Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.306
Filter
1.
Eur J Immunol ; 53(2): e2249985, 2023 02.
Article in English | MEDLINE | ID: mdl-36427489

ABSTRACT

Flagellin-induced NAIP/NLRC4 inflammasome activation and pyroptosis are critical events restricting Legionella pneumophila infection. However, the cellular and molecular dynamics of the in vivo responses against this bacterium are still unclear. We have found temporal coordination of two independent innate immunity pathways in controlling Legionella infection, the inflammasome activation and the CCR2-mediated Mo-DC recruitment. Inflammasome activation was an important player at the early stage of infection by lowering the numbers of bacteria for an efficient bacterial clearance conferred by the Mo-DC at the late stage of the infection. Mo-DC emergence highly depended on CCR2-signaling and dispensed inflammasome activation and pyroptosis. Also, Mo-DC compartment did not rely on the inflammasome machinery to deliver proper immune responses and was the most abundant cytokine-producing among the monocyte-derived cells in the infected lung. Importantly, when the CCR2- and NLRC4-dependent axes of response were simultaneously ablated, we observed an aggravated bacterial burden in the lung of infected mice. Taken together, we showed that inflammasome activation and CCR2-mediated immune response interplay in distinct pathways to restrict pulmonary bacterial infection. These findings extend our understanding of the in vivo integration and cooperation of different innate immunity arms in controlling infectious agents.


Subject(s)
Dendritic Cells , Inflammasomes , Legionella pneumophila , Legionnaires' Disease , Monocytes , Animals , Mice , Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins/metabolism , Chemotaxis, Leukocyte/genetics , Chemotaxis, Leukocyte/immunology , Dendritic Cells/metabolism , Inflammasomes/genetics , Inflammasomes/metabolism , Legionella pneumophila/immunology , Legionnaires' Disease/genetics , Legionnaires' Disease/immunology , Macrophages , Mice, Knockout , Monocytes/metabolism , Receptors, CCR2/metabolism
2.
J Exp Med ; 219(3)2022 03 07.
Article in English | MEDLINE | ID: mdl-35195682

ABSTRACT

Leukocyte trafficking between blood and tissues is an essential function of the immune system that facilitates humoral and cellular immune responses. Within tissues, leukocytes perform surveillance and effector functions via cell motility and migration toward sites of tissue damage, infection, or inflammation. Neurotransmitters that are produced by the nervous system influence leukocyte trafficking around the body and the interstitial migration of immune cells in tissues. Neural regulation of leukocyte dynamics is influenced by circadian rhythms and altered by stress and disease. This review examines current knowledge of neuro-immune interactions that regulate leukocyte migration and consequences for protective immunity against infections and cancer.


Subject(s)
Leukocytes/immunology , Neuroimmunomodulation/immunology , Cell Movement/immunology , Chemotaxis, Leukocyte/immunology , Circadian Rhythm/immunology , Humans , Models, Immunological , Models, Neurological , Neural Pathways/immunology , Sympathetic Nervous System/immunology , Tumor Microenvironment/immunology
3.
Elife ; 112022 02 03.
Article in English | MEDLINE | ID: mdl-35112667

ABSTRACT

Infection is a major co-morbidity that contributes to impaired healing in diabetic wounds. Although impairments in diabetic neutrophils have been blamed for this co-morbidity, what causes these impairments and whether they can be overcome, remain largely unclear. Diabetic neutrophils, isolated from diabetic individuals, exhibit chemotaxis impairment but this peculiar functional impairment has been largely ignored because it appears to contradict the clinical findings which blame excessive neutrophil influx as a major impediment to healing in chronic diabetic ulcers. Here, we report that exposure to glucose in diabetic range results in impaired chemotaxis signaling through the formyl peptide receptor (FPR) in neutrophils, culminating in reduced chemotaxis and delayed neutrophil trafficking in the wound of Leprdb (db/db) type two diabetic mice, rendering diabetic wound vulnerable to infection. We further show that at least some auxiliary receptors remain functional under diabetic conditions and their engagement by the pro-inflammatory cytokine CCL3, overrides the requirement for FPR signaling and substantially improves infection control by jumpstarting the neutrophil trafficking toward infection, and stimulates healing in diabetic wound. We posit that CCL3 may have therapeutic potential for the treatment of diabetic foot ulcers if it is applied topically after the surgical debridement process which is intended to reset chronic ulcers into acute fresh wounds.


