Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 241
Filter
1.
Cell ; 172(5): 1022-1037.e14, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29429633

ABSTRACT

Conventional type 1 dendritic cells (cDC1) are critical for antitumor immunity, and their abundance within tumors is associated with immune-mediated rejection and the success of immunotherapy. Here, we show that cDC1 accumulation in mouse tumors often depends on natural killer (NK) cells that produce the cDC1 chemoattractants CCL5 and XCL1. Similarly, in human cancers, intratumoral CCL5, XCL1, and XCL2 transcripts closely correlate with gene signatures of both NK cells and cDC1 and are associated with increased overall patient survival. Notably, tumor production of prostaglandin E2 (PGE2) leads to evasion of the NK cell-cDC1 axis in part by impairing NK cell viability and chemokine production, as well as by causing downregulation of chemokine receptor expression in cDC1. Our findings reveal a cellular and molecular checkpoint for intratumoral cDC1 recruitment that is targeted by tumor-derived PGE2 for immune evasion and that could be exploited for cancer therapy.


Subject(s)
Dendritic Cells/immunology , Killer Cells, Natural/immunology , Neoplasms/immunology , Neoplasms/pathology , Tumor Microenvironment/immunology , Animals , Cell Line, Tumor , Chemokine CCL5/metabolism , Chemokines, C/metabolism , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Gene Expression Regulation, Neoplastic , Humans , Melanoma/genetics , Melanoma/pathology , Mice , Mutation/genetics , Prognosis , Proto-Oncogene Proteins B-raf/genetics , Survival Analysis
2.
Trends Biochem Sci ; 46(6): 433-434, 2021 06.
Article in English | MEDLINE | ID: mdl-33752957

ABSTRACT

In a recent study, Dishman et al. resurrected ancestors of the metamorphic chemokine, XCL1, inferred through phylogenetics, and found that metamorphism arose in the XCL1 lineage ~150 million years ago. A zigzagging evolutionary path suggests that the metamorphic properties are adaptive and reveals three design principles that could be used for technological applications.


Subject(s)
Chemokines, C
3.
Int J Cancer ; 154(12): 2176-2188, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38346928

ABSTRACT

Conventional type 1 dendritic cells (cDC1s) play a crucial role in antitumor immunity through the induction and activation of tumor-specific CD8+ cytotoxic T cells (CTLs). The chemokine XCL1 is a major chemotactic factor for cDC1s and its receptor XCR1 is selectively expressed on cDC1s. Here, we investigated the effect of intratumoral delivery of a highly active form of murine XCL1 (mXCL1-V21C/A59C) on cDC1-mediated antitumor immunity using a hydrophilic gel patch. The hydrophilic gel patch containing mXCL1-V21C/A59C increased cDC1 accumulation in the tumor masses and promoted their migration to the regional lymph nodes, resulting in enhanced induction of tumor-specific CTLs. Tumor-infiltrating cDC1s not only expressed XCR1 but also produced CXCL9, a ligand for CXCR3 which is highly expressed on CTLs and NK cells. Consequently, CTLs and NK cells were increased in the tumor masses of mice treated with mXCL1-V21C/A59C, while immunosuppressive cells such as monocyte-derived suppressive cells and regulatory T cells were decreased. We also confirmed that anti-CXCL9 treatment decreased the tumor infiltration of CTLs. The intratumoral delivery of mXCL1-V21C/A59C significantly decreased tumor growth and prolonged survival in E.G7-OVA and B16-F10 tumor-bearing mice. Furthermore, the antitumor effect of mXCL1-V21CA59C was enhanced in combination with anti-programmed cell death protein 1 treatment. Finally, using The Cancer Genome Atlas database, we found that XCL1 expression was positively correlated with tumor-infiltrating cDC1s and a better prognosis in melanoma patients. Collectively, our findings provide a novel therapeutic approach to enhance tumor-specific CTL responses through the selective recruitment of CXCL9-expressing cDC1s into the tumor masses.


