Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 268
Filter
1.
STAR Protoc ; 3(3): 101645, 2022 09 16.
Article in English | MEDLINE | ID: mdl-36042879

ABSTRACT

Here, we describe an in vivo approach to visualize CD11c+ cells in atherosclerosis. In particular, we use a protocol for X-Gal staining of immune cells within atherosclerotic plaques, which can be used as an alternative to analyze plaque composition and cell-specific molecules in atherogenesis. LacZ knockin mice have to be bred to mice carrying the CD11ccre recombinase-both brought onto an ApoE-/- background-to be able to visualize this cell type of interest in the plaques by X-Gal staining. With this approach, different immune cells in atherogenesis can be examined. For complete details on the use and execution of this protocol, please refer to Sauter et al. (2021).


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Animals , Atherosclerosis/genetics , CD11c Antigen/genetics , Lac Operon/genetics , Mice , Mice, Knockout , Plaque, Atherosclerotic/genetics
2.
J Pathol Clin Res ; 8(5): 436-447, 2022 09.
Article in English | MEDLINE | ID: mdl-35715938

ABSTRACT

Diffuse large B-cell lymphoma (DLBCL) is the most common type of non-Hodgkin lymphoma and is a potentially curable disease. However, it is heterogenous, and the prognosis is poor if the tumor cells harbor fusions involving MYC and BCL2 or MYC and BCL6 (double-hit [DH] lymphoma), or fusions involving all three genes (triple-hit [TH] lymphoma). Fluorescence in situ hybridization is currently the gold standard for confirming the presence of DH/TH genotypes. However, the test is laborious and not readily available in some laboratories. Germinal center B (GCB) signatures and dual expression of MYC and BCL2 are commonly used as initial screening markers (traditional model) in clinical practice. Our study proposes immunohistochemical markers for more conveniently and accessibly screening DH/TH genotypes in DLBCL. We retrospectively reviewed the clinical and pathological parameters of patients with DLBCL. We assessed the proliferative index, apoptotic index, and tumor microenvironment (TME), with regard to T cells and CD11c(+) dendritic cells, in formalin-fixed paraffin-embedded tissue. We then generated a decision tree as a screening algorithm to predict DH/TH genotypes and employed decision curve analysis to demonstrate the superiority of this new model in prediction. We also assessed the prognostic significance of related parameters. Our study revealed that GCB subtypes, a Ki67 proliferative index higher than 70%, and BCL2 expression were significantly associated with DH/TH genotypes. Decreased CD11c(+) dendritic cells in the TME indicated additional risk. Our proposed screening algorithm outperformed a traditional model in screening for the DH/TH genotypes. In addition, decreased CD11c(+) dendritic cells in the DLBCL TME were an independent unfavorable prognosticator. In conclusion, we provide a convenient, well-performing model that predicts DH/TH genotypes in DLBCL. The prognostic significance of CD11c(+) dendritic cells in the TME might influence the classification and development of immunotherapy for DLBCL in the future.


Subject(s)
CD11c Antigen , Dendritic Cells , Lymphoma, Large B-Cell, Diffuse , Proto-Oncogene Proteins , Tumor Microenvironment , Algorithms , CD11c Antigen/genetics , CD11c Antigen/metabolism , Cell Survival , Dendritic Cells/metabolism , Dendritic Cells/pathology , Genotype , Humans , In Situ Hybridization, Fluorescence , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/mortality , Lymphoma, Large B-Cell, Diffuse/pathology , Models, Biological , Oncogene Fusion/genetics , Oncogene Fusion/physiology , Prognosis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Retrospective Studies , Survival Analysis , Tumor Microenvironment/genetics , Tumor Microenvironment/physiology
3.
Sci Rep ; 12(1): 931, 2022 01 18.
Article in English | MEDLINE | ID: mdl-35042931

ABSTRACT

Gland macrophages are primed for gland development and functions through interactions within their niche. However, the phenotype, ontogeny, and function of steady-state salivary gland (SG) macrophages remain unclear. We herein identified CD11c+ and CD11c- subsets among CD64+ macrophages in steady-state murine SGs. CD11c- macrophages were predominant in the SGs of embryonic and newborn mice and decreased with advancing age. CD11c+ macrophages were rarely detected in the embryonic period, but rapidly expanded after birth. CD11c+, but not CD11c-, macrophage numbers decreased in mice treated with a CCR2 antagonist, suggesting that CD11c+ macrophages accumulate from bone marrow-derived progenitors in a CCR2-dependent manner, whereas CD11c- macrophages were derived from embryonic progenitors in SGs. CD11c+ and CD11c- macrophages strongly expressed colony-stimulating factor (CSF)-1 receptor, the injection of an anti-CSF-1 receptor blocking antibody markedly reduced both subsets, and SGs strongly expressed CSF-1, indicating the dependency of SG resident macrophage development on CSF-1. The phagocytic activity of SG macrophages was extremely weak; however, the gene expression profile of SG macrophages indicated that SG macrophages regulate gland development and functions in SGs. These results suggest that SG CD11c+ and CD11c- macrophages are developed and instructed to perform SG-specific functions in steady-state SGs.


Subject(s)
CD11 Antigens/genetics , Macrophages/metabolism , Salivary Glands/metabolism , Animals , CD11 Antigens/metabolism , CD11c Antigen/genetics , CD11c Antigen/metabolism , Cell Differentiation , Dendritic Cells/immunology , Female , Gene Expression/genetics , Gene Expression Regulation, Developmental/genetics , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/immunology , Male , Mice/embryology , Mice, Inbred C57BL , Phagocytes/metabolism , Salivary Glands/immunology
4.
Cell Rep ; 37(11): 110111, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34910922

ABSTRACT

Blockade of the inhibitory checkpoint SIRPα-CD47 promotes phagocytosis of cancer cells by macrophages and is a promising avenue in anti-cancer therapy. Productive phagocytosis is strictly predicated on co-engagement of pro-phagocytic receptors-namely, Fc receptors (FcRs), integrin CD11b, or SLAMF7-by their ligands on cancer cells. Here, we examine whether additional pro-phagocytic receptors could be harnessed to broaden the scope of phagocytosis. Inflammatory stimuli, including multiple cytokines and Toll-like receptor (TLR) ligands, augment phagocytosis efficiency and fully alleviate the requirement of FcRs, CD11b, and SLAMF7 for phagocytosis. These effects are mediated by the unconventional pro-phagocytic integrins CD11a and CD11c, which act with CD18 to initiate actin polarization, leading to phagocytosis. Some inflammatory stimuli enable phagocytosis even in the absence of SIRPα-CD47 blockade. Higher CD11c expression in macrophage-enriched tumors correlates with improved survival in clinical studies. Thus, inflammatory macrophages exploit unconventional pro-phagocytic integrins for improved phagocytosis and anti-tumor immunity.


Subject(s)
CD11a Antigen/metabolism , CD11c Antigen/metabolism , Inflammation/immunology , Macrophages/immunology , Peritoneal Neoplasms/prevention & control , Phagocytosis , Signaling Lymphocytic Activation Molecule Family/physiology , Animals , CD11a Antigen/genetics , CD11c Antigen/genetics , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritoneal Neoplasms/immunology , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/pathology
5.
Immunology ; 164(1): 148-160, 2021 09.
Article in English | MEDLINE | ID: mdl-33934334

ABSTRACT

CD11c is a canonical dendritic cell (DC) marker with poorly defined functions in the immune system. Here, we found that blocking CD11c on human peripheral blood mononuclear cell-derived DCs (MoDCs) inhibited the proliferation of CD4+ T cells and the differentiation into IFN-γ-producing T helper 1 (Th1) cells, which were critical in acute graft-versus-host disease (aGVHD) pathogenesis. Using allogeneic bone marrow transplantation (allo-BMT) murine models, we consistently found that CD11c-deficient recipient mice had alleviated aGVHD symptoms for the decreased IFN-γ-expressing CD4+ Th1 cells and CD8+ T cells. Transcriptional analysis showed that CD11c participated in several immune regulation functions including maintaining antigen presentation of APCs. CD11c-deficient bone marrow-derived DCs (BMDCs) impaired the antigen presentation function in coculture assay. Mechanistically, CD11c interacted with MHCII and Hsp90 and participated in the phosphorylation of Akt and Erk1/2 in DCs after multiple inflammatory stimulations. Therefore, CD11c played crucial roles in triggering aGVHD and might serve as a potential target for the prevention and treatment of aGVHD.


Subject(s)
Bone Marrow Transplantation , CD11c Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Th1 Cells/immunology , Acute Disease , Animals , Antigen Presentation , CD11c Antigen/genetics , Cells, Cultured , Graft vs Host Disease , Interferon-gamma/metabolism , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Transplantation, Homologous
6.
Diabetes ; 70(3): 720-732, 2021 03.
Article in English | MEDLINE | ID: mdl-33323395

ABSTRACT

CD11c+ macrophages/dendritic cells (MDCs) are increased and display the classically activated M1-like phenotype in obese adipose tissue (AT) and may contribute to AT inflammation and insulin resistance. Stat1 is a key transcription factor for MDC polarization into the M1-like phenotype. Here, we examined the role of Stat1 in obesity-induced AT MDC polarization and inflammation and insulin resistance using mice with specific knockout of Stat1 in MDCs (cKO). Stat1 was upregulated and phosphorylated, indicating activation, early and persistently in AT and AT MDCs of wild-type mice fed a high-fat diet (HFD). Compared with littermate controls, cKO mice fed an HFD (16 weeks) had reductions in MDC (mainly CD11c+ macrophage) M1-like polarization and interferon-γ-expressing T-helper type 1 (Th1) cells but increases in interleukin 5-expressing Th2 cells and eosinophils in perigonadal and inguinal AT, and enhanced inguinal AT browning, with increased energy expenditure. cKO mice compared with controls also had significant reductions in triglyceride content in the liver and skeletal muscle and exhibited improved insulin sensitivity and glucose tolerance. Taken together, our results demonstrate that Stat1 in MDCs plays an important role in obesity-induced MDC M1-like polarization and AT inflammation and contributes to insulin resistance and metabolic dysfunctions in obese mice.


Subject(s)
Adipose Tissue/metabolism , CD11c Antigen/metabolism , Diet, High-Fat/adverse effects , Inflammation/metabolism , Insulin Resistance/physiology , Obesity/immunology , Obesity/metabolism , STAT1 Transcription Factor/metabolism , Adult , Animals , Blotting, Western , CD11c Antigen/genetics , Cells, Cultured , Female , Flow Cytometry , Humans , Immunohistochemistry , Inflammation/genetics , Inflammation/immunology , Insulin Resistance/genetics , Interleukin-5/metabolism , Male , Mice , Obesity/genetics , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/genetics
7.
Cell Rep ; 33(3): 108291, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33086061

ABSTRACT

Brain CD11c+ cells share features with microglia and dendritic cells (DCs). Sterile inflammation increases brain CD11c+ cells, but their phenotype, origin, and functions remain largely unknown. We report that, after cerebral ischemia, microglia attract DCs to the inflamed brain, and astroglia produce Flt3 ligand, supporting development and expansion of CD11c+ cells. CD11c+ cells in the inflamed brain are a complex population derived from proliferating microglia and infiltrating DCs, including a major subset of OX40L+ conventional cDC2, and also cDC1, plasmacytoid, and monocyte-derived DCs. Despite sharing certain morphological features and markers, CD11c+ microglia and DCs display differential expression of pattern recognition receptors and chemokine receptors. DCs excel CD11c- and CD11c+ microglia in the capacity to present antigen through MHCI and MHCII. Of note, cDC1s protect from brain injury after ischemia. We thus reveal aspects of the dynamics and functions of brain DCs in the regulation of inflammation and immunity.


Subject(s)
CD11 Antigens/metabolism , Dendritic Cells/metabolism , Microglia/metabolism , Animals , Antigens/metabolism , Brain/immunology , Brain/metabolism , CD11 Antigens/genetics , CD11c Antigen/genetics , CD11c Antigen/metabolism , Cytokines/metabolism , Dendritic Cells/physiology , Encephalitis/immunology , Encephalitis/metabolism , Flow Cytometry , Inflammation/immunology , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Microglia/physiology , Monocytes/metabolism , Receptors, Chemokine/metabolism
8.
Front Immunol ; 11: 2010, 2020.
Article in English | MEDLINE | ID: mdl-32922405

ABSTRACT

Dendritic cells (DCs) possess intrinsic cellular defense mechanisms to specifically inhibit HIV-1 replication. In turn, HIV-1 has evolved strategies to evade innate immune sensing by DCs resulting in suboptimal maturation and poor antiviral immune responses. We previously showed that complement-opsonized HIV-1 (HIV-C) was able to efficiently infect various DC subsets significantly higher than non-opsonized HIV-1 (HIV) and therefore also mediate a higher antiviral immunity. Thus, complement coating of HIV-1 might play a role with respect to viral control occurring early during infection via modulation of DCs. To determine in detail which complement receptors (CRs) expressed on DCs was responsible for infection and superior pro-inflammatory and antiviral effects, we generated stable deletion mutants for the α-chains of CR3, CD11b, and CR4, CD11c using CRISPR/Cas9 in THP1-derived DCs. We found that CD11c deletion resulted in impaired DC infection as well as antiviral and pro-inflammatory immunity upon exposure to complement-coated HIV-1. In contrast, sole expression of CD11b on DCs shifted the cells to an anti-inflammatory, regulatory DC type. We here illustrated that CR4 comprised of CD11c and CD18 is the major player with respect to DC infection associated with a potent early pro-inflammatory immune response. A more detailed characterization of CR3 and CR4 functions using our powerful tool might open novel avenues for early therapeutic intervention during HIV-1 infection.


Subject(s)
Dendritic Cells/immunology , HIV Infections/immunology , HIV-1/physiology , Integrin alphaXbeta2/metabolism , Macrophage-1 Antigen/metabolism , CD11b Antigen/genetics , CD11c Antigen/genetics , CD18 Antigens/genetics , CRISPR-Cas Systems , Complement System Proteins/metabolism , Humans , Immunity , Integrin alphaXbeta2/genetics , Macrophage-1 Antigen/genetics , Sequence Deletion/genetics , Signal Transduction , THP-1 Cells
9.
Int J Mol Sci ; 21(16)2020 Aug 16.
Article in English | MEDLINE | ID: mdl-32824307

ABSTRACT

L5, the most negatively charged subfraction of low-density lipoprotein (LDL), is implicated in atherogenesis, but the pathogenic association is relatively unexplored in patients with rheumatoid arthritis (RA). We examined the role of L5 LDL in macrophage foam cell formation and the association of L5 with CD11c expression in THP-1 cells and RA patients. Using quantitative real-time PCR, we determined mRNA expression levels of ITGAX, the gene for CD11c, a marker associated with vascular plaque formation and M1 macrophages in atherogenesis, in 93 RA patients. We also examined CD11c expression on THP-1 cells treated with L5 by flow cytometry analysis and the plasma levels of inflammatory mediators using a magnetic bead array. We found a dose-dependent upregulation of foam cell formation of macrophages after L5 treatment (mean ± SEM, 12.05 ± 2.35% in L5 (10 µg/mL); 50.13 ± 3.9% in L5 (25 µg/mL); 90.69 ± 1.82% in L5 (50 µg/mL), p < 0.01). Significantly higher levels of CD11c expression were observed in 30 patients with a high percentage of L5 in LDL (L5%) (0.0752 ± 0.0139-fold) compared to 63 patients with normal L5% (0.0446 ± 0.0054-fold, p < 0.05). CD11c expression levels were increased in the L5-treated group (30.00 ± 3.13% in L5 (10 µg/mL); 41.46 ± 2.77% in L5 (50 µg/mL), p < 0.05) and were positively correlated with plasma levels of interleukin (IL)-6 and IL-8. L5 augmented the expression of IL-6, IL-8, and tumor necrosis factor-α (TNF-α) on monocytes and macrophages. Our findings suggest that L5 may promote atherogenesis by augmenting macrophage foam cell formation, upregulating CD11c expression, and enhancing the expression levels of atherosclerosis-related mediators.


Subject(s)
Arthritis, Rheumatoid/metabolism , CD11c Antigen/genetics , Foam Cells/metabolism , Lipoproteins, LDL/metabolism , Aged , Arthritis, Rheumatoid/pathology , CD11c Antigen/metabolism , Female , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Male , Middle Aged , THP-1 Cells , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
10.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1865(10): 158776, 2020 10.
Article in English | MEDLINE | ID: mdl-32738301

ABSTRACT

Palmitoleic acid (POA, 16:1n-7) is a lipokine that has potential nutraceutical use to treat non-alcoholic fatty liver disease. We tested the effects of POA supplementation (daily oral gavage, 300 mg/Kg, 15 days) on murine liver inflammation induced by a high fat diet (HFD, 59% fat, 12 weeks). In HFD-fed mice, POA supplementation reduced serum insulin and improved insulin tolerance compared with oleic acid (OA, 300 mg/Kg). The livers of POA-treated mice exhibited less steatosis and inflammation than those of OA-treated mice with lower inflammatory cytokine levels and reduced toll-like receptor 4 protein content. The anti-inflammatory effects of POA in the liver were accompanied by a reduction in liver macrophages (LM, CD11c+; F4/80+; CD86+), an effect that could be triggered by peroxisome proliferator activated receptor (PPAR)-γ, a lipogenic transcription factor upregulated in livers of POA-treated mice. We also used HFD-fed mice with selective deletion of PPAR-γ in myeloid cells (PPAR-γ KOLyzCre+) to test whether the beneficial anti-inflammatory effects of POA are dependent on macrophages PPAR-γ. POA-mediated improvement of insulin tolerance was tightly dependent on myeloid PPAR-γ, while POA anti-inflammatory actions including the reduction in liver inflammatory cytokines were preserved in mice bearing myeloid cells deficient in PPAR-γ. This overlapped with increased CD206+ (M2a) cells and downregulation of CD86+ and CD11c+ liver macrophages. Moreover, POA supplementation increased hepatic AMPK activity and decreased expression of the fatty acid binding scavenger receptor, CD36. We conclude that POA controls liver inflammation triggered by fat accumulation through induction of M2a macrophages independently of myeloid cell PPAR-γ.


Subject(s)
Fatty Acids, Monounsaturated/pharmacology , Inflammation/drug therapy , Non-alcoholic Fatty Liver Disease/drug therapy , PPAR gamma/genetics , AMP-Activated Protein Kinase Kinases , Animals , B7-2 Antigen/genetics , CD11c Antigen/genetics , Diet, High-Fat/adverse effects , Fatty Acids, Monounsaturated/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Insulin Resistance/genetics , Lectins, C-Type/genetics , Liver/drug effects , Liver/metabolism , Liver/pathology , Mannose Receptor , Mannose-Binding Lectins/genetics , Mice , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Oleic Acid/metabolism , Oleic Acid/pharmacology , Protein Kinases/genetics , Receptors, Cell Surface/genetics
11.
EBioMedicine ; 55: 102774, 2020 May.
Article in English | MEDLINE | ID: mdl-32403085

ABSTRACT

BACKGROUND: Since numerous pathological conditions are evoked by unwanted dendritic cell (DC) activity, therapeutic agents modulating DC functions are of great medical interest. In regenerative medicine, cellular secretomes have gained increasing attention and valuable immunomodulatory properties have been attributed to the secretome of γ-irradiated peripheral blood mononuclear cells (PBMCs). Potential effects of the PBMC secretome (PBMCsec) on key DC functions have not been elucidated so far. METHODS: We used a hapten-mediated murine model of contact hypersensitivity (CH) to study the effects of PBMCsec on DCs in vivo. Effects of PBMCsec on human DCs were investigated in monocyte-derived DCs (MoDC) and ex vivo skin cultures. DCs were phenotypically characterised by transcriptomics analyses and flow cytometry. DC function was evaluated by cytokine secretion, antigen uptake, PBMC proliferation and T-cell priming. FINDINGS: PBMCsec significantly alleviated tissue inflammation and cellular infiltration in hapten-sensitized mice. We found that PBMCsec abrogated differentiation of MoDCs, indicated by lower expression of classical DC markers CD1a, CD11c and MHC class II molecules. Furthermore, PBMCsec reduced DC maturation, antigen uptake, lipopolysaccharides-induced cytokine secretion, and DC-mediated immune cell proliferation. Moreover, MoDCs differentiated with PBMCsec displayed diminished ability to prime naïve CD4+T-cells into TH1 and TH2 cells. Furthermore, PBMCsec modulated the phenotype of DCs present in the skin in situ. Mechanistically, we identified lipids as the main biomolecule accountable for the observed immunomodulatory effects. INTERPRETATION: Together, our data describe DC-modulatory actions of lipids secreted by stressed PBMCs and suggest PBMCsec as a therapeutic option for treatment of DC-mediated inflammatory skin conditions. FUNDING: This research project was supported by the Austrian Research Promotion Agency (Vienna, Austria; grant "APOSEC" 862068; 2015-2019) and the Vienna Business Agency (Vienna, Austria; grant "APOSEC to clinic" 2343727).


Subject(s)
Culture Media, Conditioned/chemistry , Dendritic Cells/radiation effects , Dermatitis, Contact/therapy , Immunologic Factors/pharmacology , Lipids/pharmacology , Skin/radiation effects , Adult , Animals , Antigens, CD1/genetics , Antigens, CD1/immunology , Biomarkers/analysis , CD11c Antigen/genetics , CD11c Antigen/immunology , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Dermatitis, Contact/etiology , Dermatitis, Contact/genetics , Dermatitis, Contact/immunology , Dinitrofluorobenzene/administration & dosage , Female , Gamma Rays , Gene Expression , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Humans , Immunologic Factors/isolation & purification , Lipids/isolation & purification , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Mice , Monocytes/radiation effects , Primary Cell Culture , Skin/immunology , Skin/pathology , Th1 Cells/cytology , Th1 Cells/drug effects , Th1 Cells/immunology , Th2 Cells/cytology , Th2 Cells/drug effects , Th2 Cells/immunology , Tissue Culture Techniques
12.
J Neuroimmunol ; 344: 577245, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32335319

ABSTRACT

BACKGROUND: The Cre-lox system is a non-dynamic method of gene modification and characterization. Promoters thought to be relatively cell-specific are utilized for generation of cell-lineage-specific gene modifications. METHODS: CD11c.Cre+ITGA4fl/fl mice were generated to abolish the expression of ITGA (α4-integrin) in CD11c+ cells. Ex vivo flow cytometry studies were used to assess the expression of cellular surface markers in different lymphoid compartments and leukocytes subsets after Cre-mediated recombination. RESULTS: A significant reduction of α4-integrin expression among CD11c+- cells was achieved in CD11c.Cre+ITGA4fl/fl mice in primary and secondary lymphoid tissues. A similar reduction in the expression of α4-integrin was also observed in CD11c- cells. CONCLUSION: Cre-lox-mediated cell lineage-specific gene deletion is limited by the transient expression of recombination regulating sequences in hematopoietic cell lines. These methodological issues indicate the need to consider when to employ non-dynamic DNA recombination models in animal models of CNS autoimmunity. An experimental algorithm to address the biological complexities of non-dynamic gene recombination is provided.


Subject(s)
CD11c Antigen/biosynthesis , CD11c Antigen/genetics , Cell Lineage/physiology , Integrins/biosynthesis , Integrins/genetics , Recombination, Genetic/physiology , Zebrafish Proteins/biosynthesis , Zebrafish Proteins/genetics , Animals , Cells, Cultured , Female , Mice , Mice, Inbred C57BL , Mice, Transgenic
13.
Neoplasia ; 22(2): 86-97, 2020 02.
Article in English | MEDLINE | ID: mdl-31896526

ABSTRACT

While recent research points to the importance of glycans in cancer immunity, knowledge on functional mechanisms is lacking. In lung carcinoma among other tumors, anti-tumor immunity is suppressed; and while some recent therapies boost T-cell mediated immunity by targeting immune-checkpoint pathways, robust responses are uncommon. Augmenting tumor antigen-specific immune responses by endogenous dendritic cells (DCs) is appealing from a specificity standpoint, but challenging. Here, we show that restricting a heparan sulfate (HS) loss-of-function mutation in the HS sulfating enzyme Ndst1 to predominantly conventional DCs (Ndst1f/f CD11cCre+ mutation) results in marked inhibition of Lewis lung carcinoma growth along with increased tumor-associated CD8+ T cells. In mice deficient in a major DC HS proteoglycan (syndecan-4), splenic CD8+ T cells showed increased anti-tumor cytotoxic responses relative to controls. Studies examining Ndst1f/f CD11cCre + mutants revealed that mutation was associated with an increase in anti-tumor cytolysis using either splenic CD8+ T cells or tumor-infiltrating (TIL) CD8+ T cells purified ex-vivo, and tested in pooled effector-to-target cytolytic assays against tumor cells from respective animals. On glycan compositional analysis, HS purified from Ndst1f/f CD11cCre + mutant DCs had reduced overall sulfation, including reduced sulfation of a tri-sulfated disaccharide species that was intriguingly abundant on wildtype DC HS. Interestingly, antigen presentation in the context of major histocompatibility complex class-I (MHC-I) was enhanced in mutant DCs, with more striking effects in the setting of HS under-sulfation, pointing to a likely regulatory role by sulfated glycans at the antigen/MHC-I - T-cell interface; and possibly future opportunities to improve antigen-specific T cell responses by immunologic targeting of HS proteoglycans in cancer.


Subject(s)
Carcinoma, Lewis Lung/drug therapy , Major Histocompatibility Complex/genetics , Polysaccharides/genetics , Proteoglycans/genetics , Sulfotransferases/genetics , Animals , CD11c Antigen/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Dendritic Cells/immunology , Dendritic Cells/pathology , Heparitin Sulfate/pharmacology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Immunity, Cellular/genetics , Immunity, Cellular/immunology , Loss of Function Mutation/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Major Histocompatibility Complex/immunology , Mice , Polysaccharides/antagonists & inhibitors , Proteoglycans/antagonists & inhibitors , Proteoglycans/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
14.
JCI Insight ; 5(4)2020 02 27.
Article in English | MEDLINE | ID: mdl-31961828

ABSTRACT

Pulmonary Langerhans cell histiocytosis (PLCH) is a rare smoking-related lung disease characterized by dendritic cell (DC) accumulation, bronchiolocentric nodule formation, and cystic lung remodeling. Approximately 50% of patients with PLCH harbor somatic BRAF-V600E mutations in cells of the myeloid/monocyte lineage. However, the rarity of the disease and lack of animal models have impeded the study of PLCH pathogenesis. Here, we establish a cigarette smoke-exposed (CS-exposed) BRAF-V600E-mutant mouse model that recapitulates many hallmark characteristics of PLCH. We show that CD11c-targeted expression of BRAF-V600E increases DC responsiveness to stimuli, including the chemokine CCL20, and that mutant cell accumulation in the lungs of CS-exposed mice is due to both increased cellular viability and enhanced recruitment. Moreover, we report that the chemokine CCL7 is secreted from DCs and human peripheral blood monocytes in a BRAF-V600E-dependent manner, suggesting a possible mechanism for recruitment of cells known to dominate PLCH lesions. Inflammatory lesions and airspace dilation in BRAF-V600E mice in response to CS are attenuated by transitioning animals to filtered air and treatment with a BRAF-V600E inhibitor, PLX4720. Collectively, this model provides mechanistic insights into the role of myelomonocytic cells and the BRAF-V600E mutation and CS exposure in PLCH pathogenesis and provides a platform to develop biomarkers and therapeutic targets.


Subject(s)
Histiocytosis, Langerhans-Cell/etiology , Lung Diseases/etiology , Mitogen-Activated Protein Kinases/genetics , Mutation , Smoke/adverse effects , Tobacco Products , Animals , CD11c Antigen/genetics , Disease Models, Animal , Mice , Proto-Oncogene Proteins B-raf/genetics
15.
J Invest Dermatol ; 140(1): 132-142.e3, 2020 01.
Article in English | MEDLINE | ID: mdl-31260672

ABSTRACT

The cell adhesion molecule E-cadherin is a major component of adherens junctions and marks Langerhans cells (LC), the only dendritic cell (DC) population of the epidermis. LC form a dense network and attach themselves to the surrounding keratinocytes via homophilic E-cadherin binding. LC activation, mobilization, and migration require a reduction in LC E-cadherin expression. To determine whether E-cadherin plays a role in regulating LC homeostasis and function, we generated CD11c-specific E-cadherin knockout mice (CD11c-Ecaddel). In the absence of E-cadherin-mediated cell adhesion, LC numbers remained stable and similar as in control mice, even in aged animals. Intriguingly, E-cadherin-deficient LC displayed a dramatically changed morphology characterized by a more rounded cell body and fewer dendrites than wild-type cells. Nevertheless, maturation and migration of LC lacking E-cadherin was not altered, neither under steady-state nor inflammatory conditions. Accordingly, CD11c-Ecaddel and control mice developed comparable contact hypersensitivity reactions and imiquimod-triggered psoriatic skin inflammation, indicating that E-cadherin on LC does not influence their ability to orchestrate T cell-mediated immunity. In conclusion, our data demonstrate that E-cadherin is dispensable to maintain LC in the epidermis and does not regulate LC maturation, migration, and function.


Subject(s)
Cadherins/metabolism , Dermatitis, Contact/immunology , Epidermis/physiology , Langerhans Cells/physiology , Psoriasis/immunology , Animals , CD11c Antigen/genetics , CD11c Antigen/metabolism , Cadherins/genetics , Cell Differentiation , Cell Movement , Cell Shape , Cells, Cultured , Dermatitis, Contact/genetics , Disease Models, Animal , Homeostasis , Humans , Imiquimod , Mice , Mice, Knockout , Psoriasis/genetics
16.
Sci Rep ; 9(1): 16027, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31690792

ABSTRACT

With the increasing prevalence of obesity in women of reproductive age there is a need to understand the ramifications of this on offspring. The purpose of this study is to investigate the programming effects of maternal obesity during preconception and the preconception/gestational period on adiposity and adipose tissue inflammation in offspring using an animal model. Adult female C57Bl/6J mice were assigned either normal diet, high fat diet (HFD) prior to pregnancy, or HFD prior to and through pregnancy. Some offspring were maintained on normal diet while others started HFD later in life. Offspring were assessed for body composition and metabolic responses. Lipid storing tissues were evaluated for expansion and inflammation. Male offspring from the preconception group had the greatest weight gain, most subcutaneous adipose tissue, and largest liver mass when introduced to postnatal HFD. Male offspring of the preconception/gestation group had worsened glucose tolerance and an increase in resident (CD11c-) adipose tissue macrophages (ATMs) when exposed to postnatal HFD. Female offspring had no significant difference in any parameter between the diet treatment groups. In conclusion, this study demonstrates that prenatal and pregnancy windows have independent programming effects on offspring. Preconception exposure affects body composition and adiposity while gestation exposure affects metabolism and tissue immune cell phenotypes.


Subject(s)
Metabolic Diseases/etiology , Obesity/pathology , Animals , Body Weight , CD11c Antigen/deficiency , CD11c Antigen/genetics , CD11c Antigen/metabolism , Diet, High-Fat , Female , Glucose Tolerance Test , Glycerol/blood , Liver/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/veterinary , Pregnancy , Prenatal Exposure Delayed Effects , Subcutaneous Fat/metabolism , Triglycerides/metabolism
17.
Placenta ; 78: 23-28, 2019 03.
Article in English | MEDLINE | ID: mdl-30955707

ABSTRACT

INTRODUCTION: Chronic histiocytic intervillositis of unknown etiology (CIUE) is a non-infectious, most probably immunologic placenta lesion. CIUE is associated with recurrent miscarriage, intrauterine growth restriction and stillbirth. Among the pathologic-anatomic defined placental lesions this entity displays the highest risk of recurrence in following pregnancies (about 67-100%). The histiocytic cells accumulate in the placental blood space but do not infiltrate into the villi or decidua. Sparsely known is the expression profile of these intervillous cells regarding histiocytic markers. METHODS: We analysed 5-22 markers by immunohistochemistry in a total of 41 placenta samples and evaluated decidual, villous and intervillous histiocytic cells. RESULTS: In CIUE, intervillous CD163+ histiocytes over-express CD11c/CD18 and down-regulate CD206/CD209, while CD163+ decidual and Hofbauer cells show low CD11c/CD18 and higher CD206/CD209 protein expressions. DISCUSSION: CD163 expression indicates a M2-like polarisation. CD11c and CD18 form the complement receptor 4 which could be related to a complement mediated trigger for aberrant cell accumulation in CIUE.


Subject(s)
CD11c Antigen/genetics , CD18 Antigens/genetics , Histiocytosis/genetics , Placenta Diseases/genetics , Placenta/metabolism , Receptors, Complement/genetics , Adult , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , CD11c Antigen/metabolism , CD18 Antigens/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Chorionic Villi/immunology , Chorionic Villi/metabolism , Chorionic Villi/pathology , Chronic Disease , Female , Fetal Growth Retardation/genetics , Fetal Growth Retardation/immunology , Fetal Growth Retardation/pathology , Gene Expression Regulation , Gestational Age , Histiocytes/immunology , Histiocytes/metabolism , Histiocytes/pathology , Histiocytosis/immunology , Histiocytosis/metabolism , Histiocytosis/pathology , Humans , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Mannose Receptor , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/metabolism , Placenta/immunology , Placenta/pathology , Placenta Diseases/immunology , Placenta Diseases/metabolism , Placenta Diseases/pathology , Pregnancy , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Complement/metabolism , Retrospective Studies , Transcriptome , Young Adult
18.
Cell Res ; 29(5): 406-419, 2019 05.
Article in English | MEDLINE | ID: mdl-30940876

ABSTRACT

Liver Kinase B1 (LKB1) plays a key role in cellular metabolism by controlling AMPK activation. However, its function in dendritic cell (DC) biology has not been addressed. Here, we find that LKB1 functions as a critical brake on DC immunogenicity, and when lost, leads to reduced mitochondrial fitness and increased maturation, migration, and T cell priming of peripheral DCs. Concurrently, loss of LKB1 in DCs enhances their capacity to promote output of regulatory T cells (Tregs) from the thymus, which dominates the outcome of peripheral immune responses, as suggested by increased resistance to asthma and higher susceptibility to cancer in CD11cΔLKB1 mice. Mechanistically, we find that loss of LKB1 specifically primes thymic CD11b+ DCs to facilitate thymic Treg development and expansion, which is independent from AMPK signalling, but dependent on mTOR and enhanced phospholipase C ß1-driven CD86 expression. Together, our results identify LKB1 as a critical regulator of DC-driven effector T cell and Treg responses both in the periphery and the thymus.


Subject(s)
Dendritic Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes, Regulatory/metabolism , AMP-Activated Protein Kinases , Animals , Asthma/immunology , Asthma/pathology , B7-2 Antigen/metabolism , CD11b Antigen/metabolism , CD11c Antigen/deficiency , CD11c Antigen/genetics , Cell Line, Tumor , Dendritic Cells/cytology , Disease Models, Animal , Melanoma/metabolism , Melanoma/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Phospholipase C beta/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Signal Transduction , T-Lymphocytes, Regulatory/cytology , TOR Serine-Threonine Kinases/metabolism , Thymus Gland/cytology , Thymus Gland/immunology
19.
J Cell Biochem ; 120(2): 1807-1818, 2019 02.
Article in English | MEDLINE | ID: mdl-30873824

ABSTRACT

Integrin alpha x (ITGAX), a member of the integrin family, usually serves as a receptor of the extracellular matrix. Recently, accumulating evidence suggests that ITGAX may be involved in angiogenesis in dendritic cells. Herein, we report a direct role of ITGAX in angiogenesis during tumor development. Overexpression of ITGAX in human umbilical vein endothelial cells (HUVECs) enhanced their proliferation, migration, and tube formation and promoted xenograft ovarian tumor angiogenesis and growth. Further study showed that overexpression of ITGAX activated the PI3k/Akt pathway, leading to the enhanced expression of c-Myc, vascular endothelial growth factor-A (VEGF-A), and VEGF receptor 2 (VEGFR2), whereas, the treatment of cells with PI3K inhibitor diminished these effects. Besides, c-Myc was observed to bind to the VEGF-A promoter. By Co-Immunoprecipitation (Co-IP) assay, we manifested the interaction between ITGAX and VEGFR2 or the phosphorylated VEGFR2. Immunostaining of human ovarian cancer specimens suggested that endothelial cells of micro-blood vessels displayed strong expression of VEGF-A, c-Myc, VEGFR2, and the PI3K signaling molecules. Also, overexpression of ITGAX in HUVECs could stimulate the spheroid formation of ovarian cancer cells. Our study uncovered that ITGAX stimulates angiogenesis through the PI3K/Akt signaling-mediated VEGFR2/VEGF-A overexpression during cancer development.


Subject(s)
CD11c Antigen/genetics , Neovascularization, Pathologic/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cell Movement/genetics , Cell Proliferation/genetics , Chick Embryo , Gene Knockdown Techniques , HEK293 Cells , Heterografts , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Physiologic/genetics , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transfection , Tumor Burden/genetics
20.
J Microbiol Biotechnol ; 29(1): 160-170, 2019 Jan 28.
Article in English | MEDLINE | ID: mdl-30415527

ABSTRACT

The lactic acid bacteria species Lactobacillus plantarum (L. plantarum) has been used extensively for vaccine delivery. Considering to the critical role of dendritic cells in stimulating host immune response, in this study, we constructed a novel CD11c-targeting L. plantarum strain with surface-displayed variable fragments of anti-CD11c, single-chain antibody (scFv-CD11c). The newly designed L. plantarum strain, named 409-aCD11c, could adhere and invade more efficiently to bone marrow-derived DCs (BMDCs) in vitro due to the specific interaction between scFv-CD11c and CD11c located on the surface of BMDCs. After incubation with BMDCs, the 409-aCD11c strain harboring a eukaryotic vector pValac-GFP could lead to more efficient expression of GFP compared with wild-type strains shown by flow cytometry analysis, indicating the enhanced translocation of pValac-GFP from L. plantarum to BMDCs. Similar results were also observed in an in vivo study, which showed that oral administration resulted in efficient expression of GFP in both Peyer's patches (PP) and mesenteric lymph nodes (MLNs) within 7 days after the last administration. In addition, the CD11c-targeting strain significantly promoted the differentiation and maturation of DCs, the differentiation of IL-4⁺ and IL-17A⁺ T helper (Th) cells in MLNs, as well as production of B220⁺ IgA⁺ B cells in the PP. In conclusion, this study developed a novel DC-targeting L. plantarum strain which could increase the ability to deliver eukaryotic expression plasmid to host cells, indicating a promising approach for vaccine study.


Subject(s)
CD11c Antigen/genetics , CD11c Antigen/immunology , Immunomodulation , Lactobacillus plantarum/immunology , Probiotics , Single-Chain Antibodies/immunology , Animals , Antigen-Antibody Reactions , Cell Wall/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Gene Expression , Gene Transfer Techniques , Lactobacillus plantarum/genetics , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mesentery , Mice, Inbred BALB C , Peyer's Patches/immunology , Peyer's Patches/metabolism , Plasmids/genetics , Plasmids/metabolism , Probiotics/administration & dosage , Single-Chain Antibodies/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...