Subject(s)
Chemotaxis, Leukocyte/immunology , Diabetes Mellitus, Experimental/immunology , Neutrophils/pathology , Receptors, Formyl Peptide/genetics , Signal Transduction/immunology , Wound Healing/immunology , Wound Infection/microbiology , Animals , Chemokine CCL3/immunology , Diabetes Complications/microbiology , Glucose/administration & dosage , Male , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Receptors, Formyl Peptide/immunology , Wound Infection/drug therapy , Wound Infection/etiology
4.
Cancer Lett ; 529: 112-125, 2022 03 31.
Article in English | MEDLINE | ID: mdl-34999169

ABSTRACT

Though circulating monocytes are the main source of tumour-associated macrophages (TAMs), the regulatory mechanisms of their recruitment to tumours and further differentiation remain unclear. In the present study, we observed a significant decrease in CXCR2 expression in classical circulating monocytes of patients with colorectal cancer (CRC), particularly those in the late TNM stage. The percentage of CXCR2+ monocytes was negatively associated with systemic inflammatory markers and positively associated with intratumoural immunocyte infiltration. The pro-inflammatory cytokine IFN-γ, which was overexpressed in patients with CRC, down-regulated CXCR2 expression of monocytes/TAMs by promoting GRK-2 expression. In vitro, inhibition of CXCR2 signalling in monocytes led to impaired chemotaxis to the tumour cell line supernatant and lower responsiveness to lipopolysaccharide (LPS) stimulation. Finally, monocytes from patients with CRC with decreased CXCR2 expression showed distinct phenotypes and functions after differentiating into CRC cell line-educated TAMs, including expression of co-stimulatory factors and secretion profile, than those from healthy controls. GRK-2 inhibitor altered the functional characteristics of TAMs. In summary, our findings suggest that CXCR2 expression on circulating monocytes reflects CRC stages and is an important factor determining TAM composition in the tumour microenvironment.


Subject(s)
Chemotaxis, Leukocyte/genetics , Monocytes/metabolism , Receptors, Interleukin-8B/genetics , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Aged , Aged, 80 and over , Biomarkers , Cell Line, Tumor , Chemotaxis, Leukocyte/immunology , Colorectal Neoplasms/etiology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Gene Expression , Gene Knockdown Techniques , Humans , Immunophenotyping , Male , Middle Aged , Monocytes/immunology , Neoplasm Staging , Receptors, Interleukin-8B/metabolism , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
5.
Infect Immun ; 90(1): e0051621, 2022 01 25.
Article in English | MEDLINE | ID: mdl-34662214

ABSTRACT

Salmonella invades and disrupts gut epithelium integrity, creating an infection-generated electric field that can drive directional migration of macrophages, a process called galvanotaxis. Phagocytosis of bacteria reverses the direction of macrophage galvanotaxis, implicating a bioelectrical mechanism to initiate life-threatening disseminations. The force that drives direction reversal of macrophage galvanotaxis is not understood. One hypothesis is that Salmonella can alter the electrical properties of the macrophages by modifying host cell surface glycan composition, which is supported by the fact that cleavage of surface-exposed sialic acids with a bacterial neuraminidase severely impairs macrophage galvanotaxis, as well as phagocytosis. Here, we utilize N-glycan profiling by nanoLC-chip QTOF mass cytometry to characterize the bacterial neuraminidase-associated compositional shift of the macrophage glycocalyx, which revealed a decrease in sialylated and an increase in fucosylated and high mannose structures. The Salmonella nanH gene, encoding a putative neuraminidase, is required for invasion and internalization in a human colonic epithelial cell infection model. To determine whether NanH is required for the Salmonella infection-dependent direction reversal, we constructed and characterized a nanH deletion mutant and found that NanH is partially required for Salmonella infection in primary murine macrophages. However, compared to wild type Salmonella, infection with the nanH mutant only marginally reduced the cathode-oriented macrophage galvonotaxis, without canceling direction reversal. Together, these findings strongly suggest that while neuraminidase-mediated N-glycan modification impaired both macrophage phagocytosis and galvanotaxis, yet to be defined mechanisms other than NanH may play a more important role in bioelectrical control of macrophage trafficking, which potentially triggers dissemination.


Subject(s)
Chemotaxis, Leukocyte/immunology , Macrophages/immunology , Macrophages/metabolism , Neuraminidase/metabolism , Salmonella Infections/immunology , Salmonella Infections/metabolism , Salmonella/physiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Female , Host-Pathogen Interactions/immunology , Male , Mice , Models, Biological , Mutation , Phagocytosis/immunology , Polysaccharides/metabolism , Salmonella Infections/microbiology , Sialic Acids/metabolism , Virulence
7.
Front Immunol ; 12: 775447, 2021.
Article in English | MEDLINE | ID: mdl-34858434

ABSTRACT

CD11d/CD18 is the most recently discovered and least understood ß2 integrin. Known CD11d adhesive mechanisms contribute to both extravasation and mesenchymal migration - two key aspects for localizing peripheral leukocytes to sites of inflammation. Differential expression of CD11d induces differences in monocyte/macrophage mesenchymal migration including impacts on macrophage sub-set migration. The participation of CD11d/CD18 in leukocyte localization during atherosclerosis and following neurotrauma has sparked interest in the development of CD11d-targeted therapeutic agents. Whereas the adhesive properties of CD11d have undergone investigation, the signalling pathways induced by ligand binding remain largely undefined. Underlining each adhesive and signalling function, CD11d is under unique transcriptional control and expressed on a sub-set of predominately tissue-differentiated innate leukocytes. The following review is the first to capture the nearly three decades of CD11d research and discusses the emerging role of CD11d in leukocyte migration and retention during the progression of a staged immune response.