Subject(s)
Chemokines, C , Melanoma , Humans , Mice , Animals , T-Lymphocytes, Cytotoxic , Killer Cells, Natural , Melanoma/metabolism , Dendritic Cells , CD8-Positive T-Lymphocytes , Chemokine CXCL9/metabolism , Chemokines, C/genetics
4.
Brain Behav Immun ; 118: 178-189, 2024 May.
Article in English | MEDLINE | ID: mdl-38428650

ABSTRACT

Elevated levels of cytokines in maternal circulation increase the offspring's risk for neuropsychiatric disease. Because of their low homeostatic levels, circulating maternal cytokines during normal pregnancies have not been considered to play a role in fetal brain development and offspring behavior. Here we report that the T/NK cell chemotactic cytokine XCL1, a local paracrine immune signal, can function as a pregnancy hormone and is required for the proper development of placenta and male offspring approach-avoidance behavior. We found that circulating XCL1 levels were at a low pregestational level throughout pregnancy except for a midgestational rise and fall. Blunted elevation in maternal plasma XCL1 in dams with a genetic 5HT1A receptor deficit or following neutralization by anti-XCL1 antibodies increased the expression of tissue damage associated factors in WT fetal placenta and led to increased innate anxiety and stress reactivity in the WT male offspring. Therefore, chemokines like XCL1 may act as pregnancy hormones to regulate placenta development and offspring emotional behavior.


Subject(s)
Anxiety , Chemokines, C , Female , Male , Pregnancy , Chemokines, C/genetics , Cytokines/metabolism , Hormones
5.
Mycoses ; 67(1): e13680, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38214420

ABSTRACT

CIITA, a member of NOD-like receptor (NLR) family, is the major MHC II trans-activator and mediator of Th1 immunity, but its function and interaction with NLRP3 have been little studied. We found activation of NLRP3 inflammasome, increased expression of CIITA, CBP, pSTAT1, STAT1, MHC II, IFN-γ and IFN-γ-inducible chemokines (CCL1 and CXCL8), and colocalisation of NLRP3 with CIITA in Malassezia folliculitis lesions, Malassezia globosa-infected HaCaT cells and mouse skin. CoIP with anti-CIITA or anti-NLRP3 antibody pulled down NLRP3 or both CIITA and ASC. NLRP3 silencing or knockout caused CIITA downexpression and their colocalisation disappearance in HaCaT cells and mouse skin of Nlrp3-/- mice, while CIITA knockdown had no effect on NLRP3, ASC, IL-1ß and IL-18 expression. NLRP3 inflammasome inhibitors and knockdown significantly suppressed IFN-γ, CCL1, CXCL8 and CXCL10 levels in M. globosa-infected HaCaT cells. CCL1 and CXCL8 expression was elevated in Malassezia folliculitis lesions and reduced in Nlrp3-/- mice. These results demonstrate that M. globosa can activate NLRP3 inflammasome, CIITA/MHC II signalling and IFN-γ-inducible chemokines in human keratinocytes and mouse skin. NLRP3 may regulate CIITA by their binding and trigger Th1 immunity by secreting CCL1 and CXCL8/IL-8, contributing to the pathogenesis of Malassezia-associated skin diseases.


Subject(s)
Chemokines, C , Folliculitis , Malassezia , Humans , Mice , Animals , Interferon-gamma , Interferons , Histocompatibility Antigens Class II/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Inflammasomes , Promoter Regions, Genetic , Trans-Activators/genetics , Trans-Activators/metabolism , Chemokines/genetics , Keratinocytes
6.
Microb Pathog ; 174: 105962, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36572194

ABSTRACT

The XCL1-XCR1 axis has a potential role in the recruitment of immune cells to the site of inflammation. The present study aimed to examine the relation of XCL1 serum levels with Multiple sclerosis (MS) and HTLV-1-associated myelopathy (HAM), as chronic inflammatory diseases of the central nervous system (CNS). DNA was extracted to evaluate HTLV-1 proviral load (PVL) using real-time PCR. Serum levels of XCL1 was determined by using an ELISA assay. The serum level of XCL1 was significantly higher in patients with HAM than that of asymptomatic carriers (ACs) and healthy controls (HCs) (p < 0.001 and p < 0.0001, respectively) and was also higher in MS patients compared to HCs (p < 0.0001). Moreover, the concentration of XCL1 serum level was significantly different between the ACs and HCs group (p < 0.0001). In conclusion, increased expression of XCL1 might contribute to the migration of autoreactive T cells to the central nervous system and play a critical role in the development and pathogenesis of inflammatory neurological diseases including HAM and MS.


