Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Cell Mol Gastroenterol Hepatol ; 12(1): 41-58, 2021.
Article in English | MEDLINE | ID: mdl-33548597

ABSTRACT

BACKGROUND & AIMS: Pancreatic ductal adenocarcinoma (PDA) is a lethal chemoresistant cancer that exhibits early metastatic spread. The highly immunosuppressive PDA tumor microenvironment renders patients resistant to emerging immune-targeted therapies. Building from our prior work, we evaluated stimulator of interferon genes (STING) agonist activation of PDA cell interferon-α/ß-receptor (IFNAR) signaling in systemic antitumor immune responses. METHODS: PDA cells were implanted subcutaneously to wild-type, IFNAR-, or CXCR3-knockout mice. Tumor growth was monitored, and immune responses were comprehensively profiled. RESULTS: Human and mouse STING agonist ADU-S100 reduced local and distal tumor burden and activated systemic antitumor immune responses in PDA-bearing mice. Effector T-cell infiltration and inflammatory cytokine and chemokine production, including IFN-dependent CXCR3-agonist chemokines, were elevated, whereas suppressive immune populations were decreased in treated tumors. Intratumoral STING agonist treatment also generated inflammation in distal noninjected tumors and peripheral immune tissues. STING agonist treatment of type I IFN-responsive PDA tumors engrafted to IFNAR-/- recipient mice was sufficient to contract tumors and stimulate local and systemic T-cell activation. Tumor regression and CD8+ T-cell infiltration were abolished in PDA engrafted to CXCR3-/- mice treated with STING agonist. CONCLUSIONS: These data indicate that STING agonists promote T-cell infiltration and counteract immune suppression in locally treated and distant tumors. Tumor-intrinsic type I IFN signaling initiated systemic STING-mediated antitumor inflammation and required CXCR3 expression. STING-mediated induction of systemic immune responses provides an approach to harness the immune system to treat primary and disseminated pancreatic cancers.


Subject(s)
Membrane Proteins/metabolism , Receptor, Interferon alpha-beta/metabolism , Receptors, CXCR3/metabolism , Animals , Cell Line, Tumor , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/deficiency , Receptors, CXCR3/deficiency , Signal Transduction
2.
J Virol ; 94(12)2020 06 01.
Article in English | MEDLINE | ID: mdl-32295905

ABSTRACT

We demonstrate that female C57BL/6J mice are susceptible to a transient lower genital tract infection with MmuPV1 mouse papillomavirus and display focal histopathological abnormalities resembling those of human papillomavirus (HPV) infection. We took advantage of strains of genetically deficient mice to study in vivo the role of innate immune signaling in the control of papillomavirus. At 4 months, we sacrificed MmuPV1-infected mice and measured viral 757/3139 spliced transcripts by TaqMan reverse transcription-PCR (RT-PCR), localization of infection by RNAscope in situ hybridization, and histopathological abnormities by hematoxylin and eosin (H&E) staining. Among mice deficient in receptors for pathogen-associated molecular patterns, MyD88-/- and STING-/- mice had 1,350 and 80 copies of spliced transcripts/µg RNA, respectively, while no viral expression was detected in MAVS-/- and Ripk2-/- mice. Mice deficient in an adaptor molecule, STAT1-/-, for interferon signaling had 46,000 copies/µg RNA. Among mice with targeted deficiencies in the inflammatory response, interleukin-1 receptor knockout (IL-1R-/-) and caspase-1-/- mice had 350 and 30 copies/µg RNA, respectively. Among mice deficient in chemokine receptors, CCR6-/- mice had 120 copies/µg RNA, while CXCR2-/- and CXCR3-/- mice were negative. RNAscope confirmed focal infection in MyD88-/-, STAT1-/-, and CCR6-/- mice but was negative for other gene-deficient mice. Histological abnormalities were seen only in the latter mice. Our findings and the literature support a working model of innate immunity to papillomaviruses involving the activation of a MyD88-dependent pathway and IL-1 receptor signaling, control of viral replication by interferon-stimulated genes, and clearance of virus-transformed dysplastic cells by the action of the CCR6/CCL20 axis.IMPORTANCE Papillomaviruses infect stratified squamous epithelia, and the viral life cycle is linked to epithelial differentiation. Additionally, changes occur in viral and host gene expression, and immune cells are activated to modulate the infectious process. In vitro studies with keratinocytes cannot fully model the complex viral and host responses and do not reflect the contribution of local and migrating immune cells. We show that female C57BL/6J mice are susceptible to a transient papillomavirus cervicovaginal infection, and mice deficient in select genes involved in innate immune responses are susceptible to persistent infection with variable manifestations of histopathological abnormalities. The results of our studies support a working model of innate immunity to papillomaviruses, and the model provides a framework for more in-depth studies. A better understanding of mechanisms of early viral clearance and the development of approaches to induce clearance will be important for cancer prevention and the treatment of HPV-related diseases.


Subject(s)
Host-Pathogen Interactions/immunology , Myeloid Differentiation Factor 88/immunology , Papillomaviridae/immunology , Papillomavirus Infections/immunology , RNA, Messenger/immunology , RNA, Viral/immunology , Receptors, Interleukin-1 Type I/immunology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Alternative Splicing , Animals , Caspase 1/deficiency , Caspase 1/genetics , Caspase 1/immunology , Cervix Uteri/immunology , Cervix Uteri/virology , Female , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Humans , Immunity, Innate , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Papillomaviridae/growth & development , Papillomaviridae/metabolism , Papillomavirus Infections/genetics , Papillomavirus Infections/virology , RNA, Messenger/genetics , RNA, Viral/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/deficiency , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/immunology , Receptors, CCR6/deficiency , Receptors, CCR6/genetics , Receptors, CCR6/immunology , Receptors, CXCR3/deficiency , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Receptors, Interleukin-1 Type I/deficiency , Receptors, Interleukin-1 Type I/genetics , Receptors, Interleukin-8B/deficiency , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/immunology , STAT1 Transcription Factor/deficiency , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , Signal Transduction , Vagina/immunology , Vagina/virology
3.
Artif Cells Nanomed Biotechnol ; 47(1): 1200-1206, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30957556

