Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 442
Filter
1.
Cell Mol Life Sci ; 81(1): 300, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39001897

ABSTRACT

BACKGROUND: Age-associated impairments in innate immunity are believed to be a causative factor responsible for severe pathogenesis of Staphylococcus aureus (S. aureus) infection in the bone tissue. However, the basis for age-associated decline in innate immune response upon S. aureus infection remains poorly understood. RESULTS: Our transcriptional data (GEO: GSE166522) from a mouse model of S. aureus osteomyelitis show up-regulated CXCL9 and CXCL10 (CXCL9/10), which is further confirmed in vitro and in vivo by the present study. Notably, monocytes are a main source for CXCL9/10 production in bone marrow upon S. aureus challenge, but this response declines in middle-aged mice. Interestingly, conditional medium of bone marrow monocytes from middle-aged mice has a strikingly decreased effect on bactericidal functions of neutrophils and macrophages compares with that from young mice. We further show that activation of CXCL9/10-CXCR3 axis between monocytes and macrophages/neutrophils promotes the bactericidal function of the cells, whereas blocking the axis impairs such function. Importantly, treatment with either exogenous CXCL9 or CXCL10 in a middle-aged mice model enhances, while pharmacological inhibition of CXCR3 in young mice model impairs, bacterial clearance and bone marrow structure. CONCLUSIONS: These findings demonstrate that bone marrow monocytes act as a critical promotor of innate immune response via the CXLCL9/10-CXCR3 axis upon S. aureus infection, and that the increased susceptibility to S. aureus infection in skeleton in an aged host may be largely attributable to the declined induction of CXCR9/10 in monocytes.


Subject(s)
Chemokine CXCL10 , Chemokine CXCL9 , Disease Models, Animal , Immunity, Innate , Monocytes , Osteomyelitis , Staphylococcal Infections , Staphylococcus aureus , Animals , Osteomyelitis/microbiology , Osteomyelitis/immunology , Osteomyelitis/metabolism , Osteomyelitis/pathology , Monocytes/immunology , Monocytes/metabolism , Chemokine CXCL9/metabolism , Chemokine CXCL9/genetics , Staphylococcus aureus/immunology , Mice , Chemokine CXCL10/metabolism , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcal Infections/metabolism , Mice, Inbred C57BL , Receptors, CXCR3/metabolism , Receptors, CXCR3/genetics , Aging/immunology , Neutrophils/immunology , Neutrophils/metabolism , Macrophages/immunology , Macrophages/metabolism
2.
Beijing Da Xue Xue Bao Yi Xue Ban ; 56(3): 512-518, 2024 Jun 18.
Article in Chinese | MEDLINE | ID: mdl-38864138

ABSTRACT

OBJECTIVE: To investigate the characteristics of the CD8+ T cells infiltration from the 4 subtypes in medulloblastoma (MB), to analyze the relationship between CD8+ T cells infiltration and prognosis, to study the function of C-X-C motif chemokine ligand 11 (CXCL11) and its receptor in CD8+ T cells infiltration into tumors and to explore the potential mechanism, and to provide the necessary clinicopathological basis for exploring the immunotherapy of MB. METHODS: In the study, 48 clinical MB samples (12 cases in each of 4 subtypes) were selected from the multiple medical center from 2012 to 2019. The transcriptomics analysis for the tumor of 48 clinical samples was conducted on the NanoString PanCancer IO360TM Panel (NanoString Technologies). Immunohistochemistry (IHC) staining of formalin-fixed, paraffin-embedded sections from MB was carried out using CD8 primary antibody to analyze diffe-rential quantities of CD8+ T cells in the MB four subtypes. Through bioinformatics analysis, the relationship between CD8+T cells infiltration and prognosis of the patients and the expression differences of various chemokines in the different subtypes of MB were investigated. The expression of CXCR3 receptor on the surface of CD8+T cells in MB was verified by double immunofluorescence staining, and the underlying molecular mechanism of CD8+T cells infiltration into the tumor was explored. RESULTS: The characteristic index of CD8+T cells in the WNT subtype of MB was relatively high, suggesting that the number of CD8+T cells in the WNT subtype was significantly higher than that in the other three subtypes, which was confirmed by CD8 immunohistochemical staining and Gene Expression Omnibus (GEO) database analysis by using R2 online data analysis platform. And the increase of CD8+T cells infiltration was positively correlated with the patient survival. The expression level of CXCL11 in the WNT subtype MB was significantly higher than that of the other three subtypes. Immunofluorescence staining showed the presence of CXCL11 receptor, CXCR3, on the surface of CD8+T cells, suggesting that the CD8+T cells might be attracted to the MB microenvironment by CXCL11 through CXCR3. CONCLUSION: The CD8+T cells infiltrate more in the WNT subtype MB than other subtypes. The mechanism may be related to the activation of CXCL11-CXCR3 chemokine system, and the patients with more infiltration of CD8+T cells in tumor have better prognosis. This finding may provide the necessary clinicopathological basis for the regulatory mechanism of CD8+T cells infiltration in MB, and give a new potential therapeutic target for the future immunotherapy of MB.


