Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Am J Physiol Lung Cell Mol Physiol ; 325(5): L604-L616, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37724373

ABSTRACT

Basic leucine zipper transcription factor ATF-like 2 (BATF2) is a transcription factor that is emerging as an important regulator of the innate immune system. BATF2 is among the top upregulated genes in human alveolar macrophages treated with LPS, but the signaling pathways that induce BATF2 expression in response to Gram-negative stimuli are incompletely understood. In addition, the role of BATF2 in the host response to pulmonary infection with a Gram-negative pathogen like Klebsiella pneumoniae (Kp) is not known. We show that induction of Batf2 gene expression in macrophages in response to Kp in vitro requires TRIF and type I interferon (IFN) signaling, but not MyD88 signaling. Analysis of the impact of BATF2 deficiency on macrophage effector functions in vitro showed that BATF2 does not directly impact macrophage phagocytic uptake and intracellular killing of Kp. However, BATF2 markedly enhanced macrophage proinflammatory gene expression and Kp-induced cytokine responses. In vivo, Batf2 gene expression was elevated in lung tissue of wild-type (WT) mice 24 h after pulmonary Kp infection, and Kp-infected BATF2-deficient (Batf2-/-) mice displayed an increase in bacterial burden in the lung, spleen, and liver compared with WT mice. WT and Batf2-/- mice showed similar recruitment of leukocytes following infection, but in line with in vitro observations, proinflammatory cytokine levels in the alveolar space were reduced in Batf2-/- mice. Altogether, these results suggest that BATF2 enhances proinflammatory cytokine responses in macrophages in response to Kp and contributes to the early host defense against pulmonary Kp infection.NEW & NOTEWORTHY This study investigates the signaling pathways that mediate induction of BATF2 expression downstream of TLR4 and also the impact of BATF2 on the host defense against pulmonary Kp infection. We demonstrate that Kp-induced upregulation of BATF2 in macrophages requires TRIF and type I IFN signaling. We also show that BATF2 enhances Kp-induced macrophage cytokine responses and that BATF2 contributes to the early host defense against pulmonary Kp infection.


Subject(s)
Klebsiella Infections , Pneumonia , Animals , Humans , Mice , Adaptor Proteins, Vesicular Transport/metabolism , Cytokines/metabolism , Klebsiella Infections/microbiology , Klebsiella pneumoniae , Macrophages/metabolism , Mice, Inbred C57BL , Pneumonia/metabolism
2.
Life Sci Alliance ; 6(12)2023 12.
Article in English | MEDLINE | ID: mdl-37734869

ABSTRACT

MAP4K1 has been identified as a cancer immunotherapy target. Whether and how cancer cell-intrinsic MAP4K1 contributes to glioblastoma multiforme (GBM) progression remains unclear. We found that MAP4K1 was highly expressed in the glioma cells of human GBM specimens. High levels of MAP4K1 mRNA were prevalent in IDH-WT and 1p/19q non-codeletion gliomas and correlated with poor prognosis of patients. MAP4K1 silencing inhibited GBM cell proliferation and glioma growth. Transcriptome analysis of GBM cells and patient samples showed that MAP4K1 modulated cytokine‒cytokine receptor interactions and chemokine signaling pathway, including IL-18R and IL-6R Importantly, MAP4K1 loss down-regulated membrane-bound IL-18R/IL-6R by inhibiting the PI3K-AKT pathway, whereas MAP4K1 restoration rescued this phenotype and therefore GBM cell proliferation. MAP4K1 deficiency abolished GBM cell pro-proliferation responses to IL-18, suggesting an oncogenic role of MAP4K1 via the intrinsic IL-18/IL-18R pathway. In addition, GBM cell-derived MAP4K1 impaired T-cell migration and reduced CD8+ T-cell infiltration in mouse glioma models. Together, our findings provide novel insight into the pathological significance of GBM cell-intrinsic MAP4K1 in driving tumor growth and immune evasion by remodeling cytokine-chemokine networks.