Subject(s)
Chemokines, C , Human T-lymphotropic virus 1 , Multiple Sclerosis , Paraparesis, Tropical Spastic , Humans , Human T-lymphotropic virus 1/genetics , Biomarkers , Central Nervous System , Viral Load
7.
Scand J Immunol ; 95(3): e13128, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34923667

ABSTRACT

Targeting antigens to dendritic cells represent a promising method for enhancing immune responses against specific antigens. However, many studies have focused on systemic delivery (intravenous or intraperitoneally) of targeted antigen, approaches that are not easily transferable to humans. Here we evaluate the efficacy of an influenza vaccine targeting Xcr1+ cDC1 administered by intranasal immunization. Intranasal delivery of antigen fused to the chemokine Xcl1, the ligand of Xcr1, resulted in specific uptake by lung CD103+ cDC1. Interestingly, intranasal immunization with influenza A/PR/8/34 haemagglutinin (HA) fused to Xcl1, formulated with poly(I:C), resulted in enhanced induction of antigen-specific IFNγ+ CD4+ and IFNγ+ CD8+ T cell responses in lung compared non-targeted anti-NIP-HA (αNIP-HA). Induction of antibody responses was, however, similar in Xcl1-HA and αNIP-HA immunized mice, but significantly higher than in mice immunized with monomeric HA. Both Xcl1-HA and αNIP-HA vaccines induced full protection when mice were challenged with a lethal dose of influenza PR8 virus, reflecting the strong induction of HA-specific antibodies. Our results demonstrate that i.n. delivery of Xcl1-HA is a promising vaccine strategy for enhancing T cell responses in addition to inducing strong antibody responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chemokines, C/metabolism , Influenza Vaccines/immunology , Influenza, Human/immunology , Orthomyxoviridae Infections/immunology , Poly I-C/immunology , Animals , Antibodies, Viral/immunology , Antibody Formation/immunology , Antigens/immunology , Antigens, CD/immunology , Cell Line , Dendritic Cells/immunology , Dogs , Female , HEK293 Cells , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Integrin alpha Chains/immunology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C
8.
Int J Mol Sci ; 23(16)2022 Aug 10.
Article in English | MEDLINE | ID: mdl-36012168

ABSTRACT

The human C-C chemokine receptor type 7 (CCR7) has two endogenous ligands, C-C chemokine ligand 19 (CCL19) and CCL21, displaying biased agonism reflected by a pronounced difference in the level of ß-arrestin recruitment. Detecting this preferential activation generally requires the use of separate, pathway-specific label-based assays. In this study, we evaluated an alternative methodology to study CCR7 signalling. Cellular electrical impedance (CEI) is a label-free technology which yields a readout that reflects an integrated cellular response to ligand stimulation. CCR7-expressing HEK293 cells were stimulated with CCL19 or CCL21, which induced distinct impedance profiles with an apparent bias during the desensitisation phase of the response. This discrepancy was mainly modulated by differential ß-arrestin recruitment, which shaped the impedance profile but did not seem to contribute to it directly. Pathway deconvolution revealed that Gαi-mediated signalling contributed most to the impedance profile, but Gαq- and Gα12/13-mediated pathways were also involved. To corroborate these results, label-based pathway-specific assays were performed. While CCL19 more potently induced ß-arrestin2 recruitment and receptor internalisation than CCL21, both chemokines showed a similar level of Gαi protein activation. Altogether, these findings indicate that CEI is a powerful method to analyse receptor signalling and biased agonism.


Subject(s)
Chemokine CCL21 , Chemokines, C , Chemokine CCL19/metabolism , Chemokine CCL21/metabolism , Chemokines/metabolism , Chemokines, C/metabolism , Electric Impedance , HEK293 Cells , Humans , Ligands , Receptors, CCR7/metabolism , beta-Arrestins/metabolism
9.
Int J Mol Sci ; 23(21)2022 Oct 26.
Article in English | MEDLINE | ID: mdl-36361722

ABSTRACT

Acute respiratory distress syndrome (ARDS) consists of uncontrolled inflammation that causes hypoxemia and reduced lung compliance. Since it is a complex process, not all details have been elucidated yet. In a well-controlled experimental murine model of lipopolysaccharide (LPS)-induced ARDS, the activity and viability of macrophages and neutrophils dictate the beginning and end phases of lung inflammation. C-C chemokine receptor type 2 (CCR2) is a critical chemokine receptor that mediates monocyte/macrophage activation and recruitment to the tissues. Here, we used CCR2-deficient mice to explore mechanisms that control lung inflammation in LPS-induced ARDS. CCR2-/- mice presented higher total numbers of pulmonary leukocytes at the peak of inflammation as compared to CCR2+/+ mice, mainly by enhanced influx of neutrophils, whereas we observed two to six-fold lower monocyte or interstitial macrophage numbers in the CCR2-/-. Nevertheless, the time needed to control the inflammation was comparable between CCR2+/+ and CCR2-/-. Interestingly, CCR2-/- mice presented higher numbers and increased proliferative rates of alveolar macrophages from day 3, with a more pronounced M2 profile, associated with transforming growth factor (TGF)-ß and C-C chemokine ligand (CCL)22 production, decreased inducible nitric oxide synthase (Nos2), interleukin (IL)-1ß and IL-12b mRNA expression and increased mannose receptor type 1 (Mrc1) mRNA and CD206 protein expression. Depletion of alveolar macrophages significantly delayed recovery from the inflammatory insult. Thus, our work shows that the lower number of infiltrating monocytes in CCR2-/- is partially compensated by increased proliferation of resident alveolar macrophages during the inflammation control of experimental ARDS.