Subject(s)
CD8-Positive T-Lymphocytes , Chemokine CXCL11 , Medulloblastoma , Receptors, CXCR3 , Humans , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Medulloblastoma/immunology , Medulloblastoma/pathology , Medulloblastoma/classification , Medulloblastoma/genetics , Medulloblastoma/metabolism , Receptors, CXCR3/metabolism , Receptors, CXCR3/genetics , Chemokine CXCL11/metabolism , Chemokine CXCL11/genetics , Prognosis , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Cerebellar Neoplasms/immunology , Cerebellar Neoplasms/pathology , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/classification , Cerebellar Neoplasms/metabolism , Male , Female
3.
Am J Physiol Lung Cell Mol Physiol ; 327(2): L160-L172, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38771132

ABSTRACT

The alveolar type II epithelial cells (AEC2s) act as stem cells in the lung for alveolar epithelial maintenance and repair. Chemokine C-X-C motif chemokine 10 (CXCL10) is expressed in injured tissues, modulating multiple cellular functions. AEC2s, previously reported to release chemokines to recruit leukocytes, were found in our study to secrete CXCL10 after bleomycin injury. We found that Sftpc-Cxcl10 transgenic mice were protected from bleomycin injury. The transgenic mice showed an increase in the AEC2 population in the lung by flow cytometry analysis. Both endogenous and exogenous CXCL10 promoted the colony formation efficiency of AEC2s in a three-dimensional (3-D) organoid growth assay. We identified that the regenerative effect of CXCL10 was CXCR3 independent using Cxcr3-deficient mice, but it was related to the TrkA pathway. Binding experiments showed that CXCL10 interacted with TrkA directly and reversibly. This study demonstrates a previously unidentified AEC2 autocrine signaling of CXCL10 to promote their regeneration and proliferation, probably involving a CXCR3-independent TrkA pathway.NEW & NOTEWORTHY CXCL10 may aid in lung injury recovery by promoting the proliferation of alveolar stem cells and using a distinct regulatory pathway from the classical one.


Subject(s)
Alveolar Epithelial Cells , Chemokine CXCL10 , Receptors, CXCR3 , Animals , Mice , Alveolar Epithelial Cells/metabolism , Cell Proliferation , Chemokine CXCL10/metabolism , Chemokine CXCL10/genetics , Lung Injury/metabolism , Lung Injury/pathology , Mice, Inbred C57BL , Mice, Transgenic , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Receptors, CXCR3/metabolism , Receptors, CXCR3/genetics , Regeneration , Signal Transduction
4.
Theriogenology ; 225: 43-54, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38788628

ABSTRACT

Extensive research has been conducted on the role of CXCR3 in immune responses and inflammation. However, the role of CXCR3 in the reproductive system, particularly in oocyte development, remains unknown. In this study, we present findings on the involvement of CXCR3 in the meiotic division process of mouse oocytes. We found CXCR3 was expressed consistently throughout the entire maturation process of mouse oocyte. Inhibition of CXCR3 impaired the asymmetric division of oocyte, while the injection of Cxcr3 mRNA was capable of restoring these defects. Further study showed that inhibition of CXCR3 perturbed spindle migration by affecting LIMK/cofilin pathway-mediated actin remodeling. Knockout of CXCR3 led to an upregulation of actin-binding protein and an increased ATP level in GV-stage oocytes, while maintaining normal actin dynamics during the process of meiosis. Additionally, we noticed the expression level of DYNLT1 is markedly elevated in CXCR3-null oocytes. DYNLT1 bound with the Arp2/3 complex, and knockdown of DYNLT1 in CXCR3-null oocytes impaired the organization of cytoplasmic actin, suggesting the regulatory role of DYNLT1 in actin organization, and the compensatory expression of DYNLT1 may contribute to maintain normal actin dynamics in CXCR3-knockout oocytes. In summary, our findings provide insights into the intricate network of actin dynamics associated with CXCR3 during oocyte meiosis.