Subject(s)
Glioblastoma , Glioma , Animals , Humans , Mice , Cytokines , Disease Models, Animal , Glioblastoma/genetics , Interleukin-18/genetics , Phosphatidylinositol 3-Kinases
3.
Front Cell Infect Microbiol ; 13: 1150658, 2023.
Article in English | MEDLINE | ID: mdl-37056705

ABSTRACT

Introduction: Klebsiella pneumoniae (Kp) is a common cause of hospital-acquired pneumonia. Although previous studies have suggested that evasion of phagocytic uptake is a virulence determinant of Kp, few studies have examined phagocytosis sensitivity in clinical Kp isolates. Methods: We screened 19 clinical respiratory Kp isolates that were previously assessed for mucoviscosity for their sensitivity to macrophage phagocytic uptake, and evaluated phagocytosis as a functional correlate of in vivo Kp pathogenicity. Results: The respiratory Kp isolates displayed heterogeneity in the susceptibility to macrophage phagocytic uptake, with 14 out of 19 Kp isolates displaying relative phagocytosis-sensitivity compared to the reference Kp strain ATCC 43816, and 5 out of 19 Kp isolates displaying relative phagocytosis-resistance. Intratracheal infection with the non-mucoviscous phagocytosis-sensitive isolate S17 resulted in a significantly lower bacterial burden compared to infection with the mucoviscous phagocytosis-resistant isolate W42. In addition, infection with S17 was associated with a reduced inflammatory response, including reduced bronchoalveolar lavage fluid (BAL) polymorphonuclear (PMN) cell count, and reduced BAL TNF, IL-1ß, and IL-12p40 levels. Importantly, host control of infection with the phagocytosis-sensitive S17 isolate was impaired in alveolar macrophage (AM)-depleted mice, whereas AM-depletion had no significant impact on host defense against infection with the phagocytosis-resistant W42 isolate. Conclusion: Altogether, these findings show that phagocytosis is a primary determinant of pulmonary clearance of clinical Kp isolates.


Subject(s)
Klebsiella Infections , Klebsiella pneumoniae , Animals , Mice , Lung/microbiology , Phagocytosis , Macrophages, Alveolar , Neutrophils , Klebsiella Infections/microbiology , Anti-Bacterial Agents/pharmacology
4.
Front Pharmacol ; 13: 807440, 2022.
Article in English | MEDLINE | ID: mdl-35370674

ABSTRACT

Antibiotics resistance is one of the most significant public health threats globally. Strategies that strengthen host defenses to control pathogen infection has become a hot research field. Macrophages are part of early host defense mechanisms, and are activated via host pattern recognition receptors (PRRs), such as Toll-like receptor 4 (TLR4), which then facilitates phagocytosis and elimination of invading pathogens. However, few activators of PRRs have been approved for clinical use because of their toxic effects. This study aimed to investigate whether Strongylocentrotus nudus eggs polysaccharide (SEP), a non-toxic extract from seafood, contributes to host defense against bacterial infection. Results showed that SEP promoted bacterial clearance by enhancing phagocytosis by macrophages during E. coli infection in vitro, but was inhibited by TLR4 specific inhibitor TAK-242, STAT3 inhibitor Stattic or blockade of CD64. In addition, SEP protected mice from E. coli induced mortality, reduced pulmonary inflammation and inhibited dissemination of bacteria to organs, while TAK-242 retarded the protection of SEP. Overall, SEP strengthened innate host defense and improved the outcome in bacterial infection, suggesting that SEP could be used as a potential immunomodulator in host-directed therapies.

5.
Chin J Nat Med ; 19(6): 432-441, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34092294

ABSTRACT

Esculetin, a natural derivative from the traditional and widely-used Chinese medicinal herb Cortex Fraxini, has a variety of pharmacological effects, especially in anti-inflammation. However, it is not clear whether esculetin has a therapeutic effect on sepsis. This study aimed to investigate the anti-inflammatory and protective effects of esculetin on early sepsis. The results showed that the lung injury was significantly relieved with the treatment of esculetin, accompanied with the restrained production of inflammatory factors including IL-1ß, IL-6, TNF-α, CCL2 and iNOS during the early phase of E.coli-induced sepsis. Of note, activation of NF-κB and STAT1/STAT3 signals, the main upstream signals of many inflammatory factors, were attenuated by esculetin in both lung tissues from septic mice and LPS-stimulated macrophage. These findings suggested that the protection of esculetin against early sepsis should be related to its anti-inflammatory effect, which was at least partly due to its inhibition on NF-κB and STAT1/STAT3 signaling pathway in macrophage. Thus, esculetin could serve as a potential therapeutic agent by rebalancing innate immune response in macrophage for the treatment of early sepsis.


