate into macrophage and DC populations exhibiting a M1 pro-inflammatory phenotype, which in-vitro effectively stimulates Th1 and Th17 responses in T cells [53, 66, 67, 74] . Similarly, in models of viral encephalitis, Ly6C hi monocytes have been shown to give rise to M1 pro-inflammatory CD45 hi macrophages and CD11c + DC populations, which express high levels of nitric oxide (NO) and TNF during HSV, WNV, MHV, TMEV and JEV ( [11] [12] [13] [14] 30, 69] , CvV et al., unpublished observations). We have shown that these CD45 hi macrophages are highly effective at processing and presenting antigen and effectively stimulate T cell proliferation [30] . Microglia are the resident macrophage population of the brain. Similar to other tissue resident cells such as Kupffer cells of the liver and Langerhans cells of the epidermis, microglia originate from the yolk sac during embryogenesis, from a myeloid lineage that is independent of BM HSC and therefore distinct from that of BM-derived monocytes [123] [124] [125] . Microglia can be distinguished from infiltrating monocyte-derived macrophages and DC by their low to intermediate expression of CD45 and lack of Ly6C expression [11, 126] . In most infections, resident microglia play functionally distinct roles from that of monocyte-derived cells. For example, during acute WNV encephalitis, resident microglia express lower levels of proinflammatory mediators such as NO, express lower levels of MHC-II, and show a significantly reduced capacity to process antigen and stimulate T cell proliferation compared to the highly activated infiltrating macrophages [30] . In comparison, in acute TMEV infection, resident microglia and infiltrating macrophages express similar levels of proinflammatory cytokines and show similar antigen processing and presentation capacity; however, in chronic stages of disease, macrophages are more efficient at stimulating T cell responses [127] . There is evidence to suggest that infiltrating monocytes have the capacity to give rise to microglial cells in some models of CNS inflammation, including AD, Parkinson's disease, EAE, as well as in infectious models such as scrapie and bacterial meningitis [128] [129] [130] [131] [132] [133] [134] . These immigrant microglial cells appear to play distinct functional roles compared to their resident counterparts during disease. For example, immigrant microglia are more efficient at clearing amyloid plaques than resident microglia during AD [128, 135] . However, a caveat of these studies has been in the use of irradiation to generate BM chimeras to distinguish resident microglial from BMderived cells. There are currently no immunophenotypic markers that can definitively separate these two populations. As a result, the generation of chimeras can be used distinguish tissue resident and BM-derived populations. However, irradiation can disrupt the blood-brain barrier (BBB) and promote CCL2 production, resulting in the recruitment of monocytes to the CNS [136] . Therefore, it is difficult to conclude whether monocyte engraftment is a normal feature of disease in unperturbed animals or whether it is primarily the result of brain preconditioning by irradiation. A recent study using the parabiosis model in place of irradiated BM chimeras has shown that engraftment of monocyte-derived microglia during EAE is only a transient response [137] . The parabiosis models have also been employed to show that there is no significant engraftment of monocytederived microglia in facial nerve axonomy or amyotrophic lateral sclerosis [138] . Also, another recent study has compared the recruitment of monocyte-derived microglia into brain during AD, using chimeric mice generated with or without head protection during irradiation. They found that these cells do not engraft the brain of protected animals [99] . However, one major caveat of the head-protection model is the existence of BM in the skull that may be capable of reconstituting the animal. Further studies are required to definitively determine whether monocyte-derived cells can give rise to microglia and if these cells truly engraft the parenchyma and remain there if/when disease is resolved. There are few studies that examine the recruitment of monocyte-derived microglia during viral infection of the CNS. We have shown that in WNV encephalitis, inflammatory monocytes not only give rise to CD45 hi macrophages in the brain, but also to a CD45 int subset, which is phenotypically analogous to activated resident microglia, apart from the