key: cord-0811139-xago1ts3 authors: Zhou, Ya-Qun; Liu, Dai-Qiang; Chen, Shu-Ping; Sun, Jia; Zhou, Xue-Rong; Xing, Cui; Ye, Da-Wei; Tian, Yu-Ke title: The Role of CXCR3 in Neurological Diseases date: 2019-02-03 journal: Curr Neuropharmacol DOI: 10.2174/1570159x15666171109161140 sha: 0c92f5b237572a3461ae2205a62ba7622c07a6ab doc_id: 811139 cord_uid: xago1ts3 BACKGROUND: Neurological diseases have become an obvious challenge due to insufficient therapeutic intervention. Therefore, novel drugs for various neurological disorders are in desperate need. Recently, compelling evidence has demon-strated that chemokine receptor CXCR3, which is a G protein-coupled receptor in the CXC chemokine receptor family, may play a pivotal role in the development of neurological diseases. The aim of this review is to provide evidence for the potential of CXCR3 as a therapeutic target for neurological diseases. METHODS: English journal articles that focused on the invovlement of CXCR3 in neurological diseases were searched via PubMed up to May 2017. Moreover, reference lists from identified articles were included for overviews. RESULTS: The expression level of CXCR3 in T cells was significantly elevated in several neurological diseases, including multiple sclerosis (MS), glioma, Alzheimer’s disease (AD), chronic pain, human T-lymphotropic virus type 1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) and bipolar disorder. CXCR3 antagonists showed therapeutic effects in these neurological diseases. CONCLUSION: These studies provided hard evidence that CXCR3 plays a vital role in the pathogenesis of MS, glioma, AD, chronic pain, HAM/TSP and bipolar disorder. CXCR3 is a crucial molecule in neuroinflammatory and neurodegenerative diseases. It regulates the activation of infiltrating cells and resident immune cells. However, the exact functions of CXCR3 in neurological diseases are inconclusive. Thus, it is important to understand the topic of chemokines and the scope of their ac-tivity in neurological diseases. With the substantial prolongation of average life expectancy, the prevalence of neurological diseases increase markedly in the past decades, leading them to become a major public health problem [1, 2] . Unfortunately, few drugs are currently available to cure these diseases. Thus, new drugs for various neurological disorders are in desperate need. Chemokines are important modulators of neuroinflammation and neurodegenerative processes [3, 4] . These small proteins are important in the activation and migration *Address correspondence to these authors at the Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Tel: 00862783663409; Fax: 00862783662853; E-mail: dy0711@gmail.com and Department of Obstetrics & Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Tel: 00862783663215; Fax: 00862783663215; E-mail: dellaxing@163.com of immune cells to lesion sites [5] . The chemokine receptor CXCR3, which belongs to the CXC chemokine receptor family, is a G protein-coupled receptor that plays a vital role in mediating chemotactic migration, cell proliferation, and survival [6] [7] [8] . CXCR3 is mainly expressed by various effector T lymphocytes including CD4 + Th1 cells, CD8 + cytotoxic T cells, and natural killer (NK) cells [9, 10]. The principle chemokine ligands of CXCR3 are CXCL4, CXCL9, CXCL10 and CXCL11 [11] [12] [13] [14] . CXCL4, a plateletderived CXCR3 ligand, is weakly chemotactic for neutrophils, monocytes and fibroblasts [15] . Mainly induced by interferon gamma (IFN-γ), CXCL9, CXCL10 and CXCL11 are potent chemoattractants for monocytes, T cells, NK cells and dendritic cells. Although they share the same chemokine receptor CXCR3, these three ligands are regulated by unique promoters and exhibit distinct temporal and spatial expression patterns ( Table 1) . Recent studies have shown that the chemokine receptor CXCR3 was expressed in central nerv-ous system (CNS) diseases and involved in their pathogenesis [16] [17] [18] [19] [20] . It was shown that the expression levels of CXCR3 and its ligands were elevated in the periphery blood and cerebrospinal fluid (CSF) of patients with neurological diseases [21] [22] [23] , and were correlated with prognosis in certain cases [24] . Additionally, treatment with CXCR3 antagonists could alleviate bone cancer pain (BCP) induced mechanical allodynia [25] . These studies suggested an important role of CXCR3 in neurological diseases, indicating that targeting CXCR3 may reveal novel therapeutic interventions for the management of neurological diseases. Thus, here we summarize the current evidence supporting a role of CXCR3 in the pathogenesis of neurological diseases, including multiple sclerosis, glioma, Alzheimer's disease, chronic pain and human T-lymphotropic virus type 1-associated myelopathy. Multiple sclerosis (MS), also known as disseminated sclerosis, is a chronic inflammatory disease of the CNS which is characterized by destruction of myelin and oligodendrocytes in the brain and spinal cord [35, 36] . Mounting evidence has suggested that CXCR3 plays a vital part in MS. Balashov et al. [21] first reported that CXCR3 positive T cells were increased in blood of relapsing/remitting and progressive MS compared with controls. They also found that CXCL10, one of the CXCR3 ligands, was expressed by astrocytes in MS brain lesions but not unaffected white matter of control or MS subjects. These results suggested that CXCL10/CXCR3 expression level may be used for immunologic staging of MS and provided a rationale for the use of agents blocking CXCR3 as a therapeutic approach in the treatment of MS. Using immunocytochemistry, Simpson et al. [37] confirmed the expression of CXCL9 and CXCL10, and their receptor CXCR3 in post-mortem CNS tissue from MS cases at different stages of lesion development. Their results showed that both macrophages and astrocytes were active in demyelinating lesions predominantly expressed CXCL9 and CXCL10, and CXCR3 was expressed by T cells and by astrocytes within the plaque. The differential expression of chemokines indicated that blocking chemokine receptors may serve as an anti-inflammatory therapy for MS. On the other hand, CXCL10 and CXCR3 were significantly increased in the CSF of patients with MS compared with controls [38] [39] [40] . Moreover, the increased level of CXCL10 was associated with clinical relapses in MS. Compared with secondary progressive MS, the concentration of CXCL10 was significantly greater in patients with relapsing/remitting, which was correlated significantly with CXCR3 expression on CSF CD4 + T cells from patients with MS. In another study, Sindern et al. [41] demonstrated that the increased level of CXCR3 positive T-cells in the CSF was strongly associated with active MRI lesion appearance in patients with relapsing/remitting MS, which might be the result of migration of activated T-cells from the circulation into the CSF. Consistent with previous reports, this study confirmed the hypothesis that CXCR3 might be involved in the development of acute MS lesions, leading to therapeutic intervention via blocking CXCR3. By analyzing the expression of CXCR3 on peripheral lymphocytes in 18 MS patients, Mahad et al. [42] found that the increased expression of CXCR3 on peripheral blood CD4 + lymphocytes was associated with all relapses and that the fluctuations of CXCR3 expression was significantly greater in patients with MS than controls. This study provided further evidence for the potential therapeutic value of CXCR3 antagonists. The therapeutic effect of IFN-β on patients with MS is well established [43] [44] [45] . After treatment with IFN-β for three months, Sorensen et al. [46] found that the expression of CXCR3 on CD4 + and CD8 + T cells was significantly reduced, whereas the expression of other receptors (e.g. CCR1, CCR2, CCR3, CCR5) was unaltered. This results indicated that IFN-β may exert its therapeutic effect on MS patients by suppressing the expression of CXCR3. Experimental autoimmune encephalomyelitis (EAE), a rodent model of human MS, is a CD4 + Th1 cell-and Th17 cell-mediated demyelinating disease of the CNS [47] . The role of CXCR3 in EAE has been widely studied. Considering that CXCR3 and its ligands are elevated in MS patients, it was proposed that blocking CXCR3 or neutralization of its ligands would inhibit the development of EAE. However, Narumi et al. [48] reported that EAE rats treated with monoclonal antibody (mAb) against CXCL10 exacerbated the disease scores with less enlarged draining lymph nodes than treated with control mAb. The smaller draining lymph nodes in EAE rats treated with anti-CXCL10 mAb might be explained by the following