ABSTRACT

OBJECTIVE: To study the effect of miR-16-5p on lung cancer cell injury and apoptosis, and its mechanism. METHODS: LPS induced lung cancer cell A549 injury; qRT-PCR method was applied to detect the expression of miR-16-5p and CXCR3 in A549 cells. Con (without LPS treatment), LPS + miR-NC group (transfected negative control samples), LPS + miR-16-5p group (transfected miR-16-5p mimics); LPS + si-NC group (transfected negative control samples), LPS + si-CXCR3 group (transfected si-CXCR3); LPS + miR-16-5p + pcDNA3.1 group (co-transfected miR-16-5p mimics and pcDNA3.1), LPS + miR-16-5p + pcDNA3.1-CXCR3 group (co-transfected miR-16-5p mimics and pcDNA3.1-CXCR3) were transfected into A549 cells by liposome method. Western blot was used to detect protein expression of CXCR3, IL-6 and TNF-α in A549 cells; apoptosis of A549 cells was detected by flow cytometry. RESULTS: Compared with the control group, the expression of miR-16-5p mRNA was significantly decreased in A549 cells in LPS group, and the mRNA and protein expression of CXCR3 were significantly increased (p < .05). Overexpression of miR-16-5p and knockdown of CXCR3 both can down-regulated protein expression of IL-6 and TNF-α, and up-regulated apoptosis in LPS-induced A549 cell; CXCR3 is a target of miR-16-5p. Overexpression of CXCR3 rescued the protective effect of miR-16-5p on LPS-induced A549 cell injury. CONCLUSION: miR-16-5p can protect LPS-induced A549 cell injury, and its mechanism may be related to the targeted regulation of CXCR3, which could provide a new target for targeted therapy of lung cancer.


Subject(s)
Lipopolysaccharides/pharmacology , MicroRNAs/genetics , Receptors, CXCR3/genetics , A549 Cells , Apoptosis/drug effects , Apoptosis/genetics , Base Sequence , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Humans , Interleukin-6/genetics , Receptors, CXCR3/deficiency , Tumor Necrosis Factor-alpha/genetics
4.
Transplantation ; 103(9): 1809-1820, 2019 09.
Article in English | MEDLINE | ID: mdl-30830040

ABSTRACT

BACKGROUND: We previously reported the novel activity of alloprimed CD8 T cells that suppress posttransplant alloantibody production. The purpose of the study is to investigate the expression and role of CXCR5 on antibody-suppressor CD8 T-cell function. METHODS: C57BL/6 mice were transplanted with FVB/N hepatocytes. Alloprimed CD8 T cells were retrieved on day 7 from hepatocyte transplant recipients. Unsorted or flow-sorted (CXCR5CXCR3 and CXCR3CXCR5) alloprimed CD8 T-cell subsets were analyzed for in vitro cytotoxicity and capacity to inhibit in vivo alloantibody production following adoptive transfer into C57BL/6 or high alloantibody-producing CD8 knock out (KO) hepatocyte transplant recipients. Alloantibody titer was assessed in CD8 KO mice reconstituted with naive CD8 T cells retrieved from C57BL/6, CXCR5 KO, or CXCR3 KO mice. Antibody suppression by ovalbumin (OVA)-primed monoclonal OVA-specific t-cell receptor transgenic CD8+ T cells (OT-I) CXCR5 or CXCR3 CD8 T-cell subsets was also investigated. RESULTS: Alloprimed CXCR5CXCR3CD8 T cells mediated in vitro cytotoxicity of alloprimed "self" B cells, while CXCR3CXCR5CD8 T cells did not. Only flow-sorted alloprimed CXCR5CXCR3CD8 T cells (not flow-sorted alloprimed CXCR3CXCR5CD8 T cells) suppressed alloantibody production and enhanced graft survival when transferred into transplant recipients. Unlike CD8 T cells from wild-type or CXCR3 KO mice, CD8 T cells from CXCR5 KO mice do not develop alloantibody-suppressor function. Similarly, only flow-sorted CXCR5CXCR3 (and not CXCR3CXCR5) OVA-primed OT-I CD8 T cells mediated in vivo suppression of anti-OVA antibody production. CONCLUSIONS: These data support the conclusion that expression of CXCR5 by antigen-primed CD8 T cells is critical for the function of antibody-suppressor CD8 T cells.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , B-Lymphocytes/immunology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Graft Rejection/prevention & control , Hepatocytes/transplantation , Liver Transplantation , Receptors, CXCR5/immunology , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , CD8 Antigens/deficiency , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Coculture Techniques , Female , Graft Rejection/immunology , Graft Rejection/metabolism , Graft Rejection/pathology , Graft Survival , Hepatocytes/immunology , Hepatocytes/metabolism , Liver Transplantation/adverse effects , Male , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR3/deficiency , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Receptors, CXCR5/deficiency , Receptors, CXCR5/genetics , Signal Transduction , Time Factors
5.
J Cell Physiol ; 234(8): 13894-13905, 2019 08.
Article in English | MEDLINE | ID: mdl-30656691