Subject(s)
CD11 Antigens/genetics , CD18 Antigens/genetics , Chemotaxis, Leukocyte/genetics , Chemotaxis, Leukocyte/immunology , Gene Expression Regulation , Integrin alpha Chains/genetics , Leukocytes/physiology , Animals , CD11 Antigens/chemistry , CD11 Antigens/metabolism , CD18 Antigens/chemistry , CD18 Antigens/metabolism , Disease Susceptibility , Drug Development , Humans , Integrin alpha Chains/chemistry , Integrin alpha Chains/metabolism , Lymphopoiesis/genetics , Molecular Targeted Therapy , Organ Specificity/genetics , Phagocytosis/genetics , Phagocytosis/immunology , Protein Binding , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational , Structure-Activity Relationship , Transcription Factors
8.
Int J Med Sci ; 18(16): 3631-3643, 2021.
Article in English | MEDLINE | ID: mdl-34790035

ABSTRACT

Background: Acyl-CoA dehydrogenase short-chain (ACADS) is a crucial enzyme in the fatty acid metabolism pathway located in mitochondria. However, the expression level and prognostic value of ACADS in colorectal cancer (CRC) remain unclear. Methods: The mRNA and protein expression data of ACADS was obtained from The Cancer Genome Atlas (TCGA), Clinical Proteomic Tumor Analysis Consortium (CPTAC), and Oncomine. Prognostic values of ACADS were calculated using Kaplan-Meier survival analysis. Correlations between ACADS and immune infiltration were estimated using TIMER, CIBERSORT, EPIC, quanTIseq, and xCell. The UALCAN and MEXPRESS databases were utilized for Methylation analysis. The co-expression analysis based on mRNA expression and interaction network of ACADS were performed via several online tools. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis on ACADS co-expressed genes were performed using the Metascape. Results: The expression analysis demonstrated that ACADS was down-regulated in CRC tissues compared with paired normal tissue. Expression of ACADS was found to be significantly associated with clinical cancer stages and the consensus molecular subgroups (CMS) constituent ratio in CRC patients. Besides, lower ACADS expression was found to predict poor prognosis and be significantly associated with common immune checkpoint genes and MMR genes in CRC. ACADS expression levels were positively related to B cells, CD4+ T cells, CD8+ T cells, M1 macrophages, neutrophils, and Tregs, while negatively correlated with M0 macrophages, M2 macrophages. The methylation level of ACADS in normal tissues was significantly higher than that in tumor tissues, and several methylation sites were identified. The enrichment analysis suggested the co-expressed genes mainly enriched in cell mitochondrial metabolism. Conclusions: The present study provided multilevel evidences for expression of ACADS in CRC and the function of ACADS in prognostic prediction, immune infiltration, and methylation. ACADS might have the potential as the novel biomarker and therapeutic target in CRC patients.


Subject(s)
Butyryl-CoA Dehydrogenase/genetics , Butyryl-CoA Dehydrogenase/metabolism , Colorectal Neoplasms/diagnosis , Adenocarcinoma/diagnosis , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma/diagnosis , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/mortality , Cell Line, Tumor , Chemotaxis, Leukocyte/genetics , Chemotaxis, Leukocyte/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Fatty Acids/metabolism , Gene Expression Profiling , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Lipid Metabolism/genetics , Predictive Value of Tests , Prognosis , Proteomics , Survival Analysis , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
9.
Nat Commun ; 12(1): 6798, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34815397