Subject(s)
NF-kappa B , Sepsis , Signal Transduction/drug effects , Umbelliferones/pharmacology , Animals , Inflammation/drug therapy , Lipopolysaccharides , Mice , NF-kappa B/antagonists & inhibitors , STAT1 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/antagonists & inhibitors , Sepsis/drug therapy
6.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 36(11): 1073-1076, 2019 Nov 10.
Article in Chinese | MEDLINE | ID: mdl-31703128

ABSTRACT

OBJECTIVE: To assess the association of single nucleotide polymorphisms of multidrug resistance gene 1 (MDR1) with refractory epilepsy in children. METHODS: Peripheral blood samples were collected from 200 children with epilepsy and 100 healthy controls. Genomic DNA was extracted and subjected to PCR amplification, agarose gel electrophoresis and target site sequencing. Genotypes of rs1922242, rs2235048, rs10808072, rs868755 and rs1202184 loci of the MDR1 gene were analyzed. RESULTS: No significant difference was found in genotypic distribution and allelic frequencies of the rs1922242, rs2235048, rs10808072 and rs868755 loci between the drug-resistant and drug-sensitive groups. For the rs1202184 locus, a significant difference in genotypic distribution was found (P=0.008). No significant difference was found in the frequencies of various haplotypes between the two groups. CONCLUSION: Genotypes of the rs1202184 locus of the MDR1 gene are associated with refractory epilepsy in children, for which the AA genotype plays a dominant role.


Subject(s)
Drug Resistant Epilepsy/genetics , Polymorphism, Single Nucleotide , ATP Binding Cassette Transporter, Subfamily B/genetics , Case-Control Studies , Child , Gene Frequency , Genotype , Haplotypes , Humans
7.
Eur J Pharmacol ; 857: 172453, 2019 Aug 15.
Article in English | MEDLINE | ID: mdl-31202807

ABSTRACT

Aesculin, a natural product from the traditional and widely-used Chinese medicine named Cortex fraxini, has attracted attention as a novel therapeutic modulator of inflammation. However, little is known about its effect on ulcerative colitis (UC). This study aimed to investigate the protective effects and mechanisms of aesculin on colitis. The results showed that, few cytotoxicity of aesculin were shown in vivo and in the RAW264.7 macrophages, while aesculin significantly relieved the symptoms of DSS-induced colitis and restrained the expression of inflammatory factors including iNOS, IL-1ß, TNF-α in both peritoneal macrophages and colonic tissues from DSS-induced mice and RAW264.7 macrophages. Of note, aesculin attenuated the activity of NF-κB signaling while promoted the nuclear localization of PPAR-γ in both rectal tissues from DSS-induced mice and LPS-stimulated macrophages. These findings demonstrated that the protection of aesculin against ulcerative colitis might be due to its regulation on the PPAR-γ and NF-κB pathway. Thus, aesculin could serve as a potential therapeutic agent for the treatment of ulcerative colitis.


Subject(s)
Colitis/metabolism , Colitis/prevention & control , Dextran Sulfate/adverse effects , Esculin/pharmacology , NF-kappa B/metabolism , PPAR gamma/metabolism , Signal Transduction/drug effects , Animals , Colitis/chemically induced , Colitis/pathology , Cytokines/biosynthesis , Cytoprotection/drug effects , Macrophages/drug effects , Mice , RAW 264.7 Cells
8.
Front Immunol ; 9: 2272, 2018.
Article in English | MEDLINE | ID: mdl-30337925

ABSTRACT

Sepsis, defined as life-threatening organ dysfunction, is one of the most common causes of mortality in intensive care units with limited therapeutic options. However, the mechanism underlying the regulation of epigenetics on sepsis remains largely undefined. Here we showed that JMJD3, the histone lysine demethylase, played a critical role in the epigenetic regulation of innate immunity during early sepsis. Pharmacological inhibition of JMJD3 by GSKJ4 protected mice against early septic death and reduced pro-inflammatory cytokine interleukin-1ß (IL-1ß) production as well as IL-6, tumor necrosis factor-α (TNF-α), and monocyte chemotactic protein-1 (MCP-1) expression. Interestingly, GSKJ4 up-regulated the transcription of anti-inflammatory microRNA-146a (miR-146a) in peritoneal macrophages from septic mice. Mechanistically, JMJD3 negatively regulated the transcription of miR-146a via its demethylation of H3K27me3 on the promoter of miR-146a. Moreover, the transcription of miR-146a was positively regulated by nuclear factor-κB (NF-κB) p65. Inhibition of NF-κB p65 promoted JMJD3 binding to miR-146a promoter and decreased the tri-methylation level of H3K27, while the inhibition of JMJD3 did not affect the recruitment of NF-κB p65 to miR-146a promoter. These results highlight an epigenetic mechanism by which JMJD3 was inhibited by NF-κB p65 from binding to miR-146a promoter to promote the anti-inflammatory response. Taken together, our findings uncover a key role for JMJD3 in modulating the miR-146a transcription and shed light on the JMJD3 inhibitors could be potential therapeutic agents for early sepsis therapy.