ABSTRACT

Vulvovaginal candidiasis (VVC) is a common observed infection, affecting approximately 75% of women of reproductive age. Drug resistance represents a troublesome stumbling block associated with VVC therapy. Thus the aim of the present study was to provide information regarding the selection of potential drug targets for VVC. CXCR3-, CXCR4-, or CXCR/CXCR4 double-deficient mouse models of VVC were subsequently established, with changes to the load of Candida Albicans evaluated accordingly. The biological behaviors of the vaginal epithelial cells were characterized in response to the CXCR3-, CXCR4-, or CXCR3/CXCR4 double-knockout in vivo. Our initial observations revealed that in mice with VVC, CXCR3-, CXCR4-, or CXCR3 - CXCR4 double-knockout resulted in a decreased load of C. Albicans as well as reduced levels and proportion of Th17 cells. Proinflammatory cytokine production was found to be inhibited by CXCR3-, CXCR4-, or CXCR3/CXCR4 double-knockout whereby the mRNA and protein expressions CXCR3, CXCR4, IL-17, IL-6, and TNF-α exhibited decreased levels. CXCR3-, CXCR4-, or CXCR3/CXCR4 double-knockout appeared to function as positive proliferation factors, while playing a negative role in the processes of apoptosis and the cell cycle of vaginal epithelial cells. Taken together, the key findings of the study suggested that CXCR3/CXCR4 double-knockout could act to hinder the progression of VVC, highlighting its promise as a novel therapeutic target in the treatment of VVC. CXCR3 and CXCR4 genes may regulate Th17/IL-17 immune inflammatory pathways to participate in antifungal immunity.


Subject(s)
Candidiasis, Vulvovaginal/immunology , Candidiasis, Vulvovaginal/metabolism , Cytokines/biosynthesis , Inflammation Mediators/metabolism , Receptors, CXCR3/deficiency , Receptors, CXCR4/deficiency , Th17 Cells/pathology , Animals , Apoptosis , Candida albicans/physiology , Candidiasis, Vulvovaginal/blood , Candidiasis, Vulvovaginal/microbiology , Cell Cycle , Cell Proliferation , Cytokines/blood , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Epithelial Cells/pathology , Female , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, CXCR3/blood , Receptors, CXCR3/metabolism , Receptors, CXCR4/blood , Receptors, CXCR4/metabolism , Vagina/microbiology , Vagina/pathology
6.
Nat Commun ; 9(1): 4854, 2018 11 19.
Article in English | MEDLINE | ID: mdl-30451860

ABSTRACT

Natural killer (NK) cells are reported to have immunological memory, with CD49a+ liver-resident NK cells shown to confer hapten-specific memory responses, but how this memory is induced or maintained is unclear. Here we show that memory type I innate lymphoid cells (ILC1s), which express IL-7Rα, are generated in the lymph nodes (LNs) and require IL-7R signaling to maintain their longevity in the liver. Hapten sensitization initiates CXCR3-dependent recruitment of IL-7Rα+ ILC1s into skin-draining LNs, where they are primed and acquire hapten-specific memory potential. Memory IL-7Rα+ ILC1s then exit draining LNs and are preferentially recruited, via CXCR6, to reside in the liver. Moreover, long-term blockade of IL-7R signaling significantly reduces ILC1-mediated memory responses. Thus, our results identify a memory IL-7Rα+ ILC1 population and reveal a LN-liver axis that is essential for ILC1 memory generation and long-term maintenance.


Subject(s)
Immunologic Memory , Killer Cells, Natural/immunology , Liver/immunology , Lymph Nodes/immunology , Receptors, Interleukin-7/immunology , Spleen/immunology , Animals , Cell Lineage/immunology , Fingolimod Hydrochloride/pharmacology , Gene Expression , Haptens/administration & dosage , Immunity, Innate/drug effects , Immunization , Immunosuppressive Agents/pharmacology , Integrin alpha1/genetics , Integrin alpha1/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Liver/cytology , Liver/drug effects , Lymph Nodes/cytology , Lymph Nodes/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Parabiosis/methods , Primary Cell Culture , Receptors, CXCR3/deficiency , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Receptors, CXCR6/genetics , Receptors, CXCR6/immunology , Receptors, Interleukin-7/genetics , Spleen/cytology , Spleen/drug effects
7.
Lupus ; 27(11): 1854-1859, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30139297

ABSTRACT

Mesenchymal stem cell therapy is a promising candidate for the treatment of systemic lupus erythematosus (SLE). To exert their efficacy fully, mesenchymal stem cells must infiltrate efficiently into the lesion sites. Here, we examined the role of CXCR3 in mesenchymal stem cell infiltration into the kidney of MRL. Faslpr mice, which highly expressed CXCL10. The phenotypes, production of immunosuppressive mediators, and capacity to inhibit T and B cells of CXCR3-deficient mesenchymal stem cells were similar to those of wild-type mesenchymal stem cells. However, they showed less infiltration into the nephritic kidney, less conjugation with endothelial cells and weaker MMP-9 expression than did wild-type mesenchymal stem cells. Consequently, CXCR3-deficient mesenchymal stem cells did not ameliorate lupus symptoms in MRL. Faslpr mice in comparison with wild-type mesenchymal stem cells. In summary, our data suggest that upregulation of CXCR3 in mesenchymal stem cells will be a good strategy to increase their infiltration into the kidney, which will improve therapeutic outcomes in SLE.


Subject(s)
Kidney/pathology , Lupus Erythematosus, Systemic/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Animals , B-Lymphocytes/metabolism , Gene Expression , Immunoglobulin G/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Receptors, CXCR3/deficiency , Receptors, CXCR3/genetics
8.
Stem Cell Res Ther ; 8(1): 193, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28874184

ABSTRACT

BACKGROUND: Transplantation of mesenchymal stem cells (MSC) has been proposed to improve wound healing. However, as these cells only transiently survive in the implantation site, the mechanisms underlying this beneficial healing response are associated with restorative paracrine effects of MSC matricellular factors on resident stromal cells. However, this requires that the recipient has a robust reservoir of viable cells. Here, we examine the influence of MSCs on the behavior of cotransplanted fibroblasts, in a manner to provide augmented cellular reserve to debilitated individuals, specifically focusing on matrix remodeling following in-vivo wounding. METHODS: Using a Hylan-A dermal filler hydrogel containing collagen I and tenascin-C for delivery and increased survival of transplanted cells, we find that cotransplantation of MSCs with fibroblasts reduces scarring. RESULTS: Transplanted xenogeneic MSCs augmented fibroblast proliferation, migration, and extracellular matrix deposition critical for wound closure, and reduced inflammation following wounding. MSCs also corrected matrix remodeling by CXCR3-deficient fibroblasts which otherwise led to hypertrophic scarring. This effect was superior to MSC or fibroblast transplantation alone. CONCLUSIONS: Taken together, these data suggest that MSCs, even if eventually rejected, transplanted with fibroblasts normalize matrix regeneration during healing. The current study provides insight into cellular therapies as a viable method for antifibrotic treatment and demonstrates that even transiently engrafted cells can have a long-term impact via matrix modulation and education of other tissue cells.