ABSTRACT

G-protein-coupled receptors (GPCRs), especially chemokine receptors, play a central role in the regulation of T cell migration. Various GPCRs are upregulated in activated CD4 T cells, including P2Y10, a putative lysophospholipid receptor that is officially still considered an orphan GPCR, i.e., a receptor with unknown endogenous ligand. Here we show that in mice lacking P2Y10 in the CD4 T cell compartment, the severity of experimental autoimmune encephalomyelitis and cutaneous contact hypersensitivity is reduced. P2Y10-deficient CD4 T cells show normal activation, proliferation and differentiation, but reduced chemokine-induced migration, polarization, and RhoA activation upon in vitro stimulation. Mechanistically, CD4 T cells release the putative P2Y10 ligands lysophosphatidylserine and ATP upon chemokine exposure, and these mediators induce P2Y10-dependent RhoA activation in an autocrine/paracrine fashion. ATP degradation impairs RhoA activation and migration in control CD4 T cells, but not in P2Y10-deficient CD4 T cells. Importantly, the P2Y10 pathway appears to be conserved in human T cells. Taken together, P2Y10 mediates RhoA activation in CD4 T cells in response to auto-/paracrine-acting mediators such as LysoPS and ATP, thereby facilitating chemokine-induced migration and, consecutively, T cell-mediated diseases.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Receptors, Purinergic P2Y/metabolism , Receptors, Purinergic P2/metabolism , Adenosine Triphosphate/metabolism , Adult , Aged , Animals , Autocrine Communication/immunology , CD4-Positive T-Lymphocytes/metabolism , Case-Control Studies , Cells, Cultured , Chemokines/metabolism , Chemotaxis, Leukocyte/immunology , Encephalomyelitis, Autoimmune, Experimental/blood , Female , Gene Knockdown Techniques , Gene Knockout Techniques , Humans , Lysophospholipids/metabolism , Male , Mice , Mice, Transgenic , Middle Aged , Multiple Sclerosis/blood , Paracrine Communication/immunology , Primary Cell Culture , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2Y/genetics , rhoA GTP-Binding Protein/metabolism
10.
Proc Natl Acad Sci U S A ; 118(43)2021 10 26.
Article in English | MEDLINE | ID: mdl-34675073

ABSTRACT

Neutrophils sense and migrate through an enormous range of chemoattractant gradients through adaptation. Here, we reveal that in human neutrophils, calcium-promoted Ras inactivator (CAPRI) locally controls the GPCR-stimulated Ras adaptation. Human neutrophils lacking CAPRI (caprikd ) exhibit chemoattractant-induced, nonadaptive Ras activation; significantly increased phosphorylation of AKT, GSK-3α/3ß, and cofilin; and excessive actin polymerization. caprikd cells display defective chemotaxis in response to high-concentration gradients but exhibit improved chemotaxis in low- or subsensitive-concentration gradients of various chemoattractants, as a result of their enhanced sensitivity. Taken together, our data reveal that CAPRI controls GPCR activation-mediated Ras adaptation and lowers the sensitivity of human neutrophils so that they are able to chemotax through a higher-concentration range of chemoattractant gradients.


Subject(s)
Chemotaxis, Leukocyte/immunology , Neutrophils/immunology , ras GTPase-Activating Proteins/immunology , ras Proteins/antagonists & inhibitors , Actins/immunology , Cell Movement , Cell Polarity , Gene Knockdown Techniques , HL-60 Cells , Humans , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophil Activation/drug effects , Neutrophil Activation/genetics , Neutrophil Activation/immunology , Neutrophils/drug effects , Neutrophils/physiology , Receptors, G-Protein-Coupled/immunology , Shelterin Complex/immunology , Signal Transduction , Telomere-Binding Proteins/immunology , ras GTPase-Activating Proteins/deficiency , ras GTPase-Activating Proteins/genetics , ras Proteins/immunology
11.
Front Immunol ; 12: 744155, 2021.
Article in English | MEDLINE | ID: mdl-34691047

ABSTRACT

Immunological non-responders (INR), a subgroup of people living with HIV (PLHIV) who fail to restore CD4+ T cell numbers upon effective antiretroviral treatment, have impaired gut mucosal barrier function and an inferior clinical prognosis compared with immunological responders (IR). The contribution of gut-homing and exhaustion of mucosal T cells to the INR phenotype was previously unknown. Flow cytometry analysis of mononuclear cells from peripheral blood and ileal and colonic lamina propria showed that INR had higher fractions of gut-homing CD4+ T cells in blood compared with IR. In addition, gut-homing cells were more likely to display signs of exhaustion in INR. The increased CD4+ T cell exhaustion in INR was ubiquitous and not restricted to subpopulations defined by activation, differentiation or regulatory T cell markers. In INR, colon CD4+ T cell exhaustion correlated negatively with the fraction of CD4+ T cells in the same compartment, this was not apparent in the ileum. The fraction of exhausted mucosal CD4+ T cells correlated with I-FABP and REG3α, markers of enterocyte damage. We conclude that alterations of gut-homing and exhaustion of T cells may contribute to impaired gut immune and barrier functions associated with immunological non-response in PLHIV.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Drug Resistance/immunology , HIV Infections/immunology , Immunosenescence/immunology , Intestinal Mucosa/immunology , Adult , Aged , Anti-HIV Agents/therapeutic use , CD4-Positive T-Lymphocytes/pathology , Chemotaxis, Leukocyte/immunology , HIV Infections/drug therapy , Humans , Male , Middle Aged
12.
Front Immunol ; 12: 715559, 2021.
Article in English | MEDLINE | ID: mdl-34539647