Subject(s)
Benzazepines/pharmacology , Inflammation Mediators/immunology , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , MicroRNAs/immunology , Pyrimidines/pharmacology , Sepsis/prevention & control , Up-Regulation/drug effects , Animals , Female , Jumonji Domain-Containing Histone Demethylases/immunology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/pathology , Mice , Mice, Inbred ICR , Promoter Regions, Genetic/immunology , RAW 264.7 Cells , Sepsis/immunology , Sepsis/pathology , Transcription Factor RelA/immunology , Transcription, Genetic/immunology , Up-Regulation/immunology
9.
Acta Biochim Biophys Sin (Shanghai) ; 50(9): 888-896, 2018 Sep 01.
Article in English | MEDLINE | ID: mdl-30060081

ABSTRACT

Inflammasomes are protein complexes responsible for the release of IL-1 family cytokines, and they play critical roles in immunity and inflammation. The best-characterized inflammasome, the NOD-like receptor protein 3 (NLRP3) inflammasome, is involved in the development of multiple autoimmune diseases. However, the underlying mechanisms of abnormal NLRP3 inflammasome activation in systemic lupus erythematosus (SLE) remain elusive. Here, western blot analysis was used to detect the level of NLRP3 components and mTORC1/2 substrate in the kidney tissues from B6.MRL-FASlpr/J lupus mice and C57BL/6 mice, and the results showed that mammalian target of rapamycin (mTOR) complex 1/2 (mTORC1/2) and the NLRP3 inflammasome were hyperactivated in B6.MRL-FASlpr/J lupus mice. The inhibition of mTOR by INK128, a novel mTORC1/2 inhibitor, suppressed LPS/ATP and LPS/nigericin-induced NLRP3 inflammasome activation in bone marrow-derived macrophages (BMDMs) in vitro. INK128 decreased both the mRNA and protein levels of NLRP3 in an NF-κB-independent manner. Moreover, we reported for the first time that the inhibition of mTOR suppressed mitochondrial reactive oxygen species (ROS) production in BMDMs stimulated by an NLRP3 agonist. Furthermore, N-acetyl-L-cysteine, a ROS inhibitor, decreased NLRP3 expression, and rotenone, a robust ROS inducer, partially reversed the inhibitory effect of INK128 on NLRP3. These results demonstrated that mTOR regulated the activation of the NLRP3 inflammasome at least partially via ROS-induced NLRP3 expression. Importantly, in vivo data demonstrated that INK128 treatment prominently attenuated lupus nephritis and suppressed NLRP3 inflammasome activation in B6.MRL-FASlpr/J lupus mice. Taken together, our results suggest that activation of mTOR/ROS/NLRP3 signaling may contribute to the development of SLE.


Subject(s)
Inflammasomes/metabolism , Lupus Erythematosus, Systemic/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reactive Oxygen Species/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Benzoxazoles/pharmacology , Cells, Cultured , Female , Inflammasomes/drug effects , Lipopolysaccharides/pharmacology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/prevention & control , Lupus Nephritis/genetics , Lupus Nephritis/metabolism , Lupus Nephritis/prevention & control , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Pyrimidines/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors
11.
Carbohydr Polym ; 173: 360-371, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28732877

ABSTRACT

The polysaccharide SEP has been reported to activate NK and T cells via TLR2/4. Here, the combination of gemcitabine (GEM) and SEP against HepG-2 was investigated. SEP apparently enhanced antitumor activity of gemcitabine against liver cancer through stimulating NKG2D and DAP10/Akt pathway to activate NK cells. The NKG2D upregulation could improve the sensitivity of NK-92 cells targeting to its ligand MICA expressed on HepG-2 cells. Meanwhile, GEM up-regulated MICA expression and attenuated soluble MICA secretion through inhibiting ADAM10 expression, which in turn enhanced the cytotoxicity of NK-92 cells against cancer cells. SEP remarkably enhanced GEM antitumor activity with an inhibitory rate of 79.1% in an H22-bearing mouse model. Moreover, SEP reversed atrophy and apoptosis caused by GEM in both spleen and bone marrow through suppressing ROS secretion in vivo. The data indicated that the combination of SEP and GEM is a potential chemo-immunotherapy strategy for liver cancer treatment clinically.