Subject(s)
Actins , Oocytes , Receptors, CXCR3 , Animals , Oocytes/metabolism , Oocytes/physiology , Mice , Actins/metabolism , Actins/genetics , Receptors, CXCR3/metabolism , Receptors, CXCR3/genetics , Female , Meiosis/physiology , Mice, Knockout
5.
Sci Rep ; 14(1): 8196, 2024 04 08.
Article in English | MEDLINE | ID: mdl-38589444

ABSTRACT

In atherosclerotic lesions, monocyte-derived macrophages are major source of interferon gamma (IFN-γ), a pleotropic cytokine known to regulate the expression of numerous genes, including the antiviral gene RSAD2. While RSAD2 was reported to be expressed in endothelial cells of human carotid lesions, its significance for the development of atherosclerosis remains utterly unknown. Here, we harnessed publicly available human carotid atherosclerotic data to explore RSAD2 in lesions and employed siRNA-mediated gene-knockdown to investigate its function in IFN-γ-stimulated human aortic smooth muscle cells (hAoSMCs). Silencing RSAD2 in IFN-γ-stimulated hAoSMCs resulted in reduced expression and secretion of key CXCR3-chemokines, CXCL9, CXCL10, and CXCL11. Conditioned medium from RSAD2-deficient hAoSMCs exhibited diminished monocyte attraction in vitro compared to conditioned medium from control cells. Furthermore, RSAD2 transcript was elevated in carotid lesions where it was expressed by several different cell types, including endothelial cells, macrophages and smooth muscle cells. Interestingly, RSAD2 displayed significant correlations with CXCL10 (r = 0.45, p = 0.010) and CXCL11 (r = 0.53, p = 0.002) in human carotid lesions. Combining our findings, we uncover a novel role for RSAD2 in hAoSMCs, which could potentially contribute to monocyte recruitment in the context of atherosclerosis.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Humans , Plaque, Atherosclerotic/genetics , Interferons , Endothelial Cells/metabolism , Culture Media, Conditioned/pharmacology , Chemokines/genetics , Chemokines/metabolism , Chemokine CXCL11/genetics , Chemokine CXCL11/metabolism , Chemokine CXCL9/metabolism , Interferon-gamma/pharmacology , Interferon-gamma/metabolism , Atherosclerosis/genetics , Myocytes, Smooth Muscle/metabolism , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism , Viperin Protein
6.
Int Immunopharmacol ; 132: 111929, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38555817

ABSTRACT

Increased expression of CXCL10 and its receptor CXCR3 represents an inflammatory response in cells and tissues. Macrophage polarization and autophagy are major functions in inflammatory macrophages; however, the cellular functions of the CXCL10-CXCR3 axis in macrophages are not well understood. Here, we examined the role of CXCL10-CXCR3-axis-regulated autophagy in macrophage polarization. First, in non-inflammatory macrophages, whereas CXCL10 promotes M2 polarization and inhibits M1 polarization, CXCR3 antagonist AMG487 induces the opposite macrophage polarization. Next, CXCL10 promotes the expression of autophagy proteins (Atg5-Atg12 complex, p62, LC3-II, and LAMP1) and AMG487 inhibits their expression. Knockdown of LAMP1 by short interfering RNA switches the CXCL10-induced polarization from M2 to M1 in non-inflammatory macrophages. Furthermore, in inflammatory macrophages stimulated by poly(I:C), CXCL10 induces M1 polarization and AMG487 induces M2 polarization in association with a decrease in LAMP1. Finally, AMG487 alleviates lung injury after poly(I:C) treatment in mice. In conclusion, CXCL10-CXCR3 axis differentially directs macrophage polarization in inflammatory and non-inflammatory states, and autophagy protein LAMP1 acts as the switch controlling the direction of macrophage polarization by CXCL10-CXCR3.


Subject(s)
Acetamides , Autophagy , Chemokine CXCL10 , Inflammation , Macrophages , Mice, Inbred C57BL , Pyrimidinones , Receptors, CXCR3 , Animals , Receptors, CXCR3/metabolism , Receptors, CXCR3/genetics , Chemokine CXCL10/metabolism , Chemokine CXCL10/genetics , Macrophages/immunology , Macrophages/metabolism , Mice , Autophagy/immunology , Inflammation/immunology , Inflammation/metabolism , Poly I-C/pharmacology , Lysosomal Membrane Proteins/metabolism , Lysosomal Membrane Proteins/genetics , Male , Signal Transduction , Humans , Macrophage Activation
7.
Toxicol In Vitro ; 96: 105782, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38244730

ABSTRACT

Estrogen-induced intrahepatic cholestasis (IHC) is a mild but potentially serious risk and urges for new therapeutic targets and effective treatment. Our previous study demonstrated that RORγt and CXCR3 signaling pathway of invariant natural killer T (iNKT) 17 cells play pathogenic roles in 17α-ethinylestradiol (EE)-induced IHC. Ursodeoxycholic acid (UDCA) and 18ß-glycyrrhetinic acid (GA) present a protective effect on IHC partially due to their immunomodulatory properties. Hence in present study, we aim to investigate the effectiveness of UDCA and 18ß-GA in vitro and verify the accessibility of the above targets. Biochemical index measurement indicated that UDCA and 18ß-GA presented efficacy to alleviate EE-induced cholestatic cytotoxicity. Both UDCA and 18ß-GA exhibited suppression on the CXCL9/10-CXCR3 axis, and significantly restrained the expression of RORγt in vitro. In conclusion, our observations provide new therapeutic targets of UDCA and 18ß-GA, and 18ß-GA as an alternative treatment for EE-induced cholestasis.