ABSTRACT

The involvement of gut microbiota in T-cell trafficking into tumor tissue of colorectal cancer (CRC) remains to be further elucidated. The current study aimed to evaluate the expression of major cytotoxic T-cell trafficking chemokines (CTTCs) and chemokine-associated microbiota profiles in both tumor and adjacent normal tissues during CRC progression. We analyzed the expression of chemokine C-X-C motif ligands 9, 10, and 11 (CXCL9, CXCL10, and CXCL11), and C-C motif ligand 5 (CCL5), characterized gut mucosa-associated microbiota (MAM), and investigated their correlations in CRC patients. Our results showed that the expression of CXCL9, CXCL10, and CXCL11 was significantly higher in tumor than in adjacent normal tissues in 136 CRC patients. Notably, the high expression of CXCL9 in tumor tissues was associated with enhanced CD8+ T-cell infiltration and improved survival. Moreover, the MAM in tumor tissues showed reduction of microbial diversity and increase of oral bacteria. Microbial network analysis identified differences in microbial composition and structure between tumor and adjacent normal tissues. In addition, stronger associations between oral bacteria and other gut microbes were observed. Furthermore, the correlation analysis between the defined MAM and individual CTTCs showed that the CTTCs' correlated operational taxonomic units (OTUs) in tumor and adjacent normal tissues rarely overlap with each other. Notably, all the enriched OTUs were positively correlated with the CTTCs in either tumor or adjacent normal tissues. Our findings demonstrated stronger interactions between oral bacteria and gut microbes, and a shifted correlation pattern between MAM and major CTTCs in tumor tissues, underlining possible mechanisms of gut microbiota-host interaction in CRC.


Subject(s)
Chemokines/metabolism , Chemotaxis, Leukocyte/immunology , Colorectal Neoplasms/etiology , Colorectal Neoplasms/metabolism , Gastrointestinal Microbiome/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Adult , Aged , Biomarkers , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Colorectal Neoplasms/pathology , Computational Biology/methods , Disease Progression , Disease Susceptibility , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Male , Metagenome , Metagenomics , Middle Aged , Neoplasm Grading , Neoplasm Staging
13.
Front Immunol ; 12: 698420, 2021.
Article in English | MEDLINE | ID: mdl-34497606

ABSTRACT

Siglec-H is a DAP12-associated receptor on plasmacytoid dendritic cells (pDCs) and microglia. Siglec-H inhibits TLR9-induced IFN-α production by pDCs. Previously, it was found that Siglec-H-deficient mice develop a lupus-like severe autoimmune disease after persistent murine cytomegalovirus (mCMV) infection. This was due to enhanced type I interferon responses, including IFN-α. Here we examined, whether other virus infections can also induce autoimmunity in Siglec-H-deficient mice. To this end we infected Siglec-H-deficient mice with influenza virus or with Lymphocytic Choriomeningitis virus (LCMV) clone 13. With both types of viruses we did not observe induction of autoimmune disease in Siglec-H-deficient mice. This can be explained by the fact that both types of viruses are ssRNA viruses that engage TLR7, rather than TLR9. Also, Influenza causes an acute infection that is rapidly cleared and the chronicity of LCMV clone 13 may not be sufficient and may rather suppress pDC functions. Siglec-H inhibited exclusively TLR-9 driven type I interferon responses, but did not affect type II or type III interferon production by pDCs. Siglec-H-deficient pDCs showed impaired Hck expression, which is a Src-family kinase expressed in myeloid cells, and downmodulation of the chemokine receptor CCR9, that has important functions for pDCs. Accordingly, Siglec-H-deficient pDCs showed impaired migration towards the CCR9 ligand CCL25. Furthermore, autoimmune-related genes such as Klk1 and DNase1l3 are downregulated in Siglec-H-deficient pDCs as well. From these findings we conclude that Siglec-H controls TLR-9-dependent, but not TLR-7 dependent inflammatory responses after virus infections and regulates chemokine responsiveness of pDCs.