expression of Ly6C [11, 30] . Although chimeras were initially utilized to investigate this phenomenon, we further confirmed that the recruitment of these monocyte-derived cells was not the result of BBB breakdown, using methods that do not use any irradiation including bone marrow adoptive transfer studies and microparticle-based systems which track these cells with minimal perturbation of the disease system [11] . Furthermore, these cells were found to contribute to the immunopathogenesis of WNV encephalitis, as CCL2 blockade significantly reduced recruitment into the CNS and prolonged survival of lethally-infected animals [11] . Current studies in our laboratory aim to determine whether monocyte-derived microglia truly engraft the brain parenchyma during WNV encephalitis, the functional role of these cells throughout infection, and whether these cells remain in the CNS after disease is resolved. Ly6C hi monocytes appear to play a paradoxical role in many disease models. For example, higher mortality rates and increased pathogen loads are seen in Toxoplasma [139, 140] , Listeria [83, 141] , Cryptococcus [142, 143] , Yersinia infections [144] , HSV-2, [145] and coronavirus [146] , as well as MHV [88] when these cells are depleted. On the other hand, Ly6C hi monocytes are direct targets for pathogens such as HIV, TMEV, Listeria and Toxoplasma [12, 69, [147] [148] [149] [150] [151] [152] . Infected monocytes can be directly responsible for the dissemination of infection in a "Trojan horse" fashion into the CNS thereby potentiating disease and increasing potential mortality [153] [154] [155] [156] . Monocytes significantly contribute to immunopathology during brain infection An arguable role of monocytes during brain infection is their potential contribution to immune-mediated pathology. In several models of CNS disease, Ly6C hi inflammatory monocytes cause significant damage and destruction in the brain, directly contributing to morbidity and mortality. Ly6C hi monocytes contribute significantly to the pathogenesis of disease during stroke [53] . Mice with CCL2 −/− and CCR2 −/− deficiency show smaller infarcts and enhanced functional outcomes relative to wild-type controls following transient cerebral ischemia [157, 158] . Similarly, in models of traumatic brain injury, CCL2 −/− mice showed reductions in macrophage infiltration and lesion volume compared to wild-type mice, corresponding with improved functional recovery after injury [159] . In addition, CCR2 −/− and CCL2 −/− mice exhibit milder symptoms and, in some models, are completely resistant to the development of EAE [100, 136, 160, 161] . Furthermore, a recent study has shown that Ly6C hi monocyte recruitment to the CNS is detrimental in amyotrophic lateral sclerosis [68] . In the case of encephalitic disease, studies in our laboratory using WNV as well as others using TMEV have shown that Ly6C hi monocytes are recruited into the infected brain where they contribute significantly to the immunopathogenesis of disease. Inhibition of inflammatory monocyte migration into the WNV or TMEVinfected brain can significantly reduce morbidity and mortality [11, 12, 69, 108] . Furthermore, abrogation of monocyte migration into the CNS during MHV encephalitis results in the delayed onset of demyelinating disease [105] . The precise pathways through which inflammatory monovcytes contribute to pathology are still under intense investigation. However, it is clear that differentiation into effector cells such as macrophages and DC plays a substantial role. Once differentiated, these cells are significant producers of NO, matrix metalloproteinases (MMP) and other factors known to culminate in tissue destruction, breakdown of the BBB, as well as neuronal damage (Table 1) . While in many organs such toxicity is not a major concern due to regenerative capabilities, the brain is largely comprised of many irreplaceable cellular subsets. As such not only is mortality a concern, in patients that survive serious CNS inflammatory insults will often suffer long-term sequelae and neurological imbalance [6] [7] [8] [9] . Although Ly6C hi monocyte infiltration is a hallmark of viral encephalitis, the role of these cells in viral clearance and immunopathology is not well defined. While it is clear that these cells are critical for the control and clearance of some viruses, they are directly responsible for recruiting others into CNS, or cause significant immunopathology. Future studies which target monocyte development and migration to the CNS in