Subject(s)
Cholestasis , Glycyrrhetinic Acid , Natural Killer T-Cells , Receptors, CXCR3 , Ursodeoxycholic Acid , Cholestasis/chemically induced , Cholestasis/drug therapy , Ethinyl Estradiol/toxicity , Glycyrrhetinic Acid/analogs & derivatives , Glycyrrhetinic Acid/pharmacology , Glycyrrhetinic Acid/therapeutic use , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism , Signal Transduction , Ursodeoxycholic Acid/pharmacology , Ursodeoxycholic Acid/therapeutic use , Animals , Mice
8.
Immunol Res ; 72(3): 396-408, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38151700

ABSTRACT

In rheumatoid arthritis (RA), immune homeostasis is maintained by T regulatory cells (Tregs) that in an inflammatory milieu can change towards T-helper-like phenotypes (Th-like Tregs). Our aim was to examine the phenotypic and functional characteristics of CD4+CD25+CD127lo/- Tregs, Th-like Tregs and T effector (Teff) cells in the peripheral blood (PB) and synovial fluid (SF) of treatment-naïve early RA, as compared to osteoarthritis (OA) and healthy control (HC) peripheral blood. Frequencies of Tregs, CXCR3, CCR6 expressing Tregs (Th-like Tregs), and Teff cells were analyzed using flow cytometry in RA (n = 80), OA (n = 20), and HC (n = 40). Cytokine concentrations of the respective T cell subsets in plasma and SF were measured using flow cytometric bead array. Tregs sorted from RA and HC PB using magnetic beads were analyzed for functional capacities by CFSE proliferation assay and FOXP3 gene expression using real-time PCR. We observed that the frequencies of Th17 cells in PB and SF were significantly higher in RA when compared to HC, whereas Tregs were lower in PB and high in SF compared to HC and OA respectively. Th1- and Th17-related pro-inflammatory cytokines IL12p70, INF-γ, TNF-α, and IL-6, and IL-17A were significantly higher in the plasma and SF of RA. Tregs expressing CXCR3 (Th1-like Tregs) and CCR6 (Th17-like Treg) were significantly higher in PB and SF of RA compared to controls and was positively associated with seropositivity and disease activity. Treg cells isolated from peripheral blood of RA showed decreased function and reduced FOXP3 gene expression compared to HC. In our study, we have demonstrated higher frequencies of Th1 and Th17 cells and increased circulatory and SF pro-inflammatory cytokines (IL12P70, INF-γ, IL-6, IL-17A, and TNF-α) in RA. This inflammatory milieu might alter total Tregs frequencies and influence conversion of Tregs into Th-like Tregs.


Subject(s)
Arthritis, Rheumatoid , Receptors, CCR6 , Receptors, CXCR3 , T-Lymphocytes, Regulatory , Humans , Arthritis, Rheumatoid/immunology , T-Lymphocytes, Regulatory/immunology , Receptors, CCR6/metabolism , Receptors, CCR6/genetics , Receptors, CXCR3/metabolism , Receptors, CXCR3/genetics , Female , Middle Aged , Male , Aged , Adult , Cytokines/metabolism , Osteoarthritis/immunology , Osteoarthritis/metabolism , Synovial Fluid/immunology , Synovial Fluid/metabolism , Th17 Cells/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics
9.
Cell Death Dis ; 14(12): 835, 2023 12 16.
Article in English | MEDLINE | ID: mdl-38104126

ABSTRACT

Glioblastoma (GBM) is a highly aggressive brain tumor associated with limited therapeutic options and a poor prognosis. CXCR3, a chemokine receptor, serves dual autocrine-paracrine functions in cancer. Despite gaps in our understanding of the functional role of the CXCR3 receptor in GBM, it has been shown to hold promise as a therapeutic target for the treatment of GBM. Existing clinical therapeutics and vaccines targeting CXCR3 ligand expression associated with the CXCR3 axes have also shown anti-tumorigenic effects in GBM. This review summarizes existing evidence on the oncogenic function of CXCR3 and its ligands CXCL9, CXCL10, and CXCL11, in GBM, and examines the controversies concerning the immunomodulatory functions of the CXCR3 receptor, including immune T cell recruitment, polarization, and positioning. The mechanisms underlying monotherpies and combination therapies targeting the CXCR3 pathways are discussed. A better understanding of the CXCR3 axes may lead to the development of strategies for overcoming the limitations of existing immunotherapies for GBM.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Glioblastoma/genetics , Glioblastoma/therapy , Chemokine CXCL10 , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism , Chemokine CXCL9 , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Brain Neoplasms/pathology , T-Lymphocytes/metabolism , Ligands
10.
Front Immunol ; 14: 1219279, 2023.
Article in English | MEDLINE | ID: mdl-37790939

ABSTRACT

The transcription factor Fli-1, a member of the ETS family of transcription factors, is implicated in the pathogenesis of lupus disease. Reduced Fli-1 expression in lupus mice leads to decreased renal Cxcl10 mRNA levels and renal infiltrating CXCR3+ T cells that parallels reduced renal inflammatory cell infiltration and renal damage. Inflammatory chemokine CXCL10 is critical for attracting inflammatory cells expressing the chemokine receptor CXCR3. The CXCL10/CXCR3 axis plays a role in the pathogenesis of various inflammatory diseases including lupus. Our data here demonstrate that renal CXCL10 protein levels are significantly lower in Fli-1 heterozygous MRL/lpr mice compared to wild-type MRL/lpr mice. Knockdown of Fli-1 significantly reduced CXCL10 secretion in mouse and human endothelial cells, and human mesangial cells, upon LPS or TNFα stimulation. The Fli-1 inhibitor, Camptothecin, significantly reduced CXCL10 production in human monocyte cells upon interferon stimulation. Four putative Ets binding sites in the Cxcl10 promoter showed significant enrichment for FLI-1; however, FLI-1 did not directly drive transcription from the human or mouse promoters, suggesting FLI-1 may regulate CXCL10 expression indirectly. Our results also suggest that the DNA binding domain of FLI-1 is necessary for regulation of human hCXCR3 promotor activity in human T cells and interactions with co-activators. Together, these results support a role for FLI-1 in modulating the CXCL10-CXCR3 axis by directly or indirectly regulating the expression of both genes to impact lupus disease development. Signaling pathways or drugs that reduce FLI-1 expression may offer novel approaches to lupus treatment.


Subject(s)
Endothelial Cells , Proto-Oncogene Protein c-fli-1 , Animals , Humans , Mice , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Endothelial Cells/metabolism , Kidney/pathology , Mice, Inbred MRL lpr , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Protein c-fli-1/metabolism , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism
11.
Eur J Immunol ; 53(12): e2350574, 2023 12.
Article in English | MEDLINE | ID: mdl-37689974

ABSTRACT

Various regulatory CD8+ T-cell subsets have been proposed for immune tolerance and have been implicated in controlling autoimmune diseases. However, their phenotypic identities and suppression mechanisms are not yet understood. This study found that coculture of T-cell receptor (TCR)- or interferon (IFN)-ß-activated CD8+ T cells significantly suppressed the cytokine production of Th1 and Th17 cells. By experimenting with the experimental autoimmune uveitis (EAU), we found that adoptive transfer of TCR or IFN-ß-activated CD8+ T cells significantly lessened disease development in an IFN-γ-dependent manner with a decreased uveitogenic Th1 and Th17 response. Interestingly, after adoptive transfer into the EAU mice, the IFN-γ+ CD8+ T cells were recruited more efficiently into the secondary lymphoid organs during the disease-priming phase. This recruitment depends on the IFN-γ-inducible chemokine receptor CXCR3; knocking out CXCR3 abolishes the protective effect of CD8+ T cells in EAU. In conclusion, we identified the critical role of IFN-γ for CD8+ T cells to inhibit Th1 and Th17 responses and ameliorate EAU. CXCR3 is necessary to recruit IFN-γ+ CD8+ T cells to the secondary lymphoid organ for the regulation of autoreactive Th1 and Th17 cells.


Subject(s)
CD8-Positive T-Lymphocytes , Interferon-gamma , Retinitis , Male , Female , Animals , Mice , Mice, Inbred C57BL , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation , Retinitis/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Interferon-gamma/immunology , Cell Polarity/immunology , Interleukin-10/immunology , Interferon-beta/pharmacology , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Protein Transport/genetics , Spleen/immunology
12.
Immunity ; 56(7): 1613-1630.e5, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37392735

ABSTRACT

Infiltration of regulatory T (Treg) cells, an immunosuppressive population of CD4+ T cells, into solid cancers represents a barrier to cancer immunotherapy. Chemokine receptors are critical for Treg cell recruitment and cell-cell interactions in inflamed tissues, including cancer, and thus are an ideal therapeutic target. Here, we show in multiple cancer models that CXCR3+ Treg cells were increased in tumors compared with lymphoid tissues, exhibited an activated phenotype, and interacted preferentially with CXCL9-producing BATF3+ dendritic cells (DCs). Genetic ablation of CXCR3 in Treg cells disrupted DC1-Treg cell interactions and concomitantly increased DC-CD8+ T cell interactions. Mechanistically, CXCR3 ablation in Treg cells increased tumor antigen-specific cross-presentation by DC1s, increasing CD8+ T cell priming and reactivation in tumors. This ultimately impaired tumor progression, especially in combination with anti-PD-1 checkpoint blockade immunotherapy. Overall, CXCR3 is shown to be a critical chemokine receptor for Treg cell accumulation and immune suppression in tumors.


Subject(s)
Neoplasms , T-Lymphocytes, Regulatory , Humans , Neoplasms/metabolism , CD8-Positive T-Lymphocytes , Immunotherapy , Dendritic Cells/metabolism , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism
13.
Curr Microbiol ; 80(6): 201, 2023 May 04.
Article in English | MEDLINE | ID: mdl-37140634

ABSTRACT

Brucella spp. can replicate in human endothelial cells, inducing an inflammatory response with increased expression of chemokines. Although Brucella infects humans, its ability to induce the production of chemokines by lung cells is unknown. Therefore, the current investigation was designed to examine the association between brucellosis and CXCL9, 10, and 11 chemokines. The patient group included 71 patients suffering from Brucella infection and the control group consisted of 50 healthy ranchers from the same geographical area. Serum levels of CXCL9, CXCL10, and CXCL11 were analyzed by ELISA. The fold changes of CXCR3 expression against ß-actin were determined by real-time-PCR technique. Western blotting analysis was also applied for evaluating the expression of CXCR3 at protein level. The results of this study showed that the serum levels of CXCL9, CXCL10, and CXCL11 are significantly increased in acute brucellosis patients in comparison to control as indicated by ELISA test, mRNA levels of CXCR3 by Real-time PCR as well as protein levels of CXCR3 by Western blot analysis. According to findings, these chemokines have the potential to serve as markers for brucellosis patients. Taken together, cytokine/chemokine network was active in acute brucellosis patients, and it is suggested to evaluate other cytokines in future studies.


Subject(s)
Brucellosis , Chemokine CXCL10 , Humans , Chemokine CXCL10/genetics , Leukocytes, Mononuclear/metabolism , Endothelial Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Chemokine CXCL9/genetics , Chemokine CXCL11/genetics , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism
14.
Cell Mol Life Sci ; 80(3): 78, 2023 Mar 02.
Article in English | MEDLINE | ID: mdl-36862204

ABSTRACT

Chemokines are pivotal players in instigation and perpetuation of synovitis through leukocytes egress from the blood circulation into the inflamed articulation. Multitudinous literature addressing the involvement of the dual-function interferon (IFN)-inducible chemokines CXCL9, CXCL10 and CXCL11 in diseases characterized by chronic inflammatory arthritis emphasizes the need for detangling their etiopathological relevance. Through interaction with their mutual receptor CXC chemokine receptor 3 (CXCR3), the chemokines CXCL9, CXCL10 and CXCL11 exert their hallmark function of coordinating directional trafficking of CD4+ TH1 cells, CD8+ T cells, NK cells and NKT cells towards inflammatory niches. Among other (patho)physiological processes including infection, cancer, and angiostasis, IFN-inducible CXCR3 ligands have been implicated in autoinflammatory and autoimmune diseases. This review presents a comprehensive overview of the abundant presence of IFN-induced CXCR3 ligands in bodily fluids of patients with inflammatory arthritis, the outcomes of their selective depletion in rodent models, and the attempts at developing candidate drugs targeting the CXCR3 chemokine system. We further propose that the involvement of the CXCR3 binding chemokines in synovitis and joint remodeling encompasses more than solely the directional ingress of CXCR3-expressing leukocytes. The pleotropic actions of the IFN-inducible CXCR3 ligands in the synovial niche reiteratively illustrate the extensive complexity of the CXCR3 chemokine network, which is based on the intercommunion of IFN-inducible CXCR3 ligands with distinct CXCR3 isoforms, enzymes, cytokines, and infiltrated and resident cells present in the inflamed joints.


Subject(s)
Arthritis , Autoimmune Diseases , Humans , CD8-Positive T-Lymphocytes , Ligands , Receptors, CXCR3/genetics , Interferons/pharmacology
15.
Cancer Sci ; 114(6): 2622-2633, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36898851

ABSTRACT

Epstein-Barr virus (EBV)-positive diffuse large B-cell lymphoma associated with chronic inflammation (DLBCL-CI) develops in the setting of long-standing inflammation. This type of lymphoma may have specific expression profiles of chemokines involved in the pathogenesis of DLBCL-CI. EBV-positive pyothorax-associated lymphoma (PAL) is a prototype of DLBCL-CI and represents a valuable model for the study of this disease category. Using a panel of PAL cell lines, we found that PAL cells expressed and secreted C-X-C motif chemokine ligands 9 and 10 (CXCL9 and CXCL10), the ligands of CXCR3, in contrast to EBV-negative DLBCL cell lines, which did not. Culture supernatants from PAL cell lines attracted CXCR3-expressing CD4+ T cells, CD8+ T cells, and CD56+ natural killer cells from human peripheral blood mononuclear cells. PAL cells injected into mice also attracted CXCR3-positive cytotoxic lymphocytes that expressed interferon-γ. The expression of CXCL9 and CXCL10 was detected in PAL tumor biopsy samples from patients, and CXCR3-positive lymphocytes were abundant in the tissue samples. Collectively, these findings suggest that CXCL9 and CXCL10 are produced by PAL cells and can elicit cytotoxic responses via CXCR3. This chemokine system is also likely to contribute to tissue necrosis, which is a signature histological feature of DLBCL-CI. Further studies are warranted to determine whether the CXCL9-CXCL10/CXCR3 axis exerts antitumor effects in DLBCL-CI.