Subject(s)
Cicatrix, Hypertrophic/prevention & control , Fibroblasts/transplantation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Surgical Wound/therapy , Wound Healing , Animals , Cell Communication , Cell- and Tissue-Based Therapy/methods , Cellulose/administration & dosage , Cicatrix, Hypertrophic/metabolism , Coculture Techniques , Drug Combinations , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Female , Fibroblasts/cytology , Fibroblasts/immunology , Fibroblasts/metabolism , Gene Deletion , Gene Expression , Hexamethonium Compounds/administration & dosage , Hyaluronic Acid/administration & dosage , Hyaluronic Acid/analogs & derivatives , Male , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Primary Cell Culture , Receptors, CXCR3/deficiency , Receptors, CXCR3/genetics , Skin/injuries , Skin/metabolism , Surgical Wound/metabolism , Surgical Wound/pathology , Tantalum/administration & dosage , Thrombin/administration & dosage , Wound Healing/drug effects
9.
J Virol ; 91(14)2017 07 15.
Article in English | MEDLINE | ID: mdl-28468883

ABSTRACT

Herpes simplex virus 1 (HSV-1) establishes latency within the sensory neurons of the trigeminal ganglia (TG). HSV-specific memory CD8+ T cells play a critical role in preventing HSV-1 reactivation from TG and subsequent virus shedding in tears that trigger recurrent corneal herpetic disease. The CXC chemokine ligand 10 (CXCL10)/CXC chemokine receptor 3 (CXCR3) chemokine pathway promotes T cell immunity to many viral pathogens, but its importance in CD8+ T cell immunity to recurrent herpes has been poorly elucidated. In this study, we determined how the CXCL10/CXCR3 pathway affects TG- and cornea-resident CD8+ T cell responses to recurrent ocular herpesvirus infection and disease using a well-established murine model in which HSV-1 reactivation was induced from latently infected TG by UV-B light. Following UV-B-induced HSV-1 reactivation, a significant increase in both the number and function of HSV-specific CXCR3+ CD8+ T cells was detected in TG and corneas of protected C57BL/6 (B6) mice, but not in TG and corneas of nonprotected CXCL10-/- or CXCR3-/- deficient mice. This increase was associated with a significant reduction in both virus shedding and recurrent corneal herpetic disease. Furthermore, delivery of exogenous CXCL10 chemokine in TG of CXCL10-/- mice, using the neurotropic adeno-associated virus type 8 (AAV8) vector, boosted the number and function of effector memory CD8+ T cells (TEM) and tissue-resident memory CD8+ T cells (TRM), but not of central memory CD8+ T cells (TCM), locally within TG, and improved protection against recurrent herpesvirus infection and disease in CXCL10-/- deficient mice. These findings demonstrate that the CXCL10/CXCR3 chemokine pathway is critical in shaping CD8+ T cell immunity, locally within latently infected tissues, which protects against recurrent herpesvirus infection and disease.IMPORTANCE We determined how the CXCL10/CXCR3 pathway affects CD8+ T cell responses to recurrent ocular herpesvirus infection and disease. Using a well-established murine model, in which HSV-1 reactivation in latently infected trigeminal ganglia was induced by UV-B light, we demonstrated that lack of either CXCL10 chemokine or its CXCR3 receptor compromised the mobilization of functional CD8+ TEM and CD8+ TRM cells within latently infected trigeminal ganglia following virus reactivation. This lack of T cell mobilization was associated with an increase in recurrent ocular herpesvirus infection and disease. Inversely, augmenting the amount of CXCL10 in trigeminal ganglia of latently infected CXCL10-deficient mice significantly restored the number of local antiviral CD8+ TEM and CD8+ TRM cells associated with protection against recurrent ocular herpes. Based on these findings, a novel "prime/pull" therapeutic ocular herpes vaccine strategy is proposed and discussed.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chemokine CXCL10/metabolism , Herpes Simplex/immunology , Immunologic Memory , Receptors, CXCR3/metabolism , Simplexvirus/immunology , Animals , Chemokine CXCL10/deficiency , Cornea/immunology , Cornea/virology , Disease Models, Animal , Herpes Simplex/prevention & control , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR3/deficiency , Recurrence , Trigeminal Ganglion/immunology , Trigeminal Ganglion/virology
10.
J Neurovirol ; 23(3): 394-403, 2017 06.
Article in English | MEDLINE | ID: mdl-28116674