Subject(s)
Cytotoxicity, Immunologic , Deoxycytidine/analogs & derivatives , Killer Cells, Natural/immunology , Liver Neoplasms, Experimental/drug therapy , Polysaccharides/pharmacology , Animals , Deoxycytidine/pharmacology , Hep G2 Cells , Histocompatibility Antigens Class I/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Mice, Nude , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Receptors, Immunologic/metabolism , Spleen/immunology , Strongylocentrotus/chemistry , Gemcitabine
12.
Acta Biochim Biophys Sin (Shanghai) ; 49(4): 328-337, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28338767

ABSTRACT

The disorder of B cells is one of the hallmarks of systemic lupus erythematosus (SLE). The activation state indicated by CD86 of B cells from SLE is well known, while the defect of regulatory B cells mediated by CD1d is also responsible for the process of SLE. In the present study, we focused on the relationship between B cell activation mediated by CD86 and B cell regulatory function mediated by CD1d. Our results showed that the level of CD1d in B cells was decreased during the early stages of B6.MRLlpr SLE mice and imiquimod-treated (IMQ-treated) mice, while the level of CD86 was significantly increased at the late stage. Moreover, the expression of CD1d showed a significantly negative correlation with CD86 level in B cells from IMQ-treated mice (r = -05741; P = 0.0022), B6.MRLlpr mice (r = -0.7091; P = 0.0268), and SLE patients (r = -0.4125; P = 0.0404). The in vivo and in vitro experiments with splenocytes demonstrated that CD1d signaling pathway could inhibit toll-like receptor 7 (TLR7)-induced CD86 expression of B cells. Further studies showed that this relationship also affected antibody production. Thus, our results confirmed the association of CD1d and CD86 levels in B cells from SLE, and demonstrated the importance to preserve the immunoregulatory function of B cells mediated by CD1d in the progression of SLE.


Subject(s)
Antigens, CD1d/immunology , B-Lymphocytes/immunology , B7-2 Antigen/immunology , Lupus Erythematosus, Systemic/immunology , Adolescent , Adult , Aminoquinolines/immunology , Animals , Antigens, CD1d/genetics , Antigens, CD1d/metabolism , B-Lymphocytes/metabolism , B7-2 Antigen/genetics , B7-2 Antigen/metabolism , Blotting, Western , Female , Flow Cytometry , Humans , Imiquimod , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Toll-Like Receptor 7/immunology , Toll-Like Receptor 7/metabolism , Young Adult
13.
Immunol Lett ; 181: 71-78, 2017 01.
Article in English | MEDLINE | ID: mdl-27923569

ABSTRACT

Systemic lupus erythematosus (SLE) possesses a gender-dependent incidence characterized by a male/female ratio 1:9. B-cell, a vital part of the immune system, plays an important role in pathogenesis of SLE. Thus, we hypothesize that gender differences of B cells may exist in SLE and relate to the onset and the progression of SLE. Here, we showed that the genes expression pattern is similar between healthy female and male. However, SLE female and SLE male showed more upregulated genes, in which the trendline of SLE male is higher than that of SLE female. The most differentially expressed genes between SLE male patients and female patients are only on two chromosomes. While the differentially expressed genes between healthy male and female are distributed on several chromosomes. There are more differentially expressed genes in SLE male vs healthy male than these in SLE female vs healthy female. OAS3, RGS13, STAG3, IFI44L, STS-1, FERIL14, ZBTB16, USP18, USP41, RSAD2, FKBP5, IL1R2, DNAPTP6 and ILI27, which top 14 significantly upregulated mRNAs in SLE patients compared with healthy donors, showed different expression pattern in gender-based analyses. Furthermore, we revealed that this difference may be related to estrogen-induced IFI44L/BAFF. Therefore, we conclude that the diagnosis and treatment of these immune-related diseases should consider the baseline gender-related differences.