a therapeutic manner will not only provide significant insight into pathways by which monocytes are recruited to the CNS, but will identify new targets for intervention during viral encephalitis. The authors declare that they have no competing interests. Authors' contributions RLT drafted the manuscript. DRG, CD, CVV, ILC and NJCK contributed to the interpretation and critical evaluation of content and revision of the manuscript. All authors read and approved the final manuscript. Diagnosis and treatment of viral encephalitis Viral encephalitis: familiar infections and emerging pathogens Outcome of and prognostic factors for herpes simplex encephalitis in adult patients: results of a multicenter study HIV-1 and the brain: connections between HIV-1-associated dementia, neuropathology and neuroimmunology Treatment of viral encephalitis The long-term neuropsychological outcome of herpes simplex encephalitis in a series of unselected survivors Herpes simplex encephalitis treated with acyclovir: diagnosis and long term outcome HIV dementia: an evolving disease Exacerbation of herpes simplex encephalitis after successful treatment with acyclovir Molecular mechanisms of neuroinvasion by monocytesmacrophages in HIV-1 infection Ly6c+ "inflammatory monocytes" are microglial precursors recruited in a pathogenic manner in West Nile virus encephalitis Central neuroinvasion and demyelination by inflammatory macrophages after peripheral virus infection is controlled by SHP-1 Maturation and localization of macrophages and microglia during infection with a neurotropic murine coronavirus Prolonged microglial cell activation and lymphocyte infiltration following experimental herpes encephalitis Resistance of interleukin-1beta-deficient mice to fatal Sindbis virus encephalitis Tumor necrosis factor-alpha and interleukin-1 beta play a critical role in the resistance against lethal herpes simplex virus encephalitis Interleukin-6, produced by resident cells of the central nervous system and infiltrating cells, contributes to the development of seizures following viral infection Banion MK: Chronic interleukin-1beta expression in mouse brain leads to leukocyte infiltration and neutrophil-independent blood brain barrier permeability without overt neurodegeneration Interleukin-1 and neuronal injury Interleukin-6, a mental cytokine Tumor necrosis factor-alpha mediated signaling in neuronal homeostasis and dysfunction TNF signaling inhibition in the CNS: implications for normal brain function and neurodegenerative disease Interleukin-12 (IL-12), but not IL-23, deficiency ameliorates viral encephalitis without affecting viral control Interleukin-12 promotes recovery from viral encephalitis Absence of the p55 Kd TNF-alpha receptor promotes survival in rabies virus acute encephalitis Inducible nitric oxide synthase inhibition delays death of rabies virus-infected mice Role of type I and type II interferon responses in recovery from infection with an encephalitic flavivirus Junin virus-induced astrocytosis is impaired by iNOS inhibition Inhibition of nitric oxide synthesis increases mortality in Sindbis virus encephalitis Targeted blockade in lethal West Nile virus encephalitis shows a critical role for VLA-4-dependant recruitment of nitric oxide-producing macrophages Nitric oxide in the central nervous system: neuroprotection versus neurotoxicity Matrix metalloproteinase 9 facilitates West Nile virus entry into the brain Metalloproteinases: mediators of pathology and regeneration in the CNS Metalloproteinases in biology and pathology of the nervous system Protection from fatal viral encephalomyelitis: AMPA receptor antagonists have a direct effect on the inflammatory response to infection Glutamate receptor antagonists protect from virus-induced neural degeneration Glutamate excitotoxicity is involved in the induction of paralysis in mice after infection by a human coronavirus with a single point mutation in its spike protein Macrophages and neurodegeneration A clonogenic common myeloid progenitor that gives rise to all myeloid lineages Purification and characterization of mouse hematopoietic stem cells Identification of clonogenic common lymphoid progenitors in mouse bone marrow The long-term repopulating subset of hematopoietic stem cells is deterministic and isolatable by phenotype Opposing actions of c-ets/PU.1 and c-myb protooncogene products in regulating the macrophage-specific promoters of the human and mouse colony-stimulating factor-1 receptor (c-fms) genes A two-step, PU.1-dependent