mechanism: neutralization of CXCL10 causes an increased release of Th1 cells from lymph nodes, which results in increased migration to the CNS where CXCL11, another ligand for CXCR3, is induced as well. Moreover, CXCL10-deficient mice exhibited a reduced threshold for EAE induction and developed severe EAE after immunization with low doses of myelin oligodendroglial glycoprotein (MOG)p33-55 that produced minimal disease in wild-type littermates [49] . In another study, Muller et al. examined the function of CXCR3 signaling in EAE using CXCR3 deficient (CXCR3 _ / _ ) mice [50, 51] . No significant difference was found in terms of time to onset and peak disease severity in CXCR3 _ / _ and wild-type (WT) mice. However, CXCR3 _ / _ mice had more severe chronic disease with increased demyelination and axonal damage. Additionally, the inflammatory lesions were more widespread throughout the CNS in CXCR3 _ / _ mice than WT mice in which inflammatory lesions consisted of well-demarcated perivascular mononuclear cell infiltrates. Furthermore, Foxp3 + regulatory T cells were significantly reduced in number and were scattered in the spinal cord of CXCR3 _ / _ mice. These results suggested that CXCR3 signaling plays a major protective role in EAE by constraining CD4 + T cells to the perivascular space in the CNS, promoting regulatory T cell accumulation and facilitating interaction of these cells with effector T cells, thus limiting autoimmune-mediated tissue damage. In a mouse model of neurotropic coronavirusinduced encephalomyelitis, Marques et al. [52] found that the accumulation of antibody secreting cells (ASC) in the CNS was mediated by CXCR3. In CXCR3 _ / _ mice, both the total and virus-specific ASC were reduced greater than 80%. Furthermore, neither virus-specific ASC trafficking to bone marrow nor antiviral serum antibody was reduced relative to levels in control mice. Additionally, infected CXCR3 _ / _ mice showed elevated levels of persisting viral RNA, sustained infectious virus, increased clinical disease and mortality. These results indicated that CXCR3 played a vital role in the recruitment of activated ASC into the inflamed CNS and highlighted its protective role during persistent infection. A further study demonstrated that CXCL10 is critical for the recruitment of ASC to the CNS vasculature and ASC entry into the CNS parenchyma in a mouse model of viral encephalomyelitis [53] . In this study, they found reduced CNS IgG and κ-light chain mRNA and virus-specific Ab as well as impaired ASC recruitment in CXCL10 _ / _ rather than CXCL9 _ / _ mice. Moreover, the ASC recruited to the CNS in CXCL10 _ / _ mice restricted to the vasculature, whereas it was localized in the parenchyma in wild-type and CXCL9 _ / _ mice. Recently, the CXCR3-CXCL10 axis was also demonstrated to have a role in recruiting pathogenic T lymphocytes in the brains of patients with Rasmussen encephalitis [54] . Using surgical specimens of children with Rasmussen encephalitis, they found that CXCR3 was expressed in cytotoxic T lymphocytes infiltrating the damaged areas of primary biopsies, whereas CXCL10 was expressed in neurons and astrocytes in the same areas. Additionally, in vitro study demonstrated that astrocytes upregulated the mRNA expression level of CXCL10 and the release of CXCL10 to the supernatants in response to infections, which was completely abolished by CXCR3 antagonist. These results suggested that astrocytes and neurons may recruit pathogenic T lymphocytes into areas of the brain of Rasmussen encephalitis through the CXCL10-CXCR3 axis. To conclude, the clinical studies provided compelling evidence that CXCR3 is involved in the development of MS and its antagonists may become novel agents for the management of MS. However, further study with larger sample size and longer follow-up are warranted to investigate the feasibility of CXCR3 antagonists and its exact mechanism in alleviating MS. The animal studies also proposed a protective role of CXCR3 in EAE and may provide novel insights into intervention strategies. However, the underlying mechanisms of how CXCR3 exerts its protective role in EAE need further investigations. Gliomas account for the majority of neoplasms occurring in the CNS, and are one of the most aggressive types of human cancer [55, 56] . It was reported CXCR3 