Subject(s)
Antigens/metabolism , B-Cell Activating Factor/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cytoskeletal Proteins/metabolism , Estrogens/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Animals , Antigens/genetics , Apoptosis/drug effects , Apoptosis/genetics , B-Cell Activating Factor/genetics , Case-Control Studies , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Computational Biology/methods , Cytoskeletal Proteins/genetics , Disease Models, Animal , Estrogens/pharmacology , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Interferon-alpha/metabolism , Interferon-alpha/pharmacology , Lupus Erythematosus, Systemic/genetics , Male , Mice , Mice, Transgenic , Sex Factors , Signal Transduction , Transcriptome
14.
Cell Death Dis ; 7(8): e2341, 2016 08 18.
Article in English | MEDLINE | ID: mdl-27537524

ABSTRACT

The increased death of macrophages has been considered as a pathogenic factor for systemic lupus erythematosus (SLE), and dysfunction of autophagy may contribute to improper cell death. However, the effect of autophagy on macrophage during the pathogenesis of SLE is still unclear. Here we found that the death rate and autophagy level of macrophages significantly increased in MRL/lpr lupus-prone mice. Activation of toll-like receptor 7 (TLR7) triggered macrophage death in an autophagy-dependent but caspase-independent way in vitro. Moreover, P62/SQSTM1 is thought to have an essential role in selective autophagy. We also demonstrated that P62/SQSTM1 was required for TLR7-induced autophagy, and knockdown of P62 suppressed R848-induced cell death and LC3II protein accumulation. As an important mediator for cell-cell communication, Notch signaling is responsible for cell-fate decisions. Our results showed that activation of TLR7 also upregulated the expression of Notch1, especially its downstream target gene Hairy and enhancer of split 1 (Hes-1) in macrophages. Of note, we found that Hes-1, as a transcriptional factor, controlled TLR7-induced autophagy by regulating P62 expression. Furthermore, to confirm the above results in vivo, TLR7 agonist imiquimod (IMQ)-induced lupus mouse model was prepared. Splenic macrophages from IMQ-treated mice exhibited increased autophagy and cell death as well as enhanced expressions of Notch1 and Hes-1. Our results indicate that Notch1-Hes-1 signaling controls TLR7-induced autophagic death of macrophage via regulation of P62 in mice with lupus.


Subject(s)
Autophagy , Lupus Erythematosus, Systemic/metabolism , Macrophages/pathology , Receptors, Notch/metabolism , Sequestosome-1 Protein/metabolism , Signal Transduction , Toll-Like Receptor 7/metabolism , Transcription Factor HES-1/metabolism , Animals , Disease Models, Animal , Female , Macrophages/metabolism , Macrophages/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , RAW 264.7 Cells , Up-Regulation
15.
Cell Mol Immunol ; 13(6): 764-775, 2016 11.
Article in English | MEDLINE | ID: mdl-26144250

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by B-cell hyperreactivity. The Toll-like receptor 7 (TLR7) signaling pathway is abnormally activated in SLE B cells. CyclinD3 (CCND3) plays an important role in B-cell proliferation, development, and differentiation. Although previous studies focused on the B cell-intrinsic role of TLR7 for the development of spontaneous germinal centers, the influence of TLR7 on CCND3 in SLE B cells is still not clear. Here, we used a B-cell profiling chip and found that CCND3 was related to SLE and significantly elevated in SLE B cells. Moreover, we determined that the expression level of CCND3 was higher, while miR-15b was significantly lower in the B cells from SLE patients and B6.MRL-Faslpr/J lupus mice compared to normal subjects. Furthermore, we demonstrated that the activation of TLR7 dramatically increased CCND3 expression but significantly decreased miR-15b in B cells in vitro and we identified that CCND3 is a direct target of miR-15b. To further confirm our results, we established another lupus model by topically treating C57BL/6 (B6) mice with the TLR-7 agonist imiquimod (IMQ) for 8 weeks according to the previously described protocol. Expectedly, topical treatment with IMQ also significantly increased CCND3 and decreased miR-15b in B cells of B6 mice. Taken together, our results identified that the activation of TLR7 increased CCND3 expression via the downregulation of miR-15b in B cells; thus, these findings suggest that extrinsic factor-induced CCND3 expression may contribute to the abnormality of B cell in SLE.