mechanism for developmentally regulated chromatin remodeling and transcription of the c-fms gene PU.1 regulates both cytokine-dependent proliferation and differentiation of granulocyte/macrophage progenitors Transcription factor complex formation and chromatin fine structure alterations at the murine c-fms (CSF-1 receptor) locus during maturation of myeloid precursor cells A clonogenic bone marrow progenitor specific for macrophages and dendritic cells CX3CR1+ CD115+ CD135+ common macrophage/DC precursors and the role of CX3CR1 in their response to inflammation A clonogenic bone marrow progenitor specific for macrophages and dendritic cells Blood monocytes: development, heterogeneity, and relationship with dendritic cells Blood monocytes consist of two principal subsets with distinct migratory properties Monocyte subpopulations and their differentiation patterns during infection A role for spleen monocytes in post-ischemic brain inflammation and injury Identification of splenic reservoir monocytes and their deployment to inflammatory sites Angiotensin-converting enzyme inhibition prevents the release of monocytes from their splenic reservoir in mice with myocardial infarction Rapid monocyte kinetics in acute myocardial infarction are sustained by extramedullary monocytopoiesis Development of monocytes, macrophages, and dendritic cells Subpopulations of mouse blood monocytes differ in maturation stage and inflammatory response Monocytes in atherosclerosis: subsets and functions Heterogeneous in vivo behavior of monocyte subsets in atherosclerosis Monocyte subsets differentially employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic plaques Ly-6Chi monocytes dominate hypercholesterolemia-associated monocytosis and give rise to macrophages in atheromata Targeting of Gr-1+,CCR2+ monocytes in collagen-induced arthritis Colonic eosinophilic inflammation in experimental colitis is mediated by Ly6C(high) CCR2(+) inflammatory monocyte/macrophagederived CCL11 The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions Circulating Ly-6C+ myeloid precursors migrate to the CNS and play a pathogenic role during autoimmune demyelinating disease CCR2+Ly-6Chi monocytes are crucial for the effector phase of autoimmunity in the central nervous system Modulating inflammatory monocytes with a unique microRNA gene signature ameliorates murine ALS Modulation of macrophage infiltration and inflammatory activity by the phosphatase SHP-1 in virus-induced demyelinating disease Migratory fate and differentiation of blood monocyte subsets Monocytes give rise to mucosal, but not splenic, conventional dendritic cells Accelerated dendritic cell differentiation from migrating Ly6C(lo) bone marrow monocytes in early dermal West Nile virus infection Monitoring of blood vessels and tissues by a population of monocytes with patrolling behavior Selective chemokine receptor usage by central nervous system myeloid cells in CCR2-red fluorescent protein knock-in mice Defects of B-cell lymphopoiesis and bone-marrow myelopoiesis in mice lacking the CXC chemokine PBSF/ SDF-1 Function of the chemokine receptor CXCR4 in haematopoiesis and in cerebellar development Chemokine stromal cell-derived factor-1alpha modulates VLA-4 integrin-dependent adhesion to fibronectin and VCAM-1 on bone marrow hematopoietic progenitor cells A highly efficacious lymphocyte chemoattractant, stromal cell-derived factor 1 (SDF-1) Integrin alpha4beta7 and its counterreceptor MAdCAM-1 contribute to hematopoietic progenitor recruitment into bone marrow following transplantation Chemokine stromal cell-derived factor-1alpha modulates VLA-4 integrin-mediated multiple myeloma cell adhesion to CS-1/fibronectin and VCAM-1 Monocyte emigration from bone marrow during bacterial infection requires signals mediated by chemokine receptor CCR2 Monocyte trafficking to hepatic sites of bacterial infection is chemokine independent and directed by focal intercellular adhesion molecule-1 expression Additive roles for MCP-1 and MCP-3 in CCR2-mediated recruitment of inflammatory monocytes during Listeria monocytogenes infection CCR2 mediates homeostatic and inflammatory release of Gr1(high) monocytes from the bone marrow, but is dispensable for bladder infiltration in bacterial urinary tract infection Critical roles for CCR2 and MCP-3 in monocyte mobilization from bone marrow and recruitment to inflammatory sites CCR2 and CXCR4 regulate peripheral blood monocyte pharmacodynamics and link to efficacy in experimental autoimmune encephalomyelitis Impact