Subject(s)
Chemokines, C , Pneumonia , Respiratory Distress Syndrome , Mice , Animals , Receptors, Chemokine , Macrophages, Alveolar/metabolism , Lipopolysaccharides/pharmacology , Inflammation , RNA, Messenger , Cell Proliferation , Receptors, CCR2/genetics , Mice, Inbred C57BL , Chemokine CCL2/metabolism
10.
J Cell Sci ; 133(5)2019 11 18.
Article in English | MEDLINE | ID: mdl-31649144

ABSTRACT

Dendritic cells (DCs) expressing the chemokine receptor XCR1 are specialized in antigen cross-presentation to control infections with intracellular pathogens. XCR1-positive (XCR1+) DCs are attracted by XCL1, a γ-chemokine secreted by activated CD8+ T cells and natural killer cells. Rat cytomegalovirus (RCMV) is the only virus known to encode a viral XCL1 analog (vXCL1) that competes for XCR1 binding with the endogenous chemokine. Here we show that vXCL1 from two different RCMV strains, as well as endogenous rat XCL1 (rXCL1) bind to and induce chemotaxis exclusively in rat XCR1+ DCs. Whereas rXCL1 activates the XCR1 Gi signaling pathway in rats and humans, both of the vXCL1s function as species-specific agonists for rat XCR1. In addition, we demonstrate constitutive internalization of XCR1 in XCR1-transfected HEK293A cells and in splenic XCR1+ DCs. This internalization was independent of ß-arrestin 1 and 2 and was enhanced after binding of vXCL1 and rXCL1; however, vXCL1 appeared to be a stronger agonist. These findings suggest a decreased surface expression of XCR1 during DC cultivation at 37°C, and subsequent impairment of chemotactic activity and XCR1+ DC function.


Subject(s)
Chemokines, C/metabolism , Cross-Priming , Dendritic Cells/immunology , Muromegalovirus/immunology , Receptors, Chemokine/metabolism , Animals , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Chemotaxis , Killer Cells, Natural/immunology , Rats , Receptors, G-Protein-Coupled/metabolism
11.
Biopolymers ; 112(10): e23402, 2021 Oct.
Article in English | MEDLINE | ID: mdl-32986858

ABSTRACT

The metamorphic protein XCL1 switches between two distinct native structures with different functions in the human immune system. This structural interconversion requires complete rearrangement of all hydrogen bonding networks, yet fold-switching occurs spontaneously and reversibly in solution. One structure occupies the canonical α-ß chemokine fold and binds XCL1's cognate G-protein coupled receptor, while the other structure occupies a dimeric, all-ß fold that binds glycosaminoglycans and has antimicrobial activity. Both of these functions are important for the biologic role of XCL1 in the immune system, and each structure is approximately equally populated under near-physiologic conditions. Recent work has begun to illuminate XCL1's role in combatting infection and cancer. However, without a way to control XCL1's dynamic structural interconversion, it is difficult to study the role of XCL1 fold-switching in human health and disease. Thus, a molecular tool that can regulate the fractional population of the two XCL1 structures is needed. Here, we find by heparin affinity chromatography and NMR that an engineered XCL1 variant called CC5 can trigger a dose-dependent shift in XCL1's metamorphic equilibrium such that the receptor binding structure is depleted, and the antimicrobial structure is more heavily populated. This shift likely occurs due to formation of XCL1-CC5 heterodimers in which both protomers occupy the ß-sheet structure. These findings lay the groundwork for future studies seeking to understand the functional role of XCL1 metamorphosis, as well as studies screening for a drug-like molecule that can therapeutically target XCL1 by tuning its metamorphic equilibrium. Moreover, the proof of concept presented here suggests that protein metamorphosis is druggable, opening numerous avenues for controlling biological function of metamorphic proteins by altering the population of their multiple native states.