regulates cell invasion, migration and facilitates tumor metastasis to lymph nodes [57, 58] . Furthermore, the expression level of CXCR3 correlates with prognosis of breast cancer [59] , colorectal cancer [60, 61] and renal cell cancer [62] [63] [64] . Maru et al. [65] first reported that CXCR3 and CXCL10 were increased in glioma cells compared with adult human astrocytes. Moreover, they found that the expression level of CXCR3 correlated with malignancy grade of glioma. Glioblastoma multiforme (GBM) is the most aggressive form of human glioma with mean survival approximately 12 months despite intensive and comprehensive treatment [66] . Liu et al. [67] examined the role of CXCR3 in GBM progression using the GL261 murine model of malignant glioma. They found that Murine glioma GL261 cells express CXCL10 in vitro and GL261 tumors express CXCL9 and CXCL10 in vivo. CXCR3 _ / _ mice with glioma had significantly shorter median survival time and reduced numbers of tumor-infiltrated natural killer (NK) and natural killer T (NKT) cells in contrast with WT glioma mice. However, treatment with CXCR3 antagonist NBI-74330 suppressed GL261 tumor growth and increased median survival times of both tumor-bearing WT and CXCR3 _ / _ mice compared with vehicle-treated groups, suggesting that CXCR3 may have an inhibitory effect directly on the tumor cells. Moreover, NBI-74330 had no im-pact on tumor-infiltrated NK and NKT cells, which known to express CXCR3, suggesting that CXCR3 is not the primary means by which NK and NKT cells traffic into glioma. Thus, the absence of tumor-infiltrating NK and NKT cells rather than CXCR3 may account for the shorter survival time of tumor-bearing CXCR3 _ / _ mice. Moreover, both CXCR3 and its ligands are expressed by murine and human glioma cell lines (A172, T98G, U87, U118 and U138). These results suggested that CXCR3 system may be a unique target for human GBM therapy. Recently, Pu et al. [24] investigated the potential prognostic value of CXCR3 in primary GBM and its relationship with the clinicopathological features. Using Kaplan-Meier survival curve analysis with a log-rank comparison of the 65 primary GBM patients, they found that the patients with higher expression levels of CXCR3 had shorter progression free survival and overall survival compared with those with lower expression levels of CXCR3. Using univariate Cox regression analysis, they found that high CXCR3 expression was a risk factor for primary GBM [P < 0.01, hazard ratio (HR) 2.336, 95 % confidence interval (CI) 1.341-4.071]. Furthermore, their multivariate Cox regression analysis showed that CXCR3 expression level was an independent prognostic factor for the overall survival of primary GBM patients. Taken together, these results suggested that CXCR3 might be a useful independent prognostic biomarker for primary GBM patients. Alzheimer's disease (AD) is one of the most prevalent progressive neurodegenerative brain disorders worldwide characterized by loss of neurons and synapses in the cerebral cortex and certain subcortical regions [68, 69] . Chemokines and their receptors were found to be associated with AD pathological changes [70] [71] [72] [73] . Using post-mortem human tissues of AD patients, Xia et al. [72] provided the first immunohistochemical evidence that CXCR3 was constitutively expressed on a subpopulation of neurons and neuronal processes in the neocortex, hippocampal formation, striatum, cerebellum and spinal cord. Moreover, they found greatly upregulated CXCL10 positive astrocytes in AD patients compare with controls. Additionally, CXCL10 positive astrocytes were frequently associated with amyloid deposits, suggesting an active chronic inflammatory response related to amyloid deposits occurs in AD patients. Recently, Krauthausen et al. [22] examined the impact of CXCR3 in the amyloid precursor protein (APP)/presenilin 1 (PS1) transgenic mouse model of AD. They found that amyloid beta (Aβ) deposition and Aβ levels were significantly decreased in CXCR3 _ / _ APP/PS1 mice compared with control APP/PS1 mice. Their in vitro and in vivo analysis of microglial phagocytosis showed that CXCR3 deficiency facilitated the microglial uptake of Aβ. Moreover, preincubation of CXCR3 antagonist increased primary microglial Aβ phagocytosis and reduced TNF-α secretion. Compared with