of deficiency in CCR2 and CX3CR1 receptors on monocytes trafficking in herpes simplex virus encephalitis Lack of CCR2 results in increased mortality and impaired leukocyte activation and trafficking following infection of the central nervous system with a neurotropic coronavirus Breakdown of the blood-brain barrier during tick-borne encephalitis in mice is not dependent on CD8+ T-cells Chemokine receptor CCR5 promotes leukocyte trafficking to the brain and survival in West Nile virus infection Reduced macrophage infiltration and demyelination in mice lacking the chemokine receptor CCR5 following infection with a neurotropic coronavirus The chemokine CCL5 is essential for leukocyte recruitment in a model of severe Herpes simplex encephalitis Monocyte migration and LFA-1-mediated attachment to brain microvascular endothelia is regulated by SDF-1 alpha through Lyn kinase CXCL12 limits inflammation by localizing mononuclear infiltrates to the perivascular space during experimental autoimmune encephalomyelitis HIV-1-infected and/or immune activated macrophages regulate astrocyte SDF-1 production through IL-1beta CXCR4 antagonism increases T cell trafficking in the central nervous system and improves survival from West Nile virus encephalitis Cerebral microglia recruit monocytes into the brain in response to tumor necrosis factoralpha signaling during peripheral organ inflammation Mechanisms of mononuclear phagocyte recruitment in Alzheimer's disease Distinct and Non-Redundant Roles of Microglia and Myeloid Subsets in Mouse Models of Alzheimer's Disease Resistance to experimental autoimmune encephalomyelitis in mice lacking the CC chemokine receptor (CCR)2 Production of CCL2 by central nervous system cells regulates development of murine experimental autoimmune encephalomyelitis through the recruitment of TNF-and iNOS-expressing macrophages and myeloid dendritic cells The role of chemokines during herpes simplex virus-1 infection Microglial and astrocyte chemokines regulate monocyte migration through the blood-brain barrier in human immunodeficiency virus-1 encephalitis Microglial cells initiate vigorous yet non-protective immune responses during HSV-1 brain infection Monocytes regulate T cell migration through the glia limitans during acute viral encephalitis Differential roles of CCL2 and CCR2 in host defense to coronavirus infection The pathogenesis of murine coronavirus infection of the central nervous system CCR2 regulates development of Theiler's murine encephalomyelitis virusinduced demyelinating disease Prevention of experimental autoimmune encephalomyelitis by antibodies against alpha 4 beta 1 integrin Both anti-CD11a (LFA-1) and anti-CD11b (MAC-1) therapy delay the onset and diminish the severity of experimental autoimmune encephalomyelitis The dual role of the neuroinflammatory response after ischemic stroke: modulatory effects of hypothermia An analysis of HIV-1-associated inflammatory products in brain tissue of humans and SCID mice with HIV-1 encephalitis Evidence for deficiencies in intracerebral cytokine production, adhesion molecule induction, and T cell recruitment in herpes simplex virus type-2 infected mice Role of adhesion molecules and inflammation in Venezuelan equine encephalitis virus infected mouse brain Understanding the molecular mechanism of blood-brain barrier damage in an experimental model of Japanese encephalitis: correlation with minocycline administration as a therapeutic agent A major role for VCAM-1, but not ICAM-1, in early atherosclerosis Alpha4beta1 integrin (VLA-4) blockade attenuates both early and late leukocyte recruitment and neointimal growth following carotid injury in apolipoprotein E (−/−) mice Role of vascular cell adhesion molecule-1 and fibronectin connecting segment-1 in monocyte rolling and adhesion on early atherosclerotic lesions Rapid recruitment of inflammatory monocytes is independent of neutrophil migration Timing and duration of anti-alpha4beta1 integrin treatment after spinal cord injury: effect on therapeutic efficacy Atherosclerosis and inflammation mononuclear cell recruitment and adhesion molecules with reference to the implication of ICAM-1/LFA-1 pathway in atherogenesis Inhibition of alpha4 integrin and ICAM-1 markedly attenuate macrophage homing to atherosclerotic plaques in ApoE-deficient mice Microglia derive from progenitors, originating from the yolk sac, and which proliferate in the brain Development and heterogeneity of macrophages and their related cells through their differentiation pathways