Subject(s)
Chemokines, C , Chemokines, C/metabolism , Glycosaminoglycans , Heparin , Humans , Protein Binding , Receptors, G-Protein-Coupled/metabolism
12.
J Immunol ; 203(10): 2621-2629, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31578268

ABSTRACT

Oral tolerance is defined as the specific suppression of cellular and/or humoral immune responses to an Ag by prior administration of the Ag through the oral route. Although the investigation of oral tolerance has classically involved Ag feeding, we have found that oral administration of anti-CD3 mAb induced tolerance through regulatory T (Treg) cell generation. However, the mechanisms underlying this effect remain unknown. In this study, we show that conventional but not plasmacytoid dendritic cells (DCs) are required for anti-CD3-induced oral tolerance. Moreover, oral anti-CD3 promotes XCL1 secretion by small intestine lamina propria γδ T cells that, in turn, induces tolerogenic XCR1+ DC migration to the mesenteric lymph node, where Treg cells are induced and oral tolerance is established. Consistent with this, TCRδ-/- mice did not develop oral tolerance upon oral administration of anti-CD3. However, XCL1 was not required for oral tolerance induced by fed Ags, indicating that a different mechanism underlies this effect. Accordingly, oral administration of anti-CD3 enhanced oral tolerance induced by fed MOG35-55 peptide, resulting in less severe experimental autoimmune encephalomyelitis, which was associated with decreased inflammatory immune cell infiltration in the CNS and increased Treg cells in the spleen. Thus, Treg cell induction by oral anti-CD3 is a consequence of the cross-talk between γδ T cells and tolerogenic DCs in the gut. Furthermore, anti-CD3 may serve as an adjuvant to enhance oral tolerance to fed Ags.


Subject(s)
CD3 Complex/immunology , Chemokines, C/metabolism , Immune Tolerance/drug effects , Intraepithelial Lymphocytes/immunology , Muromonab-CD3/administration & dosage , Muromonab-CD3/pharmacology , Administration, Oral , Animals , Cell Movement/immunology , Dendritic Cells/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Gene Knockout Techniques , Genes, T-Cell Receptor delta/genetics , Intestinal Mucosa/immunology , Lymph Nodes/immunology , Male , Mesentery , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/pharmacology , Peptide Fragments/pharmacology , T-Lymphocytes, Regulatory/immunology
13.
Int J Cancer ; 146(8): 2182-2193, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31904872

ABSTRACT

Most genome-wide association studies (GWASs) identify genetic variants for breast cancer occurrence. In contrast, few are for recurrence and mortality. We conducted a GWAS on breast cancer survival after diagnosis in estrogen receptor-positive patients, including 953 Taiwanese patients with 159 events. Through Cox proportional hazard models estimation, we identified 24 risk SNPs with p < 1 × 10-5 . Based on imputation and integrated analysis, one SNP, rs1024176 (located in 1q24.2, p = 2.43 × 10-5 ) was found to be a functional variant associated with breast cancer survival and XCL1 gene expression. A series of experimental approaches, including cell-based analyses and CRISPR/Cas9 genome-editing system, were then used and identified the transcription factor MYBL2 was able to discriminately bind to the A allele of rs1024176, the protective variant for breast cancer survival, which promoted XCL1 expression, but not to the G allele of rs1024176. The chemokine XCL1 attracts type 1 dendritic cells (DC1s) to the tumor microenvironment. In breast cancer tissues, we applied a two-step Mendelian randomization analysis, using expression quantitative trait loci as instrumental variables, to confirm higher XCL1 expression was correlated with higher DC1 signatures and favorable disease progression, through the causal effect of rs1024176-A allele. Our study supports the genetic effect on preventing breast cancer survival through XCL1-induced DC1 recruitment in tumor microenvironment.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/immunology , Chemokines, C/genetics , Chemokines, C/immunology , Adult , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/immunology , Chemokines, C/biosynthesis , Dendritic Cells/immunology , Dendritic Cells/pathology , Female , Gene Expression Regulation, Neoplastic , Genome-Wide Association Study , Humans , Middle Aged , Proportional Hazards Models , Quantitative Trait Loci , Trans-Activators/genetics , Trans-Activators/immunology , Young Adult
14.
Br J Cancer ; 122(8): 1185-1193, 2020 04.
Article in English | MEDLINE | ID: mdl-32066911