control APP/PS1 mice, brain tissue from CXCR3 _ / _ APP/PS1 mice had reduced concentrations of proinflmmatory cytokines and the microglia exhibited obvious morphological activation and reduced plaque association. Furthermore, Morris watermaze testing showed improved spatial memory of CXCR3 _ / _ APP/PS1 mice compared with controls, suggesting that lack of CXCR3 attenuated the behavioral deficits of APP/PS1 mice. Taken together, these results indicated that CXCR3 signaling mediated development of AD-like pathology in APP/PS1 mice and suggested that CXCR3 had the potential to be a therapeutic target for AD. Furthermore, a very recent study reported that peripheral blood mononuclear cells of Alzheimer's disease patients control CCL4 and CXCL10 levels in a human blood brain barrier model. These studies indicated a pivotal role of CXCR3 in the pathogenesis of AD. However, further studies are warranted to explore the underlying mechanisms. Pain plays a vital role in protecting us against damaging stimuli [74] [75] [76] . However, chronic pain presents a major challenge due to its complex natural history, unclear etiology, and poor response to therapy [77] [78] [79] . There is compelling evidence supporting the involvement of neuroinflammation in chronic pain [80] [81] [82] [83] [84] . Our lab has been investigating the mechanisms of chronic pain for decades [85] [86] [87] [88] [89] [90] [91] . Previously, we have shown the involvement of CXCL10/CXCR3 signaling in BCP rat models [23, 92] . After intra-tibial inoculation of Walker 256 mammary gland carcinoma cells, the BCP model was established, indicated by downregulation of paw withdrawal threshold (PWT) and bone destruction. Our real-time polymerase chain reaction (PCR) and immunohistochemical analyses showed that both CXCL10 and its receptor CXCR3 were up-regulated in the spinal cord of BCP rats. Blocking the CXCL10/CXCR3 signaling via anti-CXCL10 antibody or CXCR3 antagonist AMG487 prevented the development of BCP and microglial activation. Moreover, inhibiting microglial activation attenuated CXCL10 upregulation in BCP rats. These results suggested that CXCL10/CXCR3 signaling participated in BCP via activation of microglia in rat models. Our further study revealed the cellular mechanism of how CXCR3 mediated BCP [25] . We confirmed that CXCR3 was significantly increased in the spinal cord of BCP rats and co-localized with either neurons, microglia, and astrocytes in the spinal cord, or non-peptidergic-, peptidergic-, and A-type neurons in the dorsal root ganglion (DRG). Moreover, spinal phosphorylation of Akt and extracellular signal-regulated kinase (ERK1/2) were markedly upregulated in BCP rats in a timedependent manner. Meanwhile, CXCR3 was co-localized with either pAkt or pERK1/2. Blockage of either Akt or ERK1/2 attenuated the mechanical allodynia in BCP rats. Furthermore, CXCR3 antagonist AMG487 suppressed the upregulation of pAkt and pERK1/2. Taken together, these results indicated that the activation of spinal chemokine receptor CXCR3 mediated BCP through Akt-ERK pathway. A very recent study performed by Jiang et al. confirmed the pivotal role of CXCR3 in neuropathic pain [93] . They found that the expression of CXCR3 was significantly upregulated mainly in the spinal neuron following spinal nerve ligation (SNL). Moreover, CXCL10, a ligand of CXCR3, was also considerably increased in the spinal neurons and astrocytes in SNL mice. Additionally, inhibiting the expression of CXCR3 by CXCR3-/-mice and shRNA targeting the sequence of mice Cxcr3 as well as CXCR3 antagonist NBI-74330 attenuated SNL-induced mechanical allodynia and thermal hyperalgesia. More importantly, they revealed the epigenetic mechanism of how CXCR3 contributed to neuro-pathic pain. These results suggested that targeting CXCR3 may alleviated the established choric pain and novel CXCR3 antagonist with fewer side effects should be developed. Human T-lymphotropic virus type 1 (HTLV-1)-associated myelopathy/tropical spastic paraparesis (HAM/TSP) is a rare neurodegenerative disease characterized by chronic inflammation in the spinal cord [94, 95] . Recently, Ando et al. [96] investigated the possible role of CXCL10/CXCR3 signaling in chronic inflammation of HAM/TSP. They found that the expression level of CXCL10 was markedly high in the CSF of HAM/TSP