Subject(s)
Arenaviridae Infections/immunology , Autoimmune Diseases/immunology , Interferon Type I/immunology , Lectins/immunology , Orthomyxoviridae Infections/immunology , Receptors, Cell Surface/immunology , Animals , Autoimmune Diseases/virology , Autoimmunity/immunology , Chemotaxis, Leukocyte/immunology , Dendritic Cells/immunology , Influenza A Virus, H3N2 Subtype , Lectins/deficiency , Lymphocytic choriomeningitis virus , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cell Surface/deficiency , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism
14.
Int J Mol Sci ; 22(17)2021 Aug 26.
Article in English | MEDLINE | ID: mdl-34502120

ABSTRACT

Diabetes mellitus is a main risk factor for delayed fracture healing and fracture non-unions. Successful fracture healing requires stimuli from different immune cells, known to be affected in diabetics. Especially, application of mononuclear cells has been proposed to promote wound and fracture healing. Thus, aim was to investigate the effect of pre-/diabetic conditions on mononuclear cell functions essential to promote osteoprogenitor cell function. We here show that pre-/diabetic conditions suppress the expression of chemokines, e.g., CCL2 and CCL8 in osteoprogenitor cells. The associated MCP-1 and MCP-2 were significantly reduced in serum of diabetics. Both MCPs chemoattract mononuclear THP-1 cells. Migration of these cells is suppressed under hyperglycemic conditions, proposing that less mononuclear cells invade the site of fracture in diabetics. Further, we show that the composition of cytokines secreted by mononuclear cells strongly differ between diabetics and controls. Similar is seen in THP-1 cells cultured under hyperinsulinemia or hyperglycemia. The altered secretome reduces the positive effect of the THP-1 cell conditioned medium on migration of osteoprogenitor cells. In summary, our data support that factors secreted by mononuclear cells may support fracture healing by promoting migration of osteoprogenitor cells but suggest that this effect might be reduced in diabetics.


Subject(s)
Culture Media, Conditioned/metabolism , Diabetes Mellitus/metabolism , Fracture Healing , Monocytes/metabolism , Animals , Biomarkers , Cell Movement , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CCL8/metabolism , Chemokines/metabolism , Chemotaxis, Leukocyte/immunology , Humans , Hyperglycemia/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , MAP Kinase Signaling System , Monocytes/immunology , Osteoblasts/metabolism , Osteogenesis , THP-1 Cells
15.
Mucosal Immunol ; 14(6): 1271-1281, 2021 11.
Article in English | MEDLINE | ID: mdl-34341502

ABSTRACT

Expression of Ikaros family transcription factor IKZF3 (Aiolos) increases during murine eosinophil lineage commitment and maturation. Herein, we investigated Aiolos expression and function in mature human and murine eosinophils. Murine eosinophils deficient in Aiolos demonstrated gene expression changes in pathways associated with granulocyte-mediated immunity, chemotaxis, degranulation, ERK/MAPK signaling, and extracellular matrix organization; these genes had ATAC peaks within 1 kB of the TSS that were enriched for Aiolos-binding motifs. Global Aiolos deficiency reduced eosinophil frequency within peripheral tissues during homeostasis; a chimeric mouse model demonstrated dependence on intrinsic Aiolos expression by eosinophils. Aiolos deficiency reduced eosinophil CCR3 surface expression, intracellular ERK1/2 signaling, and CCL11-induced actin polymerization, emphasizing an impaired functional response. Aiolos-deficient eosinophils had reduced tissue accumulation in chemokine-, antigen-, and IL-13-driven inflammatory experimental models, all of which at least partially depend on CCR3 signaling. Human Aiolos expression was associated with active chromatin marks enriched for IKZF3, PU.1, and GATA-1-binding motifs within eosinophil-specific histone ChIP-seq peaks. Furthermore, treating the EOL-1 human eosinophilic cell line with lenalidomide yielded a dose-dependent decrease in Aiolos. These collective data indicate that eosinophil homing during homeostatic and inflammatory allergic states is Aiolos-dependent, identifying Aiolos as a potential therapeutic target for eosinophilic disease.


Subject(s)
Chemotaxis, Leukocyte/genetics , Chemotaxis, Leukocyte/immunology , Eosinophils/immunology , Eosinophils/metabolism , Ikaros Transcription Factor/genetics , Allergens/immunology , Animals , Disease Susceptibility , Female , Gene Expression Profiling , Gene Expression Regulation , Granulocytes/immunology , Granulocytes/metabolism , Humans , Ikaros Transcription Factor/metabolism , Immunity, Innate , Immunophenotyping , Leukocyte Count , Male , Mice , Mice, Knockout , Models, Animal , Receptors, CCR3/genetics , Receptors, CCR3/metabolism , Signal Transduction
16.
BMC Cancer ; 21(1): 922, 2021 Aug 15.
Article in English | MEDLINE | ID: mdl-34391381

ABSTRACT

OBJECTIVE: Tongue and mouth floor squamous cell carcinoma (T/MF SCC) exhibits a high rate of local recurrence and cervical lymph node metastasis. The effect of the tumor microenvironment on T/MF SCC remains unclear. MATERIALS AND METHODS: Transcriptome and somatic mutation data of patients with T/MF SCC were obtained from HNSC projects of the Cancer Genome Atlas. Immune infiltration quantification in early- (clinical stage I-II) and advanced-stage (clinical stage III-IV) T/MF SCC was performed using single sample Gene Set Enrichment Analysis and MCPcounter. Differentially expressed gene data were filtered, and their function was assessed through Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses. Kaplan-Meier survival curve analysis and Cox regression model were conducted to evaluate the survival of patients with the CCL22 signature. Maftools was used to present the overview of somatic mutations. RESULTS: In T/MF SCC, T helper (Th)2 cell counts were significantly increased in patients with early-stage disease compared to those with advanced-stage disease. Expression of the Th2 cell-related chemokine, CCL22, was downregulated in patients with advanced-stage T/MF SCC. Univariate and multivariate Cox analyses revealed that CCL22 was a good prognostic factor in T/MF SCC. A nomogram based on the expression of CCL22 was constructed to serve as a prognostic indicator for T/MF SCC. NOTCH1 mutations were found at a higher rate in patients with advanced-stage T/MF SCC than in those with early-stage T/MF SCC, resulting in the inhibition of the activation of the NOTCH1-Th2 cell differentiation pathway. The expression levels of CCL22, GATA-3, and IL4 were higher in patients with early-stage T/MF SCC than in those with advanced-stage T/MF SCC. CONCLUSION: In T/MF SCC, high expression of CCL22 may promote the recruitment of Th2 cells and help predict a better survival. Mutations in NOTCH1 inhibit the differentiation of Th2 cells, facilitating tumor progression through a decrease in Th2 cell recruitment and differentiation.


Subject(s)
Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/metabolism , Chemokine CCL22/genetics , Mouth Neoplasms/etiology , Mouth Neoplasms/metabolism , Receptor, Notch1/genetics , Th2 Cells/immunology , Th2 Cells/metabolism , Adult , Aged , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Chemotaxis, Leukocyte/genetics , Chemotaxis, Leukocyte/immunology , Computational Biology/methods , Female , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Lymphocyte Count , Lymphocytes, Tumor-Infiltrating , Male , Middle Aged , Mouth Floor/metabolism , Mouth Floor/pathology , Mouth Neoplasms/mortality , Mouth Neoplasms/pathology , Mutation , Neoplasm Staging , Prognosis , Proportional Hazards Models
17.
Immunology ; 164(4): 737-753, 2021 12.
Article in English | MEDLINE | ID: mdl-34407221

ABSTRACT

Experimental cerebral malaria (ECM) is a severe complication of Plasmodium berghei ANKA (PbA) infection in mice, characterized by CD8+ T-cell accumulation within the brain. Whilst the dynamics of CD8+ T-cell activation and migration during extant primary PbA infection have been extensively researched, the fate of the parasite-specific CD8+ T cells upon resolution of ECM is not understood. In this study, we show that memory OT-I cells persist systemically within the spleen, lung and brain following recovery from ECM after primary PbA-OVA infection. Whereas memory OT-I cells within the spleen and lung exhibited canonical central memory (Tcm) and effector memory (Tem) phenotypes, respectively, memory OT-I cells within the brain post-PbA-OVA infection displayed an enriched CD69+ CD103- profile and expressed low levels of T-bet. OT-I cells within the brain were excluded from short-term intravascular antibody labelling but were targeted effectively by longer-term systemically administered antibodies. Thus, the memory OT-I cells were extravascular within the brain post-ECM but were potentially not resident memory cells. Importantly, whilst memory OT-I cells exhibited strong reactivation during secondary PbA-OVA infection, preventing activation of new primary effector T cells, they had dampened reactivation during a fourth PbA-OVA infection. Overall, our results demonstrate that memory CD8+ T cells are systemically distributed but exhibit a unique phenotype within the brain post-ECM, and that their reactivation characteristics are shaped by infection history. Our results raise important questions regarding the role of distinct memory CD8+ T-cell populations within the brain and other tissues during repeat Plasmodium infections.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Host-Parasite Interactions/immunology , Malaria/immunology , Malaria/parasitology , Plasmodium berghei/physiology , Animals , Biomarkers , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Chemotaxis, Leukocyte/immunology , Disease Susceptibility , Epitopes, T-Lymphocyte/immunology , Erythrocytes/immunology , Erythrocytes/parasitology , Extracellular Matrix , Immunologic Memory , Immunophenotyping , Life Cycle Stages , Lymphocyte Activation/immunology , Malaria/metabolism , Malaria/pathology , Malaria, Cerebral/immunology , Malaria, Cerebral/metabolism , Malaria, Cerebral/parasitology , Mice , Mice, Transgenic , Organ Specificity/immunology
18.
Sci Rep ; 11(1): 14047, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34234274

ABSTRACT

The Apelin/APJ signalling pathway, involved in multiple physiological and pathological processes, has been attracting increasing interest recently. In our previous study, Apelin overexpression in colon26 tumor cells suppressed tumor growth by inducing vascular maturation. Here, we found that MC38 and LLC tumor growth were greater in the absence of Apelin than in wild-type (WT) mice, suggesting that Apelin acts as a tumor suppressor. Consistent with this, treating WT mice with [Pyr1]Apelin-13 inhibited tumor growth. In MC38 tumors, only endothelial cells (ECs) strongly express APJ, a cognate receptor for Apelin, indicating that EC-derived Apelin might regulate tumor formation in an autocrine manner. Comparing with WT mice, larger numbers of vessels with narrower diameters were observed in tumors of Apelin knockout mice and lack of Apelin enhanced tumor hypoxia. Investigating immune cells in the tumor revealed that [Pyr1]Apelin-13 infusion induced the accumulation of CD8+ and CD4+ T cells in central areas. Moreover, RNA-sequencing analysis showed that Apelin induces chemokine CCL8 expression in ECs. Thus, enhancing anti-tumor immunity might be one of the mechanisms by which Apelin is involved in tumor growth. Our result indicated that increased CCL8 expression might induce CD8 + T cells infiltration into tumor and tumor inhibition.


Subject(s)
Apelin/metabolism , Chemotaxis, Leukocyte/genetics , Endothelial Cells/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Animals , Apelin/genetics , Apelin Receptors/genetics , Apelin Receptors/metabolism , Cell Line, Tumor , Cell Proliferation , Chemokine CCL8/biosynthesis , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Knockout , Neoplasms/pathology , Protein Binding , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Tumor Microenvironment
19.
Eur J Immunol ; 51(8): 1980-1991, 2021 08.
Article in English | MEDLINE | ID: mdl-34060652

ABSTRACT

High mobility group box-1 protein (HMGB1) is an alarmin that, once released, promotes inflammatory responses, alone and as a complex with the chemokine CXCL12. Here, we report that the HMGB1-CXCL12 complex plays an essential role also in homeostasis by controlling the migration of B lymphocytes. We show that extracellular HMGB1 is critical for the CXCL12-dependent egress of B cells from the Peyer's patches (PP). This promigratory function of the complex was restricted to the PPs, since HMGB1 was not required for B-cell migratory processes in other locations. Accordingly, we detected higher constitutive levels of the HMGB1-CXCL12 complex in PPs than in other lymphoid organs. HMGB1-CXCL12 in vivo inhibition was associated with a reduced basal IgA production in the gut. Collectively, our results demonstrate a role for the HMGB1-CXCL12 complex in orchestrating B-cell trafficking in homeostasis, and provide a novel target to control lymphocyte migration in mucosal immunity.


Subject(s)
B-Lymphocytes/metabolism , Chemokine CXCL12/metabolism , HMGB1 Protein/metabolism , Immunity, Mucosal/immunology , Peyer's Patches/metabolism , Animals , B-Lymphocytes/immunology , Chemokine CXCL12/immunology , Chemotaxis, Leukocyte/immunology , HMGB1 Protein/immunology , Homeostasis/immunology , Mice , Mice, Inbred C57BL , Peyer's Patches/immunology
20.
Viruses ; 13(5)2021 05 07.
Article in English | MEDLINE | ID: mdl-34066999

ABSTRACT

MUC1 belongs to the family of cell surface (cs-) mucins. Experimental evidence indicates that its presence reduces in vivo influenza viral infection severity. However, the mechanisms by which MUC1 influences viral dynamics and the host immune response are not yet well understood, limiting our ability to predict the efficacy of potential treatments that target MUC1. To address this limitation, we use available in vivo kinetic data for both virus and macrophage populations in wildtype and MUC1 knockout mice. We apply two mathematical models of within-host influenza dynamics to this data. The models differ in how they categorise the mechanisms of viral control. Both models provide evidence that MUC1 reduces the susceptibility of epithelial cells to influenza virus and regulates macrophage recruitment. Furthermore, we predict and compare some key infection-related quantities between the two mice groups. We find that MUC1 significantly reduces the basic reproduction number of viral replication as well as the number of cumulative macrophages but has little impact on the cumulative viral load. Our analyses suggest that the viral replication rate in the early stages of infection influences the kinetics of the host immune response, with consequences for infection outcomes, such as severity. We also show that MUC1 plays a strong anti-inflammatory role in the regulation of the host immune response. This study improves our understanding of the dynamic role of MUC1 against influenza infection and may support the development of novel antiviral treatments and immunomodulators that target MUC1.


Subject(s)
Host-Pathogen Interactions , Influenza A virus/physiology , Macrophages/metabolism , Models, Biological , Mucin-1/metabolism , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Animals , Biomarkers , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Influenza, Human/genetics , Influenza, Human/immunology , Influenza, Human/metabolism , Influenza, Human/virology , Macrophages/immunology , Mice , Mice, Knockout , Mucin-1/genetics , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL
...