ABSTRACT

Herpes simplex virus type 1 (HSV-1) encephalitis (HSE) is the most common fatal sporadic encephalitis in developed countries. There is evidence from HSE animal models that not only direct virus-mediated damage caused but also the host's immune response contributes to the high mortality of the disease. Chemokines modulate and orchestrate this immune response. Previous experimental studies in HSE models identified the chemokine receptor CXCR3 and its ligands as molecules with a high impact on the course of HSE in mouse models. In this study, the role of the chemokine receptor CXCR3 was evaluated after intranasal infection with the encephalitogenic HSV-1 strain 17 syn+ using CXCR3-deficient mice (CXCR3-/-) and wild-type controls. We demonstrated a neurotropic viral spread into the CNS of after intranasal infection. Although viral load and histological distribution of infected neurons were independent from CXCR3 signaling early after infection, CXCR3-deficient mice cleared HSV-1 more efficiently 14 days after infection. Furthermore, CXCR3 deficiency led to a decreased weight loss in mice after HSV-1 infection. T cell infiltration and microglial activation was prominently reduced by inhibition of CXCR3 signaling. Quantitative PCR of proinflammatory cytokines and chemokines confirmed the reduced neuroinflammatory response in CXCR3-deficient mice during HSE. Our results demonstrate that the recruitment of peripheral immune cells into the CNS, induction of neuroinflammation, and consecutive weight loss during herpes encephalitis is modulated by CXCR3 signaling. Interruption of the CXCR3 pathway ameliorates the detrimental host immune response and in turn, leads paradoxically to an enhanced viral clearance after intranasal infection. Our data gives further insight into the role of CXCR3 during HSE after intranasal infection.


Subject(s)
Brain/immunology , Disease Resistance/genetics , Encephalitis, Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Receptors, CXCR3/deficiency , Administration, Intranasal , Animals , Brain/virology , Cell Movement , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , DNA, Viral/genetics , DNA, Viral/immunology , Disease Models, Animal , Encephalitis, Herpes Simplex/pathology , Encephalitis, Herpes Simplex/virology , Gene Expression Regulation , Herpesvirus 1, Human/growth & development , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Leukocytes/immunology , Leukocytes/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , Microglia/virology , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Viral Load , Weight Loss/immunology
11.
J Autoimmun ; 78: 19-28, 2017 03.
Article in English | MEDLINE | ID: mdl-28129932

ABSTRACT

CXC Chemokine Receptor 3 (CXCR3) is functionally pleiotropic and not only plays an important role in chemotaxis, but also participates in T cell differentiation and may play a critical role in inducing and maintaining immune tolerance. These observations are particularly critical for autoimmune cholangitis in which CXCR3 positive T cells are found around the portal areas of both humans and mouse models of primary biliary cholangitis (PBC). Herein, we investigated the role of CXCR3 in the pathogenesis of autoimmune cholangitis. We have taken advantage of a unique CXCR3 knockout dnTGFßRII mouse to focus on the role of CXCR3, both by direct observation of its influence on the natural course of disease, as well as through adoptive transfer studies into Rag-/- mice. We report herein that not only do CXCR3 deficient mice develop an exacerbation of autoimmune cholangitis associated with an expanded effector memory T cell number, but also selective adoptive transfer of CXCR3 deficient CD8+ T cells induces autoimmune cholangitis. In addition, gene microarray analysis of CXCR3 deficient CD8+ T cells reveal an intense pro-inflammatory profile. Our data suggests that the altered gene profiles induced by CXCR3 deficiency promotes autoimmune cholangitis through pathogenic CD8+ T cells. These data have significance for human PBC and other autoimmune liver diseases in which therapeutic intervention might be directed to chemokines and/or their receptors.


Subject(s)
Autoimmunity/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Liver Cirrhosis, Biliary/genetics , Liver Cirrhosis, Biliary/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Receptors, CXCR3/deficiency , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Cytokines/blood , Cytokines/metabolism , Disease Models, Animal , Gene Expression Profiling , Immunologic Memory , Ligands , Liver Cirrhosis, Biliary/metabolism , Liver Cirrhosis, Biliary/pathology , Mice , Mice, Knockout , Receptors, CXCR3/metabolism
12.
PLoS One ; 11(10): e0162905, 2016.
Article in English | MEDLINE | ID: mdl-27727269

ABSTRACT

Chemokine C-X-C motif receptor 3 (CXCR3) is a chemokine receptor that is mainly expressed by activated T lymphocytes. T cells play important roles in allergic pulmonary inflammation, which is a hallmark of asthma and elicits the localized accumulation of activated T cells in the lung. In China, a marked increase in the incidence rate of chronic allergic pulmonary inflammation has made it a major public health threat. In the present study, we investigated the role of CXCR3 and its ligands in airway inflammation induced by house dust mite protein (HDMP) in a CXCR3 knockout (CXCR3KO) asthma mouse model. Pathological manifestations in the lung, cell counts and bronchoalveolar lavage fluid (BALF) classifications were studied using hematoxylin and eosin (H&E) staining. The levels of IL-4 and IFN-γ in the BALF and splenocyte supernatants were measured using ELISA. CD4+ and CD8+ T cells in the lung and spleen were analyzed by flow cytometry. RT-PCR was applied to measure the mRNA transcript levels of monokines induced by IFN-γ(CXCL9) and IFN-γ inducible protein 10(CXCL10). The total cell counts, eosinophil counts, and IL-4 levels in the BALF and cultured splenocyte supernatants were significantly increased, while the levels of IFN-γ were reduced in the HDMP groups(P<0.01). Changes in the total cell counts, eosinophil counts, and lymphocyte counts, as well as the total protein levels in the BALF, the levels of IL-4 in splenocyte supernatants, and the pathological manifestations in the lung, were all greater in CXCR3KO mice than in C57BL/6 wild-type mice. Furthermore, the expression levels of CXCL9 and CXCL10 mRNA transcripts in the lungs of CXCR3KO mice were lower than those in C57BL/6 wild-type mice (P<0.05). CXCR3 and its ligands (i.e., CXCL9 and CXCL10) may play anti-inflammatory roles in this animal model. Promoting the expression of CXCR3 and its ligands may represent a novel therapeutic approach for preventing and curing asthma.