ABSTRACT

BACKGROUND: Cancer peptide vaccines show only marginal effects against cancers. Immune checkpoint inhibitors (ICIs) show significant curative effects in certain types of cancers, but the response rate is still limited. In this study, we aim to improve cancer peptide vaccination by targeting Ag peptides selectively to a dendritic cell (DC) subset, XCR1-expressing DCs (XCR1+ DCs), with high ability to support CD8+ T-cell responses. METHODS: We have generated a fusion protein, consisting of an Ag peptide presented with MHC class I, and an XCR1 ligand, XCL1, and examined its effects on antitumour immunity in mice. RESULTS: The fusion protein was delivered to XCR1+ DCs in an XCR1-dependent manner. Immunisation with the fusion protein plus an immune adjuvant, polyinosinic:polycytidylic acids (poly(I:C)), more potently induced Ag-specific CD8+ T-cell responses through XCR1 than the Ag peptide plus poly(I:C) or the Ag protein plus poly(I:C). The fusion protein plus poly(I:C) inhibited the tumour growth efficiently in the prophylactic and therapeutic tumour models. Furthermore, the fusion protein plus poly(I:C) showed suppressive effects on tumour growth in synergy with anti-PD-1 Ab. CONCLUSIONS: Cancer Ag targeting to XCR1+ DCs should be a promising procedure as a combination anticancer therapy with immune checkpoint blockade.


Subject(s)
Antigens/immunology , Cancer Vaccines/immunology , Chemokines, C/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Immune Checkpoint Inhibitors/therapeutic use , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Mice, Inbred C57BL , Neoplasms, Experimental/therapy , Poly I-C/pharmacology , Vaccines, Subunit/immunology
15.
J Pharmacol Sci ; 143(3): 182-187, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32386904

ABSTRACT

Memory CD8+ cytotoxic T-lymphocytes (CTLs) play a key role in protective immunity against infection and cancer. However, the induction of memory CTLs with currently available vaccines remains difficult. The chemokine receptor XCR1 is predominantly expressed on CD103+ cross-presenting dendritic cells (DCs). Recently, we have demonstrated that a high activity form of murine lymphotactin/XCL1 (mXCL1-V21C/A59C), a ligand of XCR1, can induce antigen-specific memory CTLs by increasing the accumulation of CD103+ DCs in the vaccination site and the regional lymph nodes. Here, we combined a hydrophilic gel patch as a transcutaneous delivery device and mXCL1-V21C/A59C as an adjuvant to further enhance memory CTL responses. The transcutaneous delivery of ovalbumin (OVA) and mXCL1-V21C/A59C by the hydrophilic gel patch increased CD103+ DCs in the vaccination site and the regional lymph nodes for a prolonged period of time compared with the intradermal injection of OVA and mXCL1-V21C/A59C. Furthermore, the hydrophilic gel patch containing OVA and mXCL1-V21C/A59C strongly induced OVA-specific memory CTLs and efficiently inhibited the growth of OVA-expressing tumors more than the intradermal injection of OVA and mXCL1-V21C/A59C. Collectively, this type of hydrophilic gel patch and a high activity form of XCL1 may provide a useful tool for the induction of memory CTL responses.


Subject(s)
Adjuvants, Immunologic/administration & dosage , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Chemokines, C/administration & dosage , Chemokines, C/immunology , Immunization/methods , Transdermal Patch , Animals , Antigens, CD , Cell Line , Dendritic Cells/immunology , Gels , Hydrophobic and Hydrophilic Interactions , Integrin alpha Chains , Mice, Inbred C57BL , Ovalbumin/administration & dosage , Ovalbumin/immunology , Time Factors
16.
J Biochem Mol Toxicol ; 34(3): e22446, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31953984

ABSTRACT

Diepoxybutane (DEB) is the most potent active metabolite of the environmental chemical 1,3-butadiene (BD). BD is a human carcinogen that exhibits multiorgan systems toxicity. Our previous studies demonstrated that the X-C motif chemokine ligand 1 (XCL1) gene expression was upregulated 3.3-fold in a p53-dependent manner in TK6 lymphoblasts undergoing DEB-induced apoptosis. The tumor-suppressor p53 protein is a transcription factor that regulates a wide variety of cellular processes, including apoptosis, through its various target genes. Thus, the objective of this study was to determine whether XCL1 is a novel direct p53 transcriptional target gene and deduce its role in DEB-induced toxicity in human lymphoblasts. We utilized the bioinformatics tool p53scan to search for known p53 consensus sequences within the XCL1 promoter region. The XCL1 gene promoter region was found to contain the p53 consensus sequences 5'-AGACATGCCTAGACATGCCT-3' at three positions relative to the transcription start site (TSS). Furthermore, the XCL1 promoter region was found, through reporter gene assays, to be transactivated at least threefold by wild-type p53 promoter in DEB-exposed human lymphoblasts. Inactivation of the XCL1 promoter p53-binding motif located at -2.579 kb relative to TSS reduced the transactivation function of p53 on this promoter in DEB-exposed cells by 97%. Finally, knockdown of XCL1 messenger RNA with specific small interfering RNA inhibited DEB-induced apoptosis in human lymphoblasts by 50%. These observations demonstrate, for the first time, that XCL1 is a novel DEB-induced direct p53 transcriptional target gene that mediates apoptosis in DEB-exposed human lymphoblasts.