patients and a great deal of CSF cells expressed the CXCL10-binding receptor CXCR3, which mainly consist of CD3 + cells (>90%) and small populations of CD14 + and CD19 + cells. Their immunofluorescence results showed that astrocytes were the major producers of CXCL10 in the spinal cords of HAM/TSP patients. Coculture of human astrocytoma cells with CD4 + T cells from HAM/TSP patients revealed that astrocytes produce CXCL10 in response to IFN-γ secreted by CD4 + T cells. Moreover, the chemotaxis assays results suggested that CXCL10 induced migration of peripheral blood mononuclear cells to the CNS and that anti-CXCL10 neutralizing antibody could disrupt this migration. These results demonstrated that HTLV-1 infected cells in the CNS produced IFN-γ that induces astrocytes to secrete CXCL10, which recruits more infected cells to the area via CXCR3, constituting a T helper type 1-centric positive feedback loop that results in chronic inflammation. Bipolar disorder (BD) is a severe disorder with high prevalence that affect mood and cognitive functions, and its pathophysiology remain largely unknown [97] . Emerging lines of evidence has shown an association of BD with the immune system [98] [99] [100] [101] . Brietzke et al. [102] first examined serum chemokine levels in 30 BD patients and to compare these results with those obtained with 30 healthy subjects. They found that the serum levels of CXCL10 were increased and CCL24 levels were decreased in BD patients in contrast with controls, suggesting an association between BD and changes in chemokine level. It is worth mentioning that CCL24 is an eotaxin, which not only binds CCR3 and recruit eosinophils and Th2 cells, but also act as an antagonist of CXCR3 [103, 104] . Barbosa et al. [17] further investigated the plasma levels of chemokines in 70 BD patients in different neuropsychiatry states (35 in euthymia and 35 in mania) compared with 50 healthy controls. Their results showed that BD patients had increased plasma levels of CXCL11, CCL24, CXCL10, and decreased plasma levels of CXCL8 in contrast with healthy controls. Moreover, these chemokines levels were not significantly different between BD patients in euthymia and in mania. Furthermore, the logistic regression stressed the main effect of increased plasma levels of CXCL10 and decreased plasma levels of CXCL8 to BD. These studies indicate that CXCL10, CXCL11 and their receptor CXCR3 should be further investigated with regard to their potential role as longstanding markers of BD. By reviewing the current evidence, we discussed the possible involvement of CXCR3 in neurological diseases. These studies provided hard evidence that CXCR3 plays a vital role in the pathogenesis of MS, glioma, AD, chronic pain, HAM/TSP and bipolar disorder. CXCR3 is a crucial molecule in neuroinflammatory and neurodegenerative diseases. It regulates the activation of infiltrating cells and resident immune cells. However, the exact functions of CXCR3 in neurological diseases are inconclusive. Thus, it is important to understand the topic of chemokines and the scope of their activity in neurological diseases. Further studies focused on chemokines and neurological diseases may lead to the discovery of novel therapeutic strategies for patients suffering from various neurological disorders connected with neuroinflammation. It is worth mentioning that the caution must be taken when considering CXCR3 as therapeutic target since it can mediate both beneficial and harmful effects during the course of CNS diseases. Not applicable. The authors declare no conflict of interest, financial or otherwise. A consensus review on the development of palliative care for patients with chronic and progressive neurological disease Lumbar transplantation of immortalized enkephalin-expressing astrocytes attenuates chronic neuropathic pain Cytokines and chemokines at the crossroads of neuroinflammation, neurodegeneration, and neuropathic pain Anti-inflammatory mechanism of bone marrow mesenchymal stem cell transplantation in rat model of spinal cord injury Chemokines and chemokine receptors: positioning cells for host defense and immunity A potential contribution of chemokine network dysfunction to the depressive disorders CXCR3 ligands in disease and therapy CXCR3 promotes plaque formation and behavioral deficits in an Alzheimer's disease model Spinal IFN-γinduced protein-10 (CXCL10) mediates metastatic breast cancerinduced bone pain by activation of microglia in rat models High expression of CXCR3 is an independent prognostic factor in glioblastoma patients that promotes an invasive phenotype Activation of spinal chemokine receptor CXCR3 mediates bone cancer pain through an Akt-ERK crosstalk pathway in rats Lymphocyte-specific chemokine receptor CXCR3: regulation, chemokine binding and gene localization CXCL4-induced migration of activated T lymphocytes is mediated by the chemokine receptor CXCR3 Platelets release CXCL4L1, a nonallelic variant of the chemokine platelet factor-4/CXCL4 and potent inhibitor of angiogenesis The role of the CXC chemokines platelet factor-4 (CXCL4/PF-4) and its variant (CXCL4L1/PF-4var) in inflammation IFN-alpha2a induces IP-10/CXCL10 and MIG/CXCL9 production in monocyte-derived dendritic cells and enhances their capacity to attract and stimulate CD8+ effector T cells Mig and IP-10: CXC chemokines that target lymphocytes CXCR3 and heparin binding sites of the chemokine IP-10 (CXCL10) Intracellular domains of CXCR3 that mediate CXCL9, CXCL10, and CXCL11 function CXCL11-dependent induction of FOXP3-negative regulatory T cells suppresses autoimmune encephalomyelitis Pathogenesis of multiple sclerosis: insights from molecular and metabolic imaging Natural killer cells and their receptors in multiple sclerosis Expression of the interferon-gamma-inducible chemokines IP-10 and Mig and their receptor, CXCR3, in multiple sclerosis lesions Expression of chemokines in the CSF and correlation with clinical disease activity in patients with multiple sclerosis Multiple sclerosis and optic neuritis: CCR5 and CXCR3 expressing T cells are augmented in blood and cerebrospinal fluid Multiple sclerosis: a study of CXCL10 and CXCR3 co-localization in the inflamed central nervous system Expression of chemokine receptor CXCR3 on cerebrospinal fluid T-cells is related to active MRI lesion appearance in patients with relapsing-remitting multiple sclerosis Longitudinal study of chemokine receptor expression on peripheral lymphocytes in multiple sclerosis: CXCR3 upregulation is associated with relapse Interferon β-1b for the treatment of primary progressive multiple sclerosis: five-year clinical trial follow-up Magnetic resonance imaging reveals therapeutic effects of interferon-beta on cytokine-induced reactivation of rat model of multiple sclerosis Daclizumab HYP versus Interferon Beta-1a in Relapsing Multiple Sclerosis Selective suppression of chemokine receptor CXCR3 expression by interferon-beta1a in multiple sclerosis Experimental autoimmune encephalomyelitis (EAE) as a model for multiple sclerosis (MS) Neutralization of IFN-inducible protein 10/CXCL10 exacerbates experimental autoimmune encephalomyelitis IFN-inducible protein 10/CXC chemokine ligand 10-independent induction of experimental autoimmune encephalomyelitis Campbell, I.L. CXCR3 signaling reduces the severity of experimental autoimmune encephalomyelitis by controlling the parenchymal distribution of effector and regulatory T cells in the central nervous system Severe disease, unaltered leukocyte migration, and reduced IFN-gamma production in CXCR3-/-mice with experimental autoimmune encephalomyelitis CXCR3-dependent plasma blast migration to the central nervous system during viral encephalomyelitis Astrocyte-derived CXCL10 drives accumulation of antibody-secreting cells in the central nervous system during viral encephalomyelitis A role for the CXCR3/CXCL10 axis in Rasmussen encephalitis CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in Isoflurane enhances the malignant potential of glioblastoma stem cells by promoting their viability, mobility in vitro and migratory capacity in vivo Pivotal role of CXCR3 in melanoma cell metastasis to lymph nodes CXCR3 as a molecular target in breast cancer metastasis: inhibition of tumor cell migration and promotion of host anti-tumor immunity CXCR3 expression is associated with poor survival in breast cancer and promotes metastasis in a murine model The prognostic significance of chemokine receptor CXCR3 expression in colorectal carcinoma The expression of chemokine receptors CXCR3 and CXCR4 in predicting postoperative tumour progression in stages I-II colon cancer: a retrospective study Predicting disease progression after nephrectomy for localized renal cell carcinoma: the utility of prognostic models and molecular biomarkers The chemokine receptor CXCR3 is an independent prognostic factor in patients with localized clear cell renal cell carcinoma Molecular signatures of localized clear cell renal cell carcinoma to predict disease-free survival after nephrectomy Chemokine production and chemokine receptor expression by human glioma cells: role of CXCL10 in tumour cell proliferation Comprehensive portrait of recurrent glioblastoma multiforme in molecular and clinical characteristics Chemokine receptor CXCR3 promotes growth of glioma Innate immunity in Alzheimer's disease Neuroinflammation in Alzheimer's disease Chemokines/chemokine receptors in the central nervous system and Alzheimer's disease Chemokines in CSF of Alzheimer's disease patients. Arq. Neuropsiquiatr Expression of the chemokine receptor CXCR3 on neurons and the elevated expression of its ligand IP-10 in reactive astrocytes: in vitro ERK1/2 activation and role in Alzheimer's disease Neuroinflammation in Alzheimer's disease: chemokines produced by astrocytes and chemokine receptors Microglia and intractable chronic pain Activation of PI3Kγ/Akt pathway mediates bone cancer pain in rats Minocycline attenuates bone cancer pain in rats by inhibiting NF-κB in spinal astrocytes Intensity of chronic pain--the wrong metric? An improved teton system for gene expression in neurons delivered by a single lentiviral vector Prevention of neuropathic pain in an animal model of spare nerve injury following oral immunization with recombinant adenovirus serotype 5-mediated NR2B gene transfer Melatonin modulates neuroinflammation and oxidative stress in experimental diabetic neuropathy: effects on NF-κB and Nrf2 cascades SOCS3-mediated blockade of JAK/STAT3 signaling pathway reveals its major contribution to spinal cord neuroinflammation and mechanical allodynia after peripheral nerve injury Emerging targets in neuroinflammation-driven chronic pain Neuroinflammation and the generation of neuropathic pain Chemokines and their receptors: potential therapeutic targets for bone cancer pain Interleukin-6: an emerging regulator of pathological pain Targeting glia for bone cancer pain The role of spinal GABAB receptors in cancerinduced bone pain in rats MHC-I promotes apoptosis of GABAergic interneurons in the spinal dorsal horn and contributes to cancer induced bone pain STAT1 as a downstream mediator of ERK signaling contributes to bone cancer pain by regulating MHC II expression in spinal microglia Slit2/Robo1 mediation of synaptic plasticity contributes to bone cancer pain PI3K/Akt pathway: a potential therapeutic target for chronic pain Activation of CXCL10/CXCR3 signaling attenuates morphine analgesia: involvement of Gi protein Promoted interaction of C/EBPα with demethylated Cxcr3 gene promoter contributes to neuropathic pain in mice Epidemiology, treatment, and prevention of human T-cell leukemia virus type 1-associated diseases Anti-rheumatic drug iguratimod (T-614) alleviates cancerinduced bone destruction via down-regulating interleukin-6 production in a nuclear factor-κB-dependent manner Positive feedback loop via astrocytes causes chronic inflammation in virus-associated myelopathy An epidemiologic and clinical overview of medical and psychopathological comorbidities in major psychoses Glia and immune cell signaling in bipolar disorder: insights from neuropharmacology and molecular imaging to clinical application Are medical comorbid conditions of bipolar disorder due to immune dysfunction? Immune dysfunction in bipolar disorder and suicide risk: is there an association between peripheral corticotropin-releasing hormone and interleukin-1β? Bipolar Disord Damage-associated molecular patterns and immune activation in bipolar disorder Abnormalities in serum chemokine levels in euthymic patients with bipolar disorder CCR3 functional responses are regulated by both CXCR3 and its ligands CXCL9, CXCL10 and CXCL11 The ligands of CXC chemokine receptor 3, I-TAC, Mig, and IP10, are natural antagonists for CCR3 This work was supported by grants from National Natural Science Foundation of P.R. China 81400917, 81371250 and 81571053. We are grateful to Amgen for kindly providing AMG487 to us.