Subject(s)
Arthropod Proteins/immunology , Asthma/physiopathology , Bronchial Hyperreactivity/pathology , Pyroglyphidae/metabolism , Receptors, CXCR3/genetics , Animals , Bronchial Hyperreactivity/immunology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Chemokine CXCL9/genetics , Chemokine CXCL9/metabolism , Disease Models, Animal , Interferon-gamma/analysis , Interleukin-4/analysis , Lung/cytology , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR3/deficiency , Spleen/cytology , Spleen/metabolism , Spleen/pathology
13.
J Immunol ; 197(5): 2016-26, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27465528

ABSTRACT

Immunotherapies have shown considerable efficacy for the treatment of various cancers, but a multitude of patients remain unresponsive for various reasons, including poor homing of T cells into tumors. In this study, we investigated the roles of the leukotriene B4 receptor, BLT1, and CXCR3, the receptor for CXCL9, CXCL10, and CXCL11, under endogenous as well as vaccine-induced antitumor immune response in a syngeneic murine model of B16 melanoma. Significant accelerations in tumor growth and reduced survival were observed in both BLT1(-/-) and CXCR3(-/-) mice as compared with wild-type (WT) mice. Analysis of tumor-infiltrating leukocytes revealed significant reduction of CD8(+) T cells in the tumors of BLT1(-/-) and CXCR3(-/-) mice as compared with WT tumors, despite their similar frequencies in the periphery. Adoptive transfer of WT but not BLT1(-/-) or CXCR3(-/-) CTLs significantly reduced tumor growth in Rag2(-/-) mice, a function attributed to reduced infiltration of knockout CTLs into tumors. Cotransfer experiments suggested that WT CTLs do not facilitate the infiltration of knockout CTLs to tumors. Anti-programmed cell death-1 (PD-1) treatment reduced the tumor growth rate in WT mice but not in BLT1(-/-), CXCR3(-/-), or BLT1(-/-)CXCR3(-/-) mice. The loss of efficacy correlated with failure of the knockout CTLs to infiltrate into tumors upon anti-PD-1 treatment, suggesting an obligate requirement for both BLT1 and CXCR3 in mediating anti-PD-1 based antitumor immune response. These results demonstrate a critical role for both BLT1 and CXCR3 in CTL migration to tumors and thus may be targeted to enhance efficacy of CTL-based immunotherapies.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Cell Movement/immunology , Gene Expression Regulation , Melanoma, Experimental/immunology , Receptors, CXCR3/metabolism , Receptors, Leukotriene B4/metabolism , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/immunology , Chemotaxis, Leukocyte , Immunotherapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR3/deficiency , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Receptors, Leukotriene B4/deficiency , Receptors, Leukotriene B4/genetics
14.
J Neuroinflammation ; 13(1): 76, 2016 Apr 11.
Article in English | MEDLINE | ID: mdl-27068264

ABSTRACT

BACKGROUND: Experimental autoimmune encephalomyelitis (EAE) is a mouse model of multiple sclerosis (MS). It has been shown that Th17 cells are critical for EAE pathogenesis. Mice lacking CXCR3 develop aggravated EAE compared with wild-type (WT) mice. This study investigated the effect of CXCR3 on Th17 expansion during EAE and further addressed the underlying mechanism. METHODS: Both active EAE and adoptive-transfer EAE experiments were employed for studying EAE pathogenesis in WT and CXCR3(-/-) mice. Demyelination and leukocyte infiltration in the spinal cord of mice were analyzed by luxol fast blue staining and flow cytometry analysis, respectively. Glial cells expressing CXCR3 in the spinal cord were analyzed by immunofluorescence staining. Cytokine and chemokine levels in the spinal cord were analyzed using quantitative real-time PCR and enzyme-linked immunosorbent assay (ELISA). The glial cell line U87MG was employed for studying the CXCR3 signaling-mediated mechanism regulating Th17 expansion. RESULTS: CXCR3(-/-) mice exhibited more severe EAE and had significantly increased central nervous system (CNS)-infiltrating Th17 cells compared with WT mice. Adoptive-transfer experiments showed that CXCR3(-/-) recipient mice that received Th17 cells polarized from splenocytes of myelin oligodendrocyte glycoprotein (MOG)-immunized CXCR3(-/-) mice or MOG-immunized WT mice always developed more severe EAE and had significantly increased CNS-infiltrating Th17 cells compared with WT recipient mice that received Th17 cells from the same origin. Furthermore, during EAE, the number of activated glial cells was increased in the CNS of MOG-immunized CXCR3(-/-) mice, and CXCR3-deficient glial cells expressed increased levels of cytokine genes required for Th17 expansion and recruitment. Finally, we found that extracellular signal-regulated kinase (ERK) activation elicited by CXCR3 signaling in U87MG cells attenuated the activation of NF-κB, a key transcription factor critical for the induction of IL-23 and CCL20, which are required for Th17 cell expansion and recruitment, respectively. CONCLUSIONS: This study demonstrates a previously unrecognized role of CXCR3 signaling in glial cells in negatively regulating Th17 cell expansion during EAE. Our results demonstrate that, in addition to its well-known role in the recruitment of immune cells, CXCR3 in CNS glial cells plays a critical role in restraining the pro-Th17 cytokine/chemokine milieu during EAE, thereby diminishing Th17 cell expansion in the CNS and suppressing disease development.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Receptors, CXCR3/immunology , Signal Transduction , Th17 Cells/immunology , Adoptive Transfer , Animals , Blotting, Western , Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Polymerase Chain Reaction , Receptors, CXCR3/deficiency
15.
Nat Immunol ; 17(2): 150-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26595890

ABSTRACT

Mucosal surfaces are exposed to environmental substances and represent a major portal of entry for microorganisms. The innate immune system is responsible for early defense against infections and it is believed that the interferons (IFNs) constitute the first line of defense against viruses. Here we identify an innate antiviral pathway that works at epithelial surfaces before the IFNs. The pathway is activated independently of known innate sensors of viral infections through a mechanism dependent on viral O-linked glycans, which induce CXCR3 chemokines and stimulate antiviral activity in a manner dependent on neutrophils. This study therefore identifies a previously unknown layer of antiviral defense that exerts its action on epithelial surfaces before the classical IFN response is operative.


Subject(s)
Immunity, Innate , Interferons/metabolism , Mucous Membrane/immunology , Mucous Membrane/metabolism , Virus Diseases/immunology , Virus Diseases/metabolism , Animals , Cell Line , Chemokine CXCL10/biosynthesis , Disease Models, Animal , Female , Gene Expression , Glycosylation , Herpes Simplex/genetics , Herpes Simplex/immunology , Herpes Simplex/metabolism , Herpes Simplex/virology , Herpesvirus 2, Human/immunology , Humans , Interferons/genetics , Ligands , Mice , Mice, Knockout , Mucous Membrane/virology , Neutrophils/immunology , Neutrophils/metabolism , Polysaccharides/immunology , Receptors, CXCR3/deficiency , Receptors, CXCR3/metabolism , Vagina/immunology , Vagina/metabolism , Vagina/virology , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Viral Load , Virus Diseases/virology
16.
Immunity ; 42(3): 524-37, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25769612

ABSTRACT

CD8(+) T cells play a critical role in limiting peripheral virus replication, yet how they locate virus-infected cells within tissues is unknown. Here, we have examined the environmental signals that CD8(+) T cells use to localize and eliminate virus-infected skin cells. Epicutaneous vaccinia virus (VV) infection, mimicking human smallpox vaccination, greatly increased expression of the CXCR3 chemokine receptor ligands CXCL9 and CXCL10 in VV-infected skin. Despite normal T cell numbers in the skin, Cxcr3(-/-) mice exhibited dramatically impaired CD8(+)-T-cell-dependent virus clearance. Intravital microscopy revealed that Cxcr3(-/-) T cells were markedly deficient in locating, engaging, and killing virus-infected cells. Further, transfer of wild-type CD8(+) T cells restored viral clearance in Cxcr3(-/-) animals. These findings demonstrate a function for CXCR3 in enhancing the ability of tissue-localized CD8(+) T cells to locate virus-infected cells and thereby exert anti-viral effector functions.


Subject(s)
Keratinocytes/immunology , Poxviridae Infections/immunology , Receptors, CXCR3/immunology , Skin/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccinia virus/immunology , Adoptive Transfer , Animals , Cell Movement , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Chemokine CXCL9/genetics , Chemokine CXCL9/immunology , Female , Gene Expression Regulation , Humans , Keratinocytes/pathology , Keratinocytes/virology , Mice, Transgenic , Poxviridae Infections/genetics , Poxviridae Infections/pathology , Poxviridae Infections/virology , Receptors, CXCR3/deficiency , Receptors, CXCR3/genetics , Signal Transduction , Skin/pathology , Skin/virology , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Cytotoxic/transplantation , Viral Load
17.
J Clin Invest ; 125(4): 1713-25, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25751061

ABSTRACT

Mammalian pregnancy requires protection against immunological rejection of the developing fetus bearing discordant paternal antigens. Immune evasion in this developmental context entails silenced expression of chemoattractant proteins (chemokines), thereby preventing harmful immune cells from penetrating the maternal-fetal interface. Here, we demonstrate that fetal wastage triggered by prenatal Listeria monocytogenes infection is driven by placental recruitment of CXCL9-producing inflammatory neutrophils and macrophages that promote infiltration of fetal-specific T cells into the decidua. Maternal CD8+ T cells with fetal specificity upregulated expression of the chemokine receptor CXCR3 and, together with neutrophils and macrophages, were essential for L. monocytogenes-induced fetal resorption. Conversely, decidual accumulation of maternal T cells with fetal specificity and fetal wastage were extinguished by CXCR3 blockade or in CXCR3-deficient mice. Remarkably, protection against fetal wastage and in utero L. monocytogenes invasion was maintained even when CXCR3 neutralization was initiated after infection, and this protective effect extended to fetal resorption triggered by partial ablation of immune-suppressive maternal Tregs, which expand during pregnancy to sustain fetal tolerance. Together, our results indicate that functionally overriding chemokine silencing at the maternal-fetal interface promotes the pathogenesis of prenatal infection and suggest that therapeutically reinforcing this pathway represents a universal approach for mitigating immune-mediated pregnancy complications.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Fetal Death/prevention & control , Listeriosis/immunology , Pregnancy Complications, Infectious/immunology , Receptors, CXCR3/physiology , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Ampicillin/therapeutic use , Animals , Anti-Bacterial Agents/therapeutic use , Chemokine CXCL9/biosynthesis , Chemokine CXCL9/genetics , Chemokine CXCL9/physiology , Chemokines/metabolism , Crosses, Genetic , Decidua/immunology , Female , Fetal Death/etiology , Fetal Resorption/immunology , Fetal Resorption/prevention & control , Listeriosis/drug therapy , Macrophages/immunology , Male , Maternal-Fetal Exchange , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neutrophils/immunology , Ovalbumin/genetics , Ovalbumin/immunology , Peptide Fragments/genetics , Peptide Fragments/immunology , Pregnancy , Pregnancy Complications, Infectious/drug therapy , Receptors, CXCR3/antagonists & inhibitors , Receptors, CXCR3/biosynthesis , Receptors, CXCR3/deficiency , Receptors, CXCR3/genetics , Spleen/immunology , T-Cell Antigen Receptor Specificity , T-Lymphocytes, Regulatory/immunology , Up-Regulation , Virulence
18.
J Neuroimmunol ; 276(1-2): 219-23, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25139013

ABSTRACT

CXCR3 deficient (CXCR3(-/-)) mice are resistant to ocular HSV-1 infection in that less mice develop encephalitis and succumb to infection in comparison to wild type (WT) animals. A region of the brain previously identified to be crucial for development of encephalitis was evaluated in HSV-1-infected CXCR3(-/-) and WT mice. In this region, known as the ependyma, viral titer, infiltrating leukocyte populations, and key anti-viral cytokine message levels were evaluated. We found that CXCR3(-/-) mice possessed significantly less HSV-1 and expressed significantly more IFN-ß mRNA in the brain ependyma compared to WT animals during the development of encephalitis.