Subject(s)
Apoptosis/drug effects , Chemokines, C/biosynthesis , Epoxy Compounds/toxicity , Lymphocytes/metabolism , Response Elements , Transcriptional Activation/drug effects , Tumor Suppressor Protein p53/metabolism , Cell Line , Humans , Lymphocytes/pathology , Transcription, Genetic/drug effects
17.
Oral Dis ; 26(8): 1668-1676, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32562323

ABSTRACT

BACKGROUND: The expression of XCR1 receptor and its metamorphic ligand lymphotactin (hLtn) has been shown in cancers but their precise role in tumorigenesis is poorly understood including the significance of the physiologically existing hLtn monomeric (CC3) and dimeric (W55D) confirmations where the latter thought to function as the receptor antagonist. The aim of this study was to explore the functional role of bioengineered hLtn variants and the role of fibroblasts in XCR1/hLtn expression regulation in oral cancer cells (OCCL). MATERIAL AND METHODS: qRT-PCR and flow cytometry were performed to evaluate mRNA and protein expression of XCR1 and hLtn. Recombinant hLtn variants (wild-type, CC3 and W55D mutant) were designed, expressed, purified and evaluated using proliferation, adhesion and chemotaxis assays. XCR1 and hLtn expression regulation by fibroblasts was determined using indirect co-culture. XCR1 and hLtn expression in primary and metastatic OSCC tissue was assessed using immunohistochemistry. RESULTS: hLtn caused a significant decrease in OCCL XCR1 surface protein expression. hLtn CC3 mutant was highly functional facilitating proliferation and migration. Conditioned media from primary cancer-associated and senescent fibroblasts significantly upregulated XCR1 and hLtn mRNA expression in OCCL. Immunohistochemistry revealed higher XCR1 and hLtn expression in metastatic tumour deposits and surrounding stroma compared to primary OSCC tissue. CONCLUSIONS: The development of hLtn biological mutants, regulation of XCR1 expression by its ligand hLtn and crosstalk with fibroblasts are novel findings suggesting an important role for the XCR1/hLtn axis within the OSCC tumour microenvironment. These discoveries build upon previous studies and suggest that the hLtn/XCR1 axis has a significant role in stromal crosstalk and OSCC progression.


Subject(s)
Chemokines, C , Mouth Neoplasms , Cell Movement , Chemokines , Chemokines, C/genetics , Humans , Lymphokines , Mouth Neoplasms/genetics , Sialoglycoproteins , Tumor Microenvironment
18.
Int J Mol Sci ; 22(1)2020 Dec 23.
Article in English | MEDLINE | ID: mdl-33374849

ABSTRACT

Chemokine-receptor interactions play multiple roles in cancer progression. It was reported that the overexpression of X-C motif chemokine receptor 1 (XCR1), a specific receptor for chemokine X-C motif chemokine ligand 1 (XCL1), stimulates the migration of MDA-MB-231 triple-negative breast cancer cells. However, the exact mechanisms of this process remain to be elucidated. Our study found that XCL1 treatment markedly enhanced MDA-MB-231 cell migration. Additionally, XCL1 treatment enhanced epithelial-mesenchymal transition (EMT) of MDA-MB-231 cells via E-cadherin downregulation and upregulation of N-cadherin and vimentin as well as increases in ß-catenin nucleus translocation. Furthermore, XCL1 enhanced the expression of hypoxia-inducible factor-1α (HIF-1α) and phosphorylation of extracellular signal-regulated kinase (ERK) 1/2. Notably, the effects of XCL1 on cell migration and intracellular signaling were negated by knockdown of XCR1 using siRNA, confirming XCR1-mediated actions. Treating MDA-MB-231 cells with U0126, a specific mitogen-activated protein kinase kinase (MEK) 1/2 inhibitor, blocked XCL1-induced HIF-1α accumulation and cell migration. The effect of XCL1 on cell migration was also evaluated in ER-/HER2+ SK-BR-3 cells. XCL1 also promoted cell migration, EMT induction, HIF-1α accumulation, and ERK phosphorylation in SK-BR-3 cells. While XCL1 did not exhibit any significant impact on the matrix metalloproteinase (MMP)-2 and -9 expressions in MDA-MB-231 cells, it increased the expression of these enzymes in SK-BR-3 cells. Collectively, our results demonstrate that activation of the ERK/HIF-1α/EMT pathway is involved in the XCL1-induced migration of both MDA-MB-231 and SK-BR-3 breast cancer cells. Based on our findings, the XCL1-XCR1 interaction and its associated signaling molecules may serve as specific targets for the prevention of breast cancer cell migration and metastasis.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement , Chemokines, C/metabolism , Signal Transduction , A549 Cells , Epithelial-Mesenchymal Transition , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Receptors, G-Protein-Coupled/metabolism
19.
J Immunol ; 199(1): 82-90, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28550205

ABSTRACT

The integrin α9ß1 is a key receptor involved in the development of autoimmune diseases. However, the detailed mechanism for the association of α9ß1 integrin with its ligands remains unclear. In this study, we introduce XCL1/lymphotactin, a member of the chemokine family, as a novel ligand for α9 integrin. Using α9 integrin-overexpressing NIH3T3 cells and endogenously α9 integrin-expressing human rhabdomyosarcoma cells, the interaction between XCL1 and α9 integrin was confirmed by pull-down assays. XCL1 enhanced α9 integrin-dependent cell migration of these cells, thus acting on α9 integrin as a chemoattractant. We also analyzed the in vivo function of XCL1 in the development of anti-type II collagen Ab-induced inflammatory arthritis (CAIA) in BALB/c mice and experimental autoimmune encephalomyelitis in C57BL/6 mice, because α9 integrin is involved in these autoimmune disease models. In CAIA, recombinant XCL1 aggravated the disease and this exacerbation was inhibited by an anti-α9 integrin Ab. An XCL1-neutralizing Ab produced in this study also ameliorated CAIA. Furthermore, the XCL1-neutralizing Ab abrogated the disease progression in experimental autoimmune encephalomyelitis. Therefore, to our knowledge this study provides the first in vitro and in vivo evidence that the interaction between XCL1 and α9 integrin has an important role for autoimmune diseases.


Subject(s)
Chemokines, C/immunology , Chemokines, C/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Integrin alpha Chains/metabolism , Animals , Antibodies, Neutralizing/immunology , Arthritis, Experimental/immunology , Cell Adhesion , Cell Movement , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Encephalomyelitis, Autoimmune, Experimental/therapy , Integrin alpha Chains/immunology , Ligands , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Rhabdomyosarcoma/immunology
20.
J Allergy Clin Immunol ; 142(6): 1781-1792.e12, 2018 12.
Article in English | MEDLINE | ID: mdl-29474842

ABSTRACT

BACKGROUND: The chemokine X-C motif chemokine ligand 1 (XCL1)-X-C motif chemokine receptor 1 (XCR1) axis has been reported to play a role in immune homeostasis and inflammation. However, it is not known whether this axis has a critical function in patients with allergic asthma. OBJECTIVE: In the present study we explored whether the invariant natural killer T (iNKT) cell-mediated XCL1-XCR1 axis regulated allergic asthma. METHODS: Ovalbumin (OVA)- or house dust mite-induced asthma was developed in XCL1 or XCR1 knockout (KO) mice. RESULTS: XCL1 or XCR1 KO mice showed attenuation in airway hyperresponsiveness (AHR), numbers of CD103+ dendritic cells (DCs), and TH2 responses in the lungs compared with wild-type (WT) mice during OVA- or house dust mite-induced asthma. These effects were reversed by intratracheal administration of recombinant XCL1 or adoptive transfer of CD103+ DCs but not CD11b+ DCs into XCL1 KO mice. Moreover, iNKT cells highly expressed XCL1 both in vitro and in vivo. On intranasal α-galactosyl ceramide challenge, CD103+ DC numbers in the lungs were increased in WT but not XCL1 KO mice. Furthermore, adoptive transfer of WT iNKT cells increased AHR, CD103+ DC recruitment, and TH2 responses in the lungs of CD1d KO mice during OVA-induced asthma, whereas adoptive transfer of XCL1-deficient iNKT cells did not. In human patients, percentages and XCL1 production capacity of iNKT cells from PBMCs were greater in patients with asthma than in healthy control subjects. CONCLUSION: These data demonstrate that the iNKT cell-mediated XCL1-XCR1 axis promotes AHR by recruiting CD103+ DCs into the lung in patients with allergic asthma.


Subject(s)
Asthma/immunology , Chemokines, C/immunology , Dendritic Cells/immunology , Natural Killer T-Cells/immunology , Receptors, Chemokine/immunology , Adult , Aged , Animals , Antigens, CD/immunology , Asthma/physiopathology , Carcinoma, Non-Small-Cell Lung/immunology , Chemokines, C/genetics , Female , Humans , Integrin alpha Chains/immunology , Lung Neoplasms/immunology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Receptors, Chemokine/genetics , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL