Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Cell Rep ; 43(4): 113981, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38520688

ABSTRACT

Cholera toxin (CT), a bacterial exotoxin composed of one A subunit (CTA) and five B subunits (CTB), functions as an immune adjuvant. CTB can induce production of interleukin-1ß (IL-1ß), a proinflammatory cytokine, in synergy with a lipopolysaccharide (LPS), from resident peritoneal macrophages (RPMs) through the pyrin and NLRP3 inflammasomes. However, how CTB or CT activates these inflammasomes in the macrophages has been unclear. Here, we clarify the roles of inositol-requiring enzyme 1 alpha (IRE1α), an endoplasmic reticulum (ER) stress sensor, in CT-induced IL-1ß production in RPMs. In RPMs, CTB is incorporated into the ER and induces ER stress responses, depending on GM1, a cell membrane ganglioside. IRE1α-deficient RPMs show a significant impairment of CT- or CTB-induced IL-1ß production, indicating that IRE1α is required for CT- or CTB-induced IL-1ß production in RPMs. This study demonstrates the critical roles of IRE1α in activation of both NLRP3 and pyrin inflammasomes in tissue-resident macrophages.


Subject(s)
Cholera Toxin , Endoplasmic Reticulum Stress , Endoribonucleases , Interleukin-1beta , Protein Serine-Threonine Kinases , Interleukin-1beta/metabolism , Animals , Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/metabolism , Endoplasmic Reticulum Stress/drug effects , Mice , Cholera Toxin/pharmacology , Cholera Toxin/metabolism , Inflammasomes/metabolism , Mice, Inbred C57BL , Macrophages/metabolism , Macrophages/drug effects , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Lipopolysaccharides/pharmacology , Endoplasmic Reticulum/metabolism
2.
Front Immunol ; 14: 1250719, 2023.
Article in English | MEDLINE | ID: mdl-37965309

ABSTRACT

Generation of memory B cells is one of the key features of adaptive immunity as they respond rapidly to re-exposure to the antigen and generate functional antibodies. Although the functions of memory B cells are becoming clearer, the regulation of memory B cell generation and maintenance is still not well understood. Here we found that transcription factor SpiB is expressed in some germinal center (GC) B cells and memory B cells and participates in the maintenance of memory B cells. Overexpression and knockdown analyses revealed that SpiB suppresses plasma cell differentiation by suppressing the expression of Blimp1 while inducing Bach2 in the in-vitro-induced germinal center B (iGB) cell culture system, and that SpiB facilitates in-vivo appearance of memory-like B cells derived from the iGB cells. Further analysis in IgG1+ cell-specific SpiB conditional knockout (cKO) mice showed that function of SpiB is critical for the generation of late memory B cells but not early memory B cells or GC B cells. Gene expression analysis suggested that SpiB-dependent suppression of plasma cell differentiation is independent of the expression of Bach2. We further revealed that SpiB upregulates anti-apoptosis and autophagy genes to control the survival of memory B cells. These findings indicate the function of SpiB in the generation of long-lasting memory B cells to maintain humoral memory.


Subject(s)
B-Lymphocytes , Memory B Cells , Mice , Animals , Transcription Factors/metabolism , Germinal Center , Basic-Leucine Zipper Transcription Factors/genetics
3.
FASEB J ; 37(7): e23032, 2023 07.
Article in English | MEDLINE | ID: mdl-37330992

ABSTRACT

The phospholipase A and acyltransferase (PLAAT) family is composed of three isoforms in mice (PLAAT1, 3, and 5), all of which function as phospholipid-metabolizing enzymes exhibiting phospholipase A1 /A2 and acyltransferase activities. Plaat3-deficient (Plaat3-/- ) mice were previously reported to show lean phenotype and remarkable hepatic fat accumulation under high-fat diet (HFD) feeding, while Plaat1-/- mice have not been analyzed. In the present study, we generated Plaat1-/- mice and investigated the effects of PLAAT1 deficiency on HFD-induced obesity, hepatic lipid accumulation, and insulin resistance. After HFD treatment, PLAAT1 deficiency caused a lower body weight gain compared to wild-type mice. Plaat1-/- mice also showed reduced liver weight with negligible hepatic lipid accumulation. In accordance with these findings, PLAAT1 deficiency improved HFD-induced hepatic dysfunction and lipid metabolism disorders. Lipidomics analysis in the liver revealed that in Plaat1-/- mice, the levels of various glycerophospholipids tended to increase, while all classes of lysophospholipids examined tended to decrease, suggesting that PLAAT1 functions as phospholipase A1 /A2 in the liver. Interestingly, the HFD treatment of wild-type mice significantly increased the mRNA level of PLAAT1 in the liver. Furthermore, the deficiency did not appear to elevate the risk of insulin resistance in contrast to PLAAT3 deficiency. These results suggested that the suppression of PLAAT1 improves HFD-induced overweight and concomitant hepatic lipid accumulation.


Subject(s)
Diet, High-Fat , Insulin Resistance , Animals , Mice , Diet, High-Fat/adverse effects , Insulin Resistance/genetics , Lipid Metabolism , Liver/metabolism , Phospholipids/metabolism , Phospholipases/metabolism , Phospholipases/pharmacology , Acyltransferases/genetics , Acyltransferases/metabolism , Mice, Inbred C57BL
4.
Biochem Biophys Res Commun ; 627: 130-136, 2022 10 30.
Article in English | MEDLINE | ID: mdl-36030654

ABSTRACT

D-Allose is classified as a 'rare sugar,' i.e., part of the group of monosaccharides that are present in low quantities in the natural world. D-Allose has been demonstrated to exert many physiological functions. The effects of the rare sugars on immune responses are largely unexplored. Here, we investigated the physiological effects of D-allose on murine dendritic cells' cytokine production. When plasmacytoid dendritic cells (pDCs) were stimulated with a Toll-like receptor 7 (TLR7) ligand, a single-stranded RNA (ssRNA), or a TLR9 ligand, CpG DNA, in the medium containing D-allose, the productions of both interferon-alpha (IFN-α) and interleukin (IL)-12p40 were severely decreased. In contrast, a normal production of these cytokines was observed when pDCs were stimulated with other TLR7 ligands, an imidazoquinoline, or a guanosine analog. In contrast to the pDCs, conventional dendritic cells (cDCs) produced IL-12p40 and tumor necrosis factor-alpha (TNF-α) in response to an imidazoquinoline or CpG DNA even though D-allose was present in the medium. D-Allose did not induce pDC death, and not inhibit the endocytic uptake of fluorophore-labeled CpG DNA into pDCs. These results suggested that D-allose exerts its inhibitory effects after CpG DNA is internalized. We analyzed the TLR7/9 signal-induced activation of downstream signaling molecules in pDCs and observed that when pDCs were stimulated with a ssRNA or CpG DNA, the phosphorylation status of the MAPK family, which includes Erk1/2, JNK/SAPK, and p38 MAPK, was attenuated in the presence of D-allose compared to D-glucose controls. The stimulation of pDCs with an imidazoquinoline induced a strong phosphorylation of these MAPK family members even in the presence of D-allose. These findings reveal that D-allose can inhibit the cytokine production by pDCs stimulated with ssRNA or CpG DNA via an attenuation of the phosphorylation of MAPK family members.


Subject(s)
Toll-Like Receptor 7 , Toll-Like Receptor 9 , Animals , Cytokines , DNA , Dendritic Cells , Glucose/pharmacology , Immunity , Ligands , Mice
5.
Int Immunol ; 34(3): 159-172, 2022 02 23.
Article in English | MEDLINE | ID: mdl-34734243

ABSTRACT

Type I IFNs (IFN-α and IFN-ß), immunomodulatory cytokines secreted from activated plasmacytoid dendritic cells (pDCs), contribute to the innate defense against pathogenic infections and the pathogenesis of the autoimmune disease psoriasis vulgaris. A previous study has shown that an E26 transformation-specific (Ets) family transcription factor Spi-B can transactivate the type I IFN promoter in synergy with IFN regulatory factor (IRF)-7 and is required for type I IFN production in pDCs. However, the mechanism of negative regulation of type I IFNs by pDCs remains unknown. In this study, we report that a basic leucine zipper (bZip) transcription factor v-maf musculoaponeurotic fibrosarcoma oncogene homolog B (MafB) suppresses the induction of type I IFNs in pDCs. The elevated expression of MafB inhibited the transactivation of type I IFN genes in a dose-dependent manner. At the molecular level, MafB interacted with the Ets domain of Spi-B and interfered with IRF-7-Spi-B complexation. Decreased MafB mRNA expression and degradation of MafB protein in the early phase of immune responses led to the enhancement of type I IFNs in pDCs. In vivo studies indicated that MafB is involved in resistance against imiquimod-induced psoriasis-like skin inflammation. Overall, these findings demonstrate that MafB acts as a negative regulator of type I IFN induction in pDCs and plays an important role in maintaining immune homeostasis.


Subject(s)
Interferon Type I , Psoriasis , Dendritic Cells , Humans , Interferon Type I/metabolism , Interferon-alpha/metabolism , MafB Transcription Factor/genetics , MafB Transcription Factor/metabolism , Promoter Regions, Genetic
6.
Clin Exp Nephrol ; 26(3): 226-233, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34698914

ABSTRACT

BACKGROUND: Galectin-9 (Gal-9) is a multifunctional lectin that moderates inflammation and organ damage. In this study, we tested whether Gal-9 has a protective role in the pathogenesis of endotoxemic acute kidney injury. METHODS: We examined the levels of Gal-9 in control mice after lipopolysaccharide (LPS) administration. We developed Gal-9 knockout (KO) mice that lack Gal-9 systemically and evaluated the role of Gal-9 in LPS-induced proinflammatory cytokines, vascular permeability, and renal injury. RESULTS: Gal-9 levels were increased in the plasma, kidney, and spleen within 4 h after LPS administration to wild-type mice. Gal-9 deficiency did not affect the LPS-induced increase in plasma tumor necrosis factor-α levels at 1 h or vascular permeability at 6 h. Lower urine volume and reduced creatinine clearance were observed in Gal-9-KO mice compared with wild-type mice after LPS administration. Gal-9-KO mice had limited improvement in urine volume after fluid resuscitation compared with wild-type mice. LPS reduced the body temperature 12 h after its administration. Hypothermia had disappeared in wild-type mice by 24 h, whereas it was sustained until 24 h in Gal-9-KO mice. Importantly, maintaining body temperature in Gal-9-KO mice improved the response of urine flow to fluid resuscitation. CONCLUSION: Deficiency in Gal-9 worsened LPS-induced hypothermia and kidney injury in mice. The accelerated hypothermia induced by Gal-9 deficiency contributed to the blunted response to fluid resuscitation.


Subject(s)
Acute Kidney Injury , Hypothermia, Induced , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Animals , Galectins/adverse effects , Galectins/genetics , Humans , Kidney/pathology , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Mice, Knockout
7.
Biochem Biophys Res Commun ; 525(2): 477-482, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32111355

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are characterized by an exclusive expression of nucleic acid sensing Toll-like receptor 7 (TLR7) and TLR9, and production of high amounts of type I interferon (IFN) in response to TLR7/9 signaling. This function is crucial for both antiviral immunity and the pathogenesis of autoimmune diseases. An Ets family transcription factor, i.e., Spi-B (which is highly expressed in pDCs) is required for TLR7/9 signal-induced type I IFN production and can transactivate IFN-α promoter in synergy with IFN regulatory factor-7 (IRF-7). Herein, we analyzed how Spi-B contributes to the transactivation of the Ifna4 promoter. We performed deletion and/or mutational analyses of the Ifna4 promoter and an electrophoretic mobility shift assay (EMSA) and observed an Spi-B binding site in close proximity to the IRF-7 binding site. The EMSA results also showed that the binding of Spi-B to the double-stranded DNA probe potentiated the recruitment of IRF-7 to its binding site. We also observed that the association of Spi-B with transcriptional coactivator p300 was required for the Spi-B-induced synergistic enhancement of the Ifna4 promoter activity by Spi-B. These results clarify the molecular mechanism of action of Spi-B in the transcriptional activation of the Ifna4 promoter.


Subject(s)
Interferon-alpha/genetics , Proto-Oncogene Proteins c-ets/metabolism , Transcriptional Activation , Animals , E1A-Associated p300 Protein/metabolism , HEK293 Cells , Humans , Mice , Mutation , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-ets/genetics
8.
Int Immunol ; 31(10): 657-668, 2019 09 18.
Article in English | MEDLINE | ID: mdl-30689886

ABSTRACT

Cholera toxin B (CTB) is a subunit of cholera toxin, a bacterial enterotoxin secreted by Vibrio cholerae and also functions as an immune adjuvant. However, it remains unclear how CTB activates immune cells. We here evaluated whether or how CTB induces production of a pro-inflammatory cytokine, interleukin-1ß (IL-1ß). CTB induced IL-1ß production not only from bone marrow-derived macrophages (BMMs) but also from resident peritoneal macrophages in synergy with O111:B4-derived lipopolysaccharide (LPS O111:B4) that can bind to CTB. Meanwhile, when prestimulated with O55:B5-derived LPS (LPS O55:B5) that fails to bind to CTB, resident peritoneal macrophages, but not BMMs, produced IL-1ß in response to CTB. The CTB-induced IL-1ß production in synergy with LPS in both peritoneal macrophages and BMMs was dependent on ganglioside GM1, which is required for internalization of CTB. Notably, not only the NLRP3 inflammasome but also the pyrin inflammasome were involved in CTB-induced IL-1ß production from resident peritoneal macrophages, while only the NLRP3 inflammasome was involved in that from BMMs. In response to CTB, a Rho family small GTPase, RhoA, which activates pyrin inflammasome upon various kinds of biochemical modification, increased its phosphorylation at serine-188 in a GM1-dependent manner. This phosphorylation as well as CTB-induced IL-1ß productions were dependent on protein kinase A (PKA), indicating critical involvement of PKA-dependent RhoA phosphorylation in CTB-induced IL-1ß production. Taken together, these results suggest that CTB, incorporated through GM1, can activate resident peritoneal macrophages to produce IL-1ß in synergy with LPS through novel mechanisms in which pyrin as well as NLRP3 inflammasomes are involved.


Subject(s)
Cholera Toxin/pharmacology , Inflammasomes/drug effects , Interleukin-1beta/biosynthesis , Macrophages, Peritoneal/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Pyrin/immunology , Animals , Humans , Inflammasomes/immunology , Macrophages, Peritoneal/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology
9.
Cell Physiol Biochem ; 50(5): 1856-1868, 2018.
Article in English | MEDLINE | ID: mdl-30396167

ABSTRACT

BACKGROUND/AIMS: Galectin-9 is a soluble immune modulator with versatile functions, including a role as an immune checkpoint molecule. Therefore, the amount of galectin-9 in the blood may reflect an individual's immunological balance. Many studies have conducted galectin-9 measurements; however, the reported galectin-9 concentration in the blood varies greatly, even within healthy controls. This study investigates the variation between the reported and actual concentrations of galectin-9. METHODS: A GalPharma ELISA and an R&D Systems ELISA kit were directly compared using the same set of plasma and a series of recombinant galectins, including degraded galectin-9. Furthermore, galectin-9 in plasma was concentrated using anti-galectin-9 antibody-conjugated beads, and subjected to western blotting to estimate the quantity and integrity of galectin-9 and assess the consistency of ELISA measurements. RESULTS: The R&D Systems' ELISA indicated a 50-fold higher median concentration of plasma galectin-9 than that indicated by the GalPharma ELISA. This variation is due to aberrantly enhanced reactivity of the R&D Systems' ELISA to degraded galectin-9 present in small quantities in the plasma. The GalPharma ELISA could detect only intact galectin-9 and its results correlated well with the plasma galectin-9 level obtained by western blotting. CONCLUSION: ELISA kits from R&D Systems reacts aberrantly higher against degraded galectin-9 than the intact galectin-9. Therefore, the existence of a small amount of degraded galectin-9 in a test sample hinders the quantification. As galectin-9 is a fragile protein, this is a serious concern when using this kit. Based on quantifications from the GalPharma ELISA, the median (25th-75th percentiles) galectin-9 concentration in healthy subjects in the current study cohort was calculated as 110 pg/mL (67 -154 pg/mL).


Subject(s)
Galectins/blood , Liver Failure, Acute/blood , Enzyme-Linked Immunosorbent Assay , Galectins/metabolism , Humans , Limit of Detection , Liver Failure, Acute/diagnosis , Reagent Kits, Diagnostic
10.
Proc Natl Acad Sci U S A ; 115(33): 8418-8423, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30061415

ABSTRACT

The local environment is crucial for shaping the identities of tissue-resident macrophages (Mϕs). When hemorrhage occurs in damaged tissues, hemoglobin induces differentiation of anti-inflammatory Mϕs with reparative function. Mucosal bleeding is one of the pathological features of inflammatory bowel diseases. However, the heme-mediated mechanism modulating activation of intestinal innate immune cells remains poorly understood. Here, we show that heme regulates gut homeostasis through induction of Spi-C in intestinal CX3CR1high Mϕs. Intestinal CX3CR1high Mϕs highly expressed Spi-C in a heme-dependent manner, and myeloid lineage-specific Spic-deficient (Lyz2-cre; Spicflox/flox ) mice showed severe intestinal inflammation with an increased number of Th17 cells during dextran sodium sulfate-induced colitis. Spi-C down-regulated the expression of a subset of Toll-like receptor (TLR)-inducible genes in intestinal CX3CR1high Mϕs to prevent colitis. LPS-induced production of IL-6 and IL-1α, but not IL-10 and TNF-α, by large intestinal Mϕs from Lyz2-cre; Spicflox/flox mice was markedly enhanced. The interaction of Spi-C with IRF5 was linked to disruption of the IRF5-NF-κB p65 complex formation, thereby abrogating recruitment of IRF5 and NF-κB p65 to the Il6 and Il1a promoters. Collectively, these results demonstrate that heme-mediated Spi-C is a key molecule for the noninflammatory signature of intestinal Mϕs by suppressing the induction of a subset of TLR-inducible genes through binding to IRF5.


Subject(s)
Colitis/drug therapy , Heme/pharmacology , Intestines/immunology , Macrophages/immunology , Animals , CX3C Chemokine Receptor 1/physiology , Cytokines/biosynthesis , DNA-Binding Proteins/physiology , Dextran Sulfate/toxicity , Iron, Dietary/administration & dosage , Mice , Mice, Inbred C57BL , Toll-Like Receptors/physiology , Transcription Factor RelA/physiology
12.
Methods Mol Biol ; 1423: 247-53, 2016.
Article in English | MEDLINE | ID: mdl-27142021

ABSTRACT

Dendritic cells (DCs) are one of the key populations controlling immune responses. To establish a cell depletion system in vivo, human diphtheria toxin (DT) receptor (DTR) is transduced to the mice in which DTR is expressed under the control of a specific promoter. In these mice, DTR-expressing cells are inducibly depleted after DT injection. Using this system, analysis of mouse models in which DTR was expressed under the CD11c promoter has contributed to our knowledge of DC biology by depleting CD11c(+) cells. Other mouse models to inducibly eliminate specific DC subsets upon DT treatment have been also generated. Here, we describe a new mouse model in which the XCR1(+) DC subset is inducibly and transiently depleted in vivo.


Subject(s)
CD11c Antigen/genetics , Dendritic Cells/immunology , Diphtheria Toxin/pharmacology , Heparin-binding EGF-like Growth Factor/metabolism , Receptors, Chemokine/metabolism , Animals , Gene Knock-In Techniques , Heparin-binding EGF-like Growth Factor/genetics , Humans , Mice , Mice, Transgenic , Models, Animal , Promoter Regions, Genetic , Receptors, Chemokine/genetics
13.
Sci Rep ; 6: 23505, 2016 Mar 23.
Article in English | MEDLINE | ID: mdl-27005831

ABSTRACT

Intestinal immune homeostasis requires dynamic crosstalk between innate and adaptive immune cells. Dendritic cells (DCs) exist as multiple phenotypically and functionally distinct sub-populations within tissues, where they initiate immune responses and promote homeostasis. In the gut, there exists a minor DC subset defined as CD103(+)CD11b(-) that also expresses the chemokine receptor XCR1. In other tissues, XCR1(+) DCs cross-present antigen and contribute to immunity against viruses and cancer, however the roles of XCR1(+) DCs and XCR1 in the intestine are unknown. We showed that mice lacking XCR1(+) DCs are specifically deficient in intraepithelial and lamina propria (LP) T cell populations, with remaining T cells exhibiting an atypical phenotype and being prone to death, and are also more susceptible to chemically-induced colitis. Mice deficient in either XCR1 or its ligand, XCL1, similarly possess diminished intestinal T cell populations, and an accumulation of XCR1(+) DCs in the gut. Combined with transcriptome and surface marker expression analysis, these observations lead us to hypothesise that T cell-derived XCL1 facilitates intestinal XCR1(+) DC activation and migration, and that XCR1(+) DCs in turn provide support for T cell survival and function. Thus XCR1(+) DCs and the XCR1/XCL1 chemokine axis have previously-unappreciated roles in intestinal immune homeostasis.


Subject(s)
Chemokines, C/metabolism , Dendritic Cells/physiology , Intestines/immunology , Receptors, Chemokine/metabolism , T-Lymphocytes/cytology , Animals , Cell Movement , Cell Proliferation , Cell Survival , Cells, Cultured , Chemokines, C/deficiency , Cross-Priming , Dendritic Cells/immunology , Gene Expression Profiling/methods , Gene Expression Regulation , Homeostasis , Intestines/cytology , Mice , Receptors, Chemokine/deficiency , T-Lymphocytes/immunology
14.
Proc Natl Acad Sci U S A ; 113(4): 1044-9, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26755602

ABSTRACT

Dendritic cells (DCs) are antigen-presenting cells specialized for activating T cells to elicit effector T-cell functions. Cross-presenting DCs are a DC subset capable of presenting antigens to CD8(+) T cells and play critical roles in cytotoxic T-cell-mediated immune responses to microorganisms and cancer. Although their importance is known, the spatiotemporal dynamics of cross-presenting DCs in vivo are incompletely understood. Here, we study the T-cell zone in skin-draining lymph nodes (SDLNs) and find it is compartmentalized into regions for CD8(+) T-cell activation by cross-presenting DCs that express the chemokine (C motif) receptor 1 gene, Xcr1 and for CD4(+) T-cell activation by CD11b(+) DCs. Xcr1-expressing DCs in the SDLNs are composed of two different populations: migratory (CD103(hi)) DCs, which immigrate from the skin, and resident (CD8α(hi)) DCs, which develop in the nodes. To characterize the dynamic interactions of these distinct DC populations with CD8(+) T cells during their activation in vivo, we developed a photoconvertible reporter mouse strain, which permits us to distinctively visualize the migratory and resident subsets of Xcr1-expressing DCs. After leaving the skin, migratory DCs infiltrated to the deep T-cell zone of the SDLNs over 3 d, which corresponded to their half-life in the SDLNs. Intravital two-photon imaging showed that after soluble antigen immunization, the newly arriving migratory DCs more efficiently form sustained conjugates with antigen-specific CD8(+) T cells than other Xcr1-expressing DCs in the SDLNs. These results offer in vivo evidence for differential contributions of migratory and resident cross-presenting DCs to CD8(+) T-cell activation.


Subject(s)
Cross-Priming , Dendritic Cells/immunology , Lymph Nodes/immunology , Skin/immunology , Animals , Antigens, CD/analysis , CD8-Positive T-Lymphocytes/immunology , Cell Movement , Immunization , Integrin alpha Chains/analysis , Mice , Mice, Inbred C57BL , Receptors, Chemokine/analysis , Receptors, Chemokine/physiology
15.
J Exp Med ; 211(12): 2425-38, 2014 Nov 17.
Article in English | MEDLINE | ID: mdl-25385757

ABSTRACT

Medullary thymic epithelial cells (mTECs) expressing the autoimmune regulator AIRE and various tissue-specific antigens (TSAs) are critical for preventing the onset of autoimmunity and may attenuate tumor immunity. However, molecular mechanisms controlling mTEC development remain elusive. Here, we describe the roles of the transcription factor Spi-B in mTEC development. Spi-B is rapidly up-regulated by receptor activator of NF-κB ligand (RANKL) cytokine signaling, which triggers mTEC differentiation, and in turn up-regulates CD80, CD86, some TSAs, and the natural inhibitor of RANKL signaling, osteoprotegerin (OPG). Spi-B-mediated OPG expression limits mTEC development in neonates but not in embryos, suggesting developmental stage-specific negative feedback regulation. OPG-mediated negative regulation attenuates cellularity of thymic regulatory T cells and tumor development in vivo. Hence, these data suggest that this negative RANKL-Spi-B-OPG feedback mechanism finely tunes mTEC development and function and may optimize the trade-off between prevention of autoimmunity and induction of antitumor immunity.


Subject(s)
Epithelial Cells/immunology , Immune Tolerance/immunology , Proto-Oncogene Proteins c-ets/immunology , Thymus Gland/immunology , Animals , Animals, Newborn , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Epithelial Cells/metabolism , Feedback, Physiological , Female , Gene Expression/immunology , Immune Tolerance/genetics , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Osteoprotegerin/genetics , Osteoprotegerin/immunology , Osteoprotegerin/metabolism , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , RANK Ligand/immunology , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/immunology , Receptor Activator of Nuclear Factor-kappa B/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , Thymus Gland/metabolism , NF-kappaB-Inducing Kinase
16.
Cell Rep ; 8(4): 974-82, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25127135

ABSTRACT

Inflammasome-mediated caspase-1 activation is involved in cell death and the secretion of the proinflammatory cytokine interleukin-1ß (IL-1ß). Although the dynamics of caspase-1 activation, IL-1ß secretion, and cell death have been examined with bulk assays in population-level studies, they remain poorly understood at the single-cell level. In this study, we conducted single-cell imaging using a genetic fluorescence resonance energy transfer sensor that detects caspase-1 activation. We determined that caspase-1 exhibits all-or-none (digital) activation at the single-cell level, with similar activation kinetics irrespective of the type of inflammasome or the intensity of the stimulus. Real-time concurrent detection of caspase-1 activation and IL-1ß release demonstrated that dead macrophages containing activated caspase-1 release a local burst of IL-1ß in a digital manner, which identified these macrophages as the main source of IL-1ß within cell populations. Our results highlight the value of single-cell analysis in enhancing understanding of the inflammasome system and chronic inflammatory diseases.


Subject(s)
Caspase 1/physiology , Inflammasomes/metabolism , Animals , Cells, Cultured , Enzyme Activation , Humans , Interleukin-1beta/physiology , Kinetics , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/immunology , Mice, Transgenic , Signal Transduction , Single-Cell Analysis
17.
J Immunol ; 190(11): 5609-19, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23630347

ABSTRACT

A key goal of vaccine immunotherapy is the generation of long-term memory CD8(+) T cells capable of mediating immune surveillance. We discovered a novel intercellular pathway governing the development of potent memory CD8(+) T cell responses against cell-associated Ags that is mediated through cross-presentation by XCR1(+) dendritic cells (DCs). Generation of CD8(+) memory T cells against tumor cells pulsed with an invariant NKT cell ligand depended on cross-talk between XCR1(+) and plasmacytoid DCs that was regulated by IFN-α/IFN-αR signals. IFN-α production by plasmacytoid DCs was stimulated by an OX40 signal from the invariant NKT cells, as well as an HMGB1 signal from the dying tumor cells. These findings reveal a previously unknown pathway of intercellular collaboration for the generation of tumor-specific CD8(+) memory T cells that can be exploited for strategic vaccination in the setting of tumor immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Communication/immunology , Dendritic Cells/immunology , Immunologic Memory , Natural Killer T-Cells/immunology , Animals , Cell Line, Tumor , Chemotaxis/immunology , Dendritic Cells/metabolism , Interferon Type I/immunology , Interferon Type I/metabolism , Interleukin-12/biosynthesis , Ligands , Mice , Neoplasms/immunology , Signal Transduction
18.
J Immunol ; 190(12): 6071-82, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23670193

ABSTRACT

Dendritic cells (DCs) consist of various subsets that play crucial roles in linking innate and adaptive immunity. In the murine spleen, CD8α(+) DCs exhibit a propensity to ingest dying/dead cells, produce proinflammatory cytokines, and cross-present Ags to generate CD8(+) T cell responses. To track and ablate CD8α(+) DCs in vivo, we generated XCR1-venus and XCR1-DTRvenus mice, in which genes for a fluorescent protein, venus, and a fusion protein consisting of diphtheria toxin receptor and venus were knocked into the gene locus of a chemokine receptor, XCR1, which is highly expressed in CD8α(+) DCs. In both mice, venus(+) cells were detected in the majority of CD8α(+) DCs, but they were not detected in any other cells, including splenic macrophages. Venus(+)CD8α(+) DCs were superior to venus(-)CD8α(+) DCs with regard to their cytokine-producing ability in response to TLR stimuli. In other tissues, venus(+) cells were found primarily in lymph node (LN)-resident CD8α(+), LN migratory and peripheral CD103(+) DCs, which are closely related to splenic CD8α(+) DCs, although some thymic CD8α(-)CD11b(-) and LN CD103(-)CD11b(-) DCs were also venus(+). In response to dsRNAs, diphtheria toxin-treated XCR1-DTR mice showed impaired CD8(+) T cell responses, with retained cytokine and augmented CD4(+) T cell responses. Furthermore, Listeria monocytogenes infection and anti-L. monocytogenes CD8(+) T cell responses were defective in diphtheria toxin-treated XCR1-DTRvenus mice. Thus, XCR1-expressing DCs were required for dsRNA- or bacteria-induced CD8(+) T cell responses. XCR1-venus and XCR1-DTRvenus mice should be useful for elucidating the functions and behavior of XCR1-expressing DCs, including CD8α(+) and CD103(+) DCs, in lymphoid and peripheral tissues.


Subject(s)
Cross-Priming/immunology , Dendritic Cells/immunology , Receptors, Chemokine/immunology , Animals , Antigen Presentation/immunology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Separation , Dendritic Cells/metabolism , Flow Cytometry , Gene Knock-In Techniques , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Receptors, Chemokine/metabolism
19.
Immunity ; 38(3): 450-60, 2013 Mar 21.
Article in English | MEDLINE | ID: mdl-23453632

ABSTRACT

Sjögren's syndrome (SS) is an autoimmune disease characterized by exocrinopathy that leads to dry eye and mouth. Although lymphocyte infiltration into exocrine glands and the generation of autoantibodies have been reported in SS, its pathogenic mechanism remains elusive. Here, we show that mice lacking the transcriptional regulator IκB-ζ developed SS-like inflammation characterized by lymphocyte-infiltrated dacryoadenitis and SS-associated autoantibodies. In particular, epithelial cells, but not hematopoietic cells, lacking IκB-ζ were essential for the development of inflammation. IκB-ζ-deficient epithelial cells in the lacrimal glands exhibited enhanced apoptosis even in the absence of lymphocytes. Administration of caspase inhibitors ameliorated the inflammation, indicating the critical role of caspase-mediated apoptosis. Furthermore, epithelial cell-specific STAT3-deficient mice developed SS-like inflammation with impaired IκB-ζ expression in the lacrimal glands. Thus, this study reveals a pathogenic mechanism of SS in which dysfunction of epithelial cells caused by disruption of STAT3-mediated IκB-ζ induction elicits the activation of self-reactive lymphocytes.


Subject(s)
Apoptosis/immunology , Autoimmune Diseases/immunology , Epithelial Cells/immunology , STAT3 Transcription Factor/immunology , Sjogren's Syndrome/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Apoptosis/genetics , Autoimmune Diseases/genetics , Autoimmune Diseases/metabolism , Epithelial Cells/metabolism , Female , Immunohistochemistry , In Situ Nick-End Labeling , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Lacrimal Apparatus/immunology , Lacrimal Apparatus/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/immunology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Sjogren's Syndrome/genetics , Sjogren's Syndrome/metabolism
20.
Blood ; 120(24): 4733-43, 2012 Dec 06.
Article in English | MEDLINE | ID: mdl-23065153

ABSTRACT

Plasmacytoid dendritic cells (pDCs), originating from hematopoietic progenitor cells in the BM, are a unique dendritic cell subset that can produce large amounts of type I IFNs by signaling through the nucleic acid-sensing TLR7 and TLR9 (TLR7/9). The molecular mechanisms for pDC function and development remain largely unknown. In the present study, we focused on an Ets family transcription factor, Spi-B, that is highly expressed in pDCs. Spi-B could transactivate the type I IFN promoters in synergy with IFN regulatory factor 7 (IRF-7), which is an essential transcription factor for TLR7/9-induced type I IFN production in pDCs. Spi-B-deficient pDCs and mice showed defects in TLR7/9-induced type I IFN production. Furthermore, in Spi-B-deficient mice, BM pDCs were decreased and showed attenuated expression of a set of pDC-specific genes whereas peripheral pDCs were increased; this uneven distribution was likely because of defective retainment of mature nondividing pDCs in the BM. The expression pattern of cell-surface molecules in Spi-B-deficient mice indicated the involvement of Spi-B in pDC development. The developmental defects of pDCs in Spi-B-deficient mice were more prominent in the BM than in the peripheral lymphoid organs and were intrinsic to pDCs. We conclude that Spi-B plays critical roles in pDC function and development.


Subject(s)
Bone Marrow Cells/metabolism , Dendritic Cells/metabolism , Gene Expression Profiling , Proto-Oncogene Proteins c-ets/genetics , Animals , Base Sequence , Bone Marrow Cells/physiology , Dendritic Cells/physiology , Flow Cytometry , HEK293 Cells , Humans , Interferon Type I/genetics , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Mutation , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-ets/metabolism , Proto-Oncogene Proteins c-ets/physiology , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/physiology , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/physiology , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...