Subject(s)
Empyema, Pleural , Epstein-Barr Virus Infections , Lymphoma, Large B-Cell, Diffuse , Mice , Animals , Humans , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Herpesvirus 4, Human/metabolism , CD8-Positive T-Lymphocytes/metabolism , Epstein-Barr Virus Infections/complications , Leukocytes, Mononuclear/metabolism , Ligands , Inflammation , Killer Cells, Natural/metabolism , Chemokine CXCL9 , Receptors, CXCR3/genetics
16.
Mol Cells ; 46(5): 281-297, 2023 May 31.
Article in English | MEDLINE | ID: mdl-36799104

ABSTRACT

CXCR3 regulates leukocyte trafficking, maturation, and various pathophysiological conditions. Alternative splicing generates three CXCR3 isoforms in humans. Previous studies investigated the roles of CXCR3 isoforms, and some biochemical data are not correlated with biological relevance analyses. RT-PCR analyses indicate that most cells express all three splicing variants, suggesting that they may mutually affect the chemokine binding and cellular responses of other splicing variants. Here, we performed an integrative analysis of the functional relations among CXCR3 splicing variants and their chemokine-dependent signaling using NanoBiT live cell protein interaction assays. The results indicated that the CXCR3 N-terminal region affected cell surface expression levels and ligand-dependent activation. CXCR3A was efficiently expressed in the plasma membrane and responded to I-TAC, IP-10, and MIG chemokines. By contrast, CXCR3B had low plasma membrane expression and mediated I-TAC-stimulated cellular responses. CXCR3Alt was rarely expressed on the cell surface and did not mediate any cell responses to the tested chemokines; however, CXCR3Alt negatively affected the plasma membrane expression of CXCR3A and CXCR3B and their chemokine-stimulated cellular responses. Jurkat cells express endogenous CXCR3, and exogenous CXCR3A expression enhanced chemotactic activity in response to I-TAC, IP-10, and MIG. By contrast, exogenous expression of CXCR3B and CXCR3Alt eliminated or reduced the CXCR3A-induced chemotactic activity. The PF-4 chemokine did not activate any CXCR3-mediated cellular responses. NanoBiT technology are useful to integrative studies of CXCR3-mediated cell signaling, and expand our knowledge of the cellular responses mediated by molecular interactions among the splicing variants, including cell surface expression, ligand-dependent receptor activation, and chemotaxis.


Subject(s)
Chemokine CXCL10 , Signal Transduction , Humans , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Ligands , Alternative Splicing , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism
17.
J Invest Dermatol ; 143(7): 1138-1146.e12, 2023 07.
Article in English | MEDLINE | ID: mdl-36708947

ABSTRACT

Morphea is characterized by initial inflammation followed by fibrosis of the skin and soft tissue. Despite its substantial morbidity, the pathogenesis of morphea is poorly studied. Previous work showed that CXCR3 ligands CXCL9 and CXCL10 are highly upregulated in the sera and lesional skin of patients with morphea. We found that an early inflammatory subcutaneous bleomycin mouse model of dermal fibrosis mirrors the clinical, histological, and immune dysregulation observed in human morphea. We used this model to examine the role of the CXCR3 chemokine axis in the pathogenesis of cutaneous fibrosis. Using the REX3 (Reporting the Expression of CXCR3 ligands) mice, we characterized which cells produce CXCR3 ligands over time. We found that fibroblasts contribute the bulk of CXCL9-RFP and CXCL10-BFP by percentage, whereas macrophages produce high amounts on a per-cell basis. To determine whether these chemokines are mechanistically involved in pathogenesis, we treated Cxcl9-, Cxcl10-, or Cxcr3-deficient mice with bleomycin and found that fibrosis is dependent on CXCL9 and CXCR3. Addition of recombinant CXCL9 but not CXCL10 to cultured mouse fibroblasts induced Col1a1 mRNA expression, indicating that the chemokine itself contributes to fibrosis. Taken together, our studies provide evidence that CXCL9 and its receptor CXCR3 are functionally required for inflammatory fibrosis.