Subject(s)
B-Lymphocytes/metabolism , Cyclin D3/genetics , Down-Regulation/genetics , Lupus Erythematosus, Systemic/genetics , MicroRNAs/genetics , Toll-Like Receptor 7/metabolism , Adult , Aminoquinolines , Animals , Antigens, CD19/metabolism , Cluster Analysis , Cyclin D3/metabolism , Disease Models, Animal , Female , Gene Expression Profiling , Gene Ontology , Gene Regulatory Networks , Humans , Imiquimod , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Mice, Inbred C57BL , MicroRNAs/metabolism , Protein Interaction Maps , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation/genetics
16.
Immunol Lett ; 168(2): 355-65, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26545567

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease with prominent chronic inflammatory aspects. Plasmacytoid dendritic cells (pDCs), which are the principal interferon-α (IFN-α)-producing cells, have known to be critically involved in SLE pathogenesis. Our previous research demonstrated that a benzenediamine derivative FC-99 possessed anti-inflammatory activities. However, the effects of FC-99 on SLE have not been investigated to date. In this study, we found that FC-99 attenuated lupus-like pathological symptoms and lupus nephritis as well as the expression of pro-inflammatory cytokines in kidneys of MRL/lpr mice. FC-99 also decreased both the total IgM, total IgG and anti-dsDNA IgG levels in sera and the activation of B cells in the PBMCs and spleens of MRL/lpr mice. Moreover, FC-99 inhibited the abnormal activation and number of pDCs from PBMCs and spleens and levels of IFN-α in MRL/lpr mice. Notably, FC-99 significantly suppressed the expression of IFN-inducible genes in peripheral blood mononuclear cells (PBMCs) and spleens from MRL/lpr mice. As expected, in vitro experiments demonstrated that FC-99 decreased both the activation and IFN-α production of pDCs and inhibited IRAK4 phosphorylation in pDCs upon TLR7 and TLR9 stimulation. We further confirm that the inhibition of FC-99 on B cell activation depended on level of pDCs-secreting IFN-α. These data indicate that FC-99 attenuated lupus-like syndrome in MRL/lpr mice related to suppression of pDC activation, especially pDCs-secreting IFN-α. This study suggests that FC-99 may be a potential therapeutic candidate for the treatment of SLE.


Subject(s)
Alkanesulfonates/pharmacology , Dendritic Cells/drug effects , Fluorocarbons/pharmacology , Lupus Erythematosus, Systemic/prevention & control , Alkanesulfonates/chemistry , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Flow Cytometry , Fluorocarbons/chemistry , Gene Expression/drug effects , Gene Expression/immunology , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunoglobulin M/immunology , Immunoglobulin M/metabolism , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Kidney/drug effects , Kidney/immunology , Kidney/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Molecular Structure , Reverse Transcriptase Polymerase Chain Reaction , Spleen/drug effects , Spleen/immunology , Spleen/metabolism , Syndrome
17.
Acta Biochim Biophys Sin (Shanghai) ; 47(8): 620-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26071573

ABSTRACT

Estrogens are strongly implicated in gender differences in immune responses by influencing the development and activation of immune cells. Recent studies have shown that myeloid-derived suppressor cells (MDSCs), derived from CD11b(+)Gr-1(+) myeloid cells under pathological conditions, play vital roles in modulating immune responses. However, it is still unknown the effects of estrogens on MDSCs. In the present study, we investigated the effects and mechanisms of estrogens on regulating the accumulation of MDSCs. It was found that, compared with male patients with systemic lupus erythematosus (SLE), female patients with SLE showed a higher frequency of MDSCs in peripheral blood mononuclear cells and a higher level of tumor necrosis factor α (TNF-α) in serum. Notably, estradiol level in the serum of female patients with SLE was positively correlated with the frequency of MDSCs. Moreover, 17ß-estradiol could promote TNF-α-induced accumulation of MDSCs in vivo by increasing the fundamental frequency of CD11b(+)Gr-1(+) cells. Furthermore, 17ß-estradiol promoted the secretion of TNF-α in vivo, which contributed to the increase of the frequency of CD11b(+)Gr-1(+) cells. In addition, it was also found that female mice showed a higher frequency of CD11b(+)Gr-1(+) cells and a higher TNF-α level in blood than the age-matched male mice. These data indicate that 17ß-estradiol contributes to the accumulation of MDSCs in blood by promoting TNF-α secretion, which increases the fundamental frequency of CD11b(+)Gr-1(+) cells. Our findings provide a new insight into the mechanism of gender difference in the prevalence of inflammation and autoimmune diseases.


Subject(s)
Estradiol/metabolism , Lupus Erythematosus, Systemic/blood , Myeloid Cells/metabolism , Tumor Necrosis Factor-alpha/blood , Animals , Female , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/physiopathology , Male , Mice , Myeloid Cells/pathology , Sex Factors , Suppressor Factors, Immunologic/blood , Tumor Necrosis Factor-alpha/metabolism
18.
Mol Cells ; 38(5): 441-51, 2015 May.
Article in English | MEDLINE | ID: mdl-25947293

ABSTRACT

Recognition of cytosolic DNA initiates a series of innate immune responses by inducing IFN-I production and subsequent triggering JAK1-STAT1 signaling which plays critical roles in the pathogenesis of infection, inflammation and autoimmune diseases through promoting B cell activation and antibody responses. The stimulator of interferon genes protein (STING) has been demonstrated to be a critical hub of type I IFN induction in cytosolic DNA-sensing pathways. However, it still remains unknown whether cytosolic DNA can directly activate the JAK1-STAT1 signaling or not. And the role of STING is also unclear in this response. In the present study, we found that dsDNA directly triggered the JAK1-STAT1 signaling by inducing phosphorylation of the Lyn kinase. Moreover, this response is not dependent on type I IFN receptors. Interestingly, STING could inhibit dsDNA-triggered activation of JAK1-STAT1 signaling by inducing SHP-1 and SHP-2 phosphorylation. In addition, compared with normal B cells, the expression of STING was significantly lower and the phosphorylation level of JAK1 was significantly higher in B cells from MRL/lpr lupus-prone mice, highlighting the close association between STING low-expression and JAK1-STAT1 signaling activation in B cells in autoimmune diseases. Our data provide a molecular insight into the novel role of STING in dsDNA-mediated inflammatory disorders.


Subject(s)
B-Lymphocytes/metabolism , DNA/metabolism , Lupus Erythematosus, Systemic/immunology , Membrane Proteins/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Adult , Animals , Female , HEK293 Cells , HeLa Cells , Humans , Janus Kinase 1/metabolism , Jurkat Cells , Male , Mice , Phosphorylation , STAT1 Transcription Factor/metabolism , Signal Transduction , Spleen/immunology
19.
Eur J Immunol ; 45(8): 2377-88, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25959715

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the overexpression of IFN-α. IFN-α induces autophagy via the JAK1-STAT1 signaling pathway, contributing to the pathogenesis of SLE. Recent studies reported that B cells from patients with SLE and NZB/W F1 mice had enhanced autophagy activity; however, the mechanism still remains unknown. Here, we show that the protein tyrosine phosphatase STS-1 (suppressor of T-cell receptor signaling 1) was significantly overexpressed in B cells from patients with SLE and MRL/lpr mice. Notably, STS-1 promoted IFN-α-induced autophagy in B cells by enhancing the JAK1-STAT1 signaling activation. STS-1 inhibited the phosphorylation of the E3 ubiquitin protein ligase c-cbl, and subsequently promoted IFN-α-induced phosphorylation of tyrosine kinase 2, leading to JAK1-STAT1 signaling activation. Furthermore, STAT1 and JAK1 inhibitors blocked the IFN-α-induced autophagy promoted by STS-1, indicating that STS-1 promotes IFN-α-induced autophagy via the JAK1-STAT1 signaling. Our results demonstrate the importance of STS-1 in regulating IFN-α-induced autophagy in B cells, and this could be used as a therapeutic approach to treat SLE.


Subject(s)
Autophagy/immunology , B-Lymphocytes/immunology , Interferon-alpha/immunology , Janus Kinase 1/immunology , Protein Tyrosine Phosphatases/immunology , Receptors, Antigen, T-Cell/immunology , STAT1 Transcription Factor/immunology , Signal Transduction/immunology , Animals , Female , Humans , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/therapy , Male , Mice , Proto-Oncogene Proteins c-cbl/immunology , TYK2 Kinase/immunology
20.
Eur J Immunol ; 45(7): 1934-45, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25929465

ABSTRACT

B cells present lipid antigens to CD1d-restricted invariant natural killer T (iNKT) cells to maintain autoimmune tolerance, and this process is disrupted in systemic lupus erythematosus (SLE). Inflammation may inhibit CD1d expression to exacerbate the pathology of lupus. However, how inflammation regulates CD1d expression on B cells is unclear in SLE. In the present study, we showed that the surface expression of CD1d