Subject(s)
Brain/pathology , Ependyma/metabolism , Gene Expression Regulation/genetics , Herpes Simplex/genetics , Herpes Simplex/pathology , Receptors, CXCR3/deficiency , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Disease Models, Animal , Ependyma/virology , Female , Flow Cytometry , Herpesvirus 1, Human , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Male , Mice , Mice, Knockout , Receptors, CXCR3/genetics , Viral Plaque Assay
19.
J Neuroinflammation ; 11: 109, 2014 Jun 16.
Article in English | MEDLINE | ID: mdl-24930935

ABSTRACT

BACKGROUND: The functional state of glial cells, like astrocytes and microglia, critically modulates the course of neuroinflammatory and neurodegenerative diseases and can have both detrimental and beneficial effects. Glial cell function is tightly controlled by cellular interactions in which cytokines are important messengers. Recent studies provide evidence that in particular chemokines are important modulators of glial cell function. During the course of CNS diseases like multiple sclerosis or Alzheimer's disease, and in the corresponding animal models, the chemokines CXCL9 and CXCL10 are abundantly expressed at sites of glial activation, arguing for an important role of these chemokines and their corresponding receptor CXCR3 in glial activation. To clarify the role of this chemokine system in glial cell activation, we characterized the impact of CXCR3 on glial activation in a model of toxic demyelination in which glial activation without a prominent influx of hematogenous cells is prototypical. METHODS: We investigated the impact of CXCR3 on cuprizone-induced demyelination, comparing CXCR3-deficient mice with wild type controls. The clinical course during cuprizone feeding was documented for five weeks and for the subsequent four days withdrawal of the cuprizone diet (5.5 weeks). Glial activation was characterized using histological, histomorphometric and phenotypic analysis. Molecular analysis for (de)myelination and neuroinflammation was applied to characterize the effect of cuprizone on CXCR3-deficient mice and control animals. RESULTS: CXCR3-deficient mice displayed a milder clinical course during cuprizone feeding and a more rapid body weight recovery after offset of diet. In the CNS, CXCR3 deficiency significantly attenuated the accumulation and activation of microglia and astrocytes. Moreover, a deficiency of CXCR3 reduced the expression of the microglial activation markers CD45 and CD11b. Compared to controls, we observed a vast reduction of RNA levels for proinflammatory cytokines and chemokines like Ccl2, Cxcl10, Tnf and Il6 within the CNS of cuprizone-treated mice. Lastly, CXCR3 deficiency had no major effects on the course of demyelination during cuprizone feeding. CONCLUSIONS: The CXCR3 chemokine system is critically involved in the intrinsic glial activation during cuprizone-induced demyelination, which significantly modulates the distribution of glial cells and the local cytokine milieu.


Subject(s)
Cuprizone/toxicity , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Monoamine Oxidase Inhibitors/toxicity , Neuroglia/metabolism , Receptors, CXCR3/genetics , Analysis of Variance , Animals , Antigens, CD/metabolism , Body Weight/drug effects , Cuprizone/administration & dosage , Cytokines/genetics , Cytokines/metabolism , Flow Cytometry , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monoamine Oxidase Inhibitors/administration & dosage , Neuroglia/drug effects , RNA, Messenger/metabolism , Receptors, CXCR3/deficiency
20.
Arterioscler Thromb Vasc Biol ; 34(7): 1374-81, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24812325

ABSTRACT

OBJECTIVE: Obesity associates with increased numbers of inflammatory cells in adipose tissue (AT), including T cells, but the mechanism of T-cell recruitment remains unknown. This study tested the hypothesis that the chemokine (C-X-C motif) receptor 3 (CXCR3) participates in T-cell accumulation in AT of obese mice and thus in the regulation of local inflammation and systemic metabolism. APPROACH AND RESULTS: Obese wild-type mice exhibited higher mRNA expression of CXCR3 in periepididymal AT-derived stromal vascular cells compared with lean mice. We evaluated the function of CXCR3 in AT inflammation in vivo using CXCR3-deficient and wild-type control mice that consumed a high-fat diet. Periepididymal AT from obese CXCR3-deficient mice contained fewer T cells than obese controls after 8 and 16 weeks on high-fat diet, as assessed by flow cytometry. Obese CXCR3-deficient mice had greater glucose tolerance than obese controls after 8 weeks, but not after 16 weeks. CXCR3-deficient mice fed high-fat diet had reduced mRNA expression of proinflammatory mediators, such as monocyte chemoattractant protein-1 and regulated on activation, normal T cell expressed and secreted, and anti-inflammatory genes, such as Foxp3, IL-10, and arginase-1 in periepididymal AT, compared with obese controls. CONCLUSIONS: These results demonstrate that CXCR3 contributes to T-cell accumulation in periepididymal AT of obese mice. Our results also suggest that CXCR3 regulates the accumulation of distinct subsets of T cells and that the ratio between these functional subsets across time likely modulates local inflammation and systemic metabolism.


Subject(s)
Adipose Tissue/immunology , Chemotaxis, Leukocyte , Obesity/immunology , Panniculitis/immunology , Receptors, CXCR3/metabolism , T-Lymphocyte Subsets/immunology , Adipose Tissue/metabolism , Animals , Disease Models, Animal , Gene Expression Regulation , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/metabolism , Panniculitis/genetics , Panniculitis/metabolism , Receptors, CXCR3/deficiency , Receptors, CXCR3/genetics , Signal Transduction , T-Lymphocyte Subsets/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...