Fate mapping analysis reveals that adult microglia derive from primitive macrophages ter Meulen V: Isolation and direct characterization of resident microglial cells from the normal and inflamed central nervous system Microglia are activated to become competent antigen presenting and effector cells in the inflammatory environment of the Theiler's virus model of multiple sclerosis Bone marrow-derived microglia play a critical role in restricting senile plaque formation in Alzheimer's disease Bone-marrow-derived cells contribute to the recruitment of microglial cells in response to beta-amyloid deposition in APP/PS1 double transgenic Alzheimer mice Experimental autoimmune encephalomyelitis repressed by microglial paralysis Bone marrow-derived microglia contribute to the neuroinflammatory response and express iNOS in the MPTP mouse model of Parkinson's disease Aguzzi A: Early and rapid engraftment of bone marrow-derived microglia in scrapie Circulating monocytes engraft in the brain, differentiate into microglia and contribute to the pathology following meningitis in mice Invasion of hematopoietic cells into the brain of amyloid precursor protein transgenic mice Ccr2 deficiency impairs microglial accumulation and accelerates progression of Alzheimer-like disease Microglia in the adult brain arise from Ly-6C(hi)CCR2(+) monocytes only under defined host conditions Infiltrating monocytes trigger EAE progression, but do not contribute to the resident microglia pool Local self-renewal can sustain CNS microglia maintenance and function throughout adult life Recruitment of Gr-1+ monocytes is essential for control of acute toxoplasmosis Inflammatory monocytes but not neutrophils are necessary to control infection with Toxoplasma gondii in mice TNF/iNOSproducing dendritic cells mediate innate immune defense against bacterial infection CCR2 mediates conventional dendritic cell recruitment and the formation of bronchovascular mononuclear cell infiltrates in the lungs of mice infected with Cryptococcus neoformans CCR2 expression determines T1 versus T2 polarization during pulmonary Cryptococcus neoformans infection Distinct CCR2(+) Gr1(+) cells control growth of the Yersinia pestis DeltayopM mutant in liver and spleen during systemic plague Recruited inflammatory monocytes stimulate antiviral Th1 immunity in infected tissue MyD88 is required for protection from lethal infection with a mouseadapted SARS-CoV Theiler's virus infection of primary cultures of bone marrow-derived monocytes/macrophages The predominant virus antigen burden is present in macrophages in Theiler's murine encephalomyelitis virus-induced demyelinating disease Monocytes/ macrophages isolated from the mouse central nervous system contain infectious Theiler's murine encephalomyelitis virus (TMEV) Observations on demyelinating lesions induced by Theiler's virus in CBA mice Identification of Theiler's virus infected cells in the central nervous system of the mouse during demyelinating disease Human immunodeficiency virus type 1 infection increases the in vivo capacity of peripheral monocytes to cross the blood-brain barrier into the brain and the in vivo sensitivity of the blood-brain barrier to disruption by lipopolysaccharide The Ly-6Chigh monocyte subpopulation transports Listeria monocytogenes into the brain during systemic infection of mice IFN-gamma triggers CCR2-independent monocyte entry into the brain during systemic infection by virulent Listeria monocytogenes Evidence of a role for monocytes in dissemination and brain invasion by Cryptococcus neoformans CD11c-and CD11b-expressing mouse leukocytes transport single Toxoplasma gondii tachyzoites to the brain The role of CC chemokine receptor 2 on microglia activation and blood-borne cell recruitment after transient focal cerebral ischemia in mice Effects of monocyte chemoattractant protein 1 on blood-borne cell recruitment after transient focal cerebral ischemia in mice Role of CCL2 (MCP-1) in traumatic brain injury (TBI): evidence from severe TBI patients and CCL2−/− mice CC chemokine receptor 2 is critical for induction of experimental autoimmune encephalomyelitis Acute and relapsing experimental autoimmune encephalomyelitis are regulated by differential expression of the CC chemokines macrophage inflammatory protein-1alpha and monocyte chemotactic Inflammatory monocytes and the pathogenesis of viral encephalitis The authors cited in this review were supported by the National Health and Medical Research Council grants 512413 and 1007757 to NJCK and ILC. RLT