Subject(s)
Dermatitis , Scleroderma, Localized , Humans , Animals , Mice , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Up-Regulation , Ligands , Chemokine CXCL9/genetics , Chemokine CXCL9/metabolism , Fibrosis , Inflammation , Fibroblasts/metabolism , Bleomycin/toxicity , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism
18.
Cancer Immunol Immunother ; 72(6): 1865-1880, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36688994

ABSTRACT

Radiotherapy (RT) not only damages tumors but also induces interferon (IFN) expression in tumors. IFNs mediate PD-L1 to exhaust CD8+ T cells, but which also directly impact tumor cells and potentially activate anti-tumor immune surveillance. Little is known about the contradictory mechanism of IFNs in regulating CD8+ T-mediated anti-tumor activity in lung cancer. This study found that RT induced IFNs and CXCL9/10 expression in the RT-treated lung cancer cells. Specifically, RT- and IFNγ-pretreated A549 significantly activated CD8+ T cells, resulting in significant inhibition of A549 colony formation. RNAseq and consequent qPCR results revealed that IFNγ induced PD-L1, CXCL10, and ICAM-1, whereas PD-L1 knockdown activated CD8+ T cells, but ICAM-1 knockdown diminished CD8+ T cell activation. We further demonstrated that CXCR3 and CXCL10 decreased in the CD8+ T cells and nonCD8+ PBMCs, respectively, in the patients with lung cancer that expressed lower reactivation as co-cultured with A549 cells. In addition, inhibitors targeting CXCR3 and LFA-1 in CD8+ T cells significantly diminished CD8+ T cell activation and splenocytes-mediated anti-LL/2shPdl1. In conclusion, we validated that RT suppressed lung cancer and overexpress PD-L1, CXCL10, and ICAM-1, which exhibited different roles in regulating CD8+ T cell activity. We propose that CXCR3highCD8+ T cells stimulated by CXCL10 exhibit anti-tumor immunity, possibly by enhancing T cells-tumor cells adhesion through CXCL10/CXCR3-activated LFA-1-ICAM-1 interaction, but CXCR3lowCD8+ T cells with low CXCL10 in patients with lung cancer were exhausted by PD-L1 dominantly. Therefore, RT potentially activates CD8+ T cells by inducing IFNs-mediated CXCL10 and ICAM-1 expression in tumors to enhance CD8+ T-tumor adhesion and recognition. This study clarified the possible mechanisms of RT and IFNs in regulating CD8+ T cell activation in lung cancer.


Subject(s)
CD8-Positive T-Lymphocytes , Lung Neoplasms , Humans , Chemokine CXCL10/metabolism , B7-H1 Antigen/metabolism , Intercellular Adhesion Molecule-1/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , Interferon-gamma/metabolism , Lung Neoplasms/radiotherapy , Lung Neoplasms/metabolism , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism
19.
Int J Biochem Cell Biol ; 152: 106311, 2022 11.
Article in English | MEDLINE | ID: mdl-36195287

ABSTRACT

Development of fibrosis leads to end stage diseases that defy treatments across all organs. This ensues as chronic inflammation is not dampened by physiologic processes that issue in the resolution phase of wound healing. Thus, these conditions can be considered diseases of "failure to heal". In the absence of broadly viable treatments, it is proposed to examine key switches in wound healing resolution to seek insights into novel approaches. Signaling through the GPCR CXCR3 has been shown to be one such critical player in this physiologic transition that limits and even reverses early fibrosis. As such, a number of investigators and early stage technology companies have posited that triggering this signaling network would limit fibrosis. While there are some conflicting results, a consensus is emerging that pharmacologic interventions that promote signaling through this pathway represent innovative ways to limit fibrotic diseases.


Subject(s)
Fibrosis , Receptors, CXCR3 , Wound Healing , Humans , Fibrosis/genetics , Receptors, CXCR3/genetics , Signal Transduction/physiology , Wound Healing/genetics , Wound Healing/physiology
20.
Nat Commun ; 13(1): 5846, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36195635

ABSTRACT

Some G protein-coupled receptor (GPCR) ligands act as "biased agonists" that preferentially activate specific signaling transducers over others. Although GPCRs are primarily found at the plasma membrane, GPCRs can traffic to and signal from many subcellular compartments. Here, we determine that differential subcellular signaling contributes to the biased signaling generated by three endogenous ligands of the GPCR CXC chemokine receptor 3 (CXCR3). The signaling profile of CXCR3 changes as it traffics from the plasma membrane to endosomes in a ligand-specific manner. Endosomal signaling is critical for biased activation of G proteins, ß-arrestins, and extracellular-signal-regulated kinase (ERK). In CD8 + T cells, the chemokines promote unique transcriptional responses predicted to regulate inflammatory pathways. In a mouse model of contact hypersensitivity, ß-arrestin-biased CXCR3-mediated inflammation is dependent on receptor internalization. Our work demonstrates that differential subcellular signaling is critical to the overall biased response observed at CXCR3, which has important implications for drugs targeting chemokine receptors and other GPCRs.


Subject(s)
GTP-Binding Proteins , Receptors, CXCR3 , Animals , Chemokines/metabolism , GTP-Binding Proteins/metabolism , Ligands , Mice , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism , Receptors, G-Protein-Coupled/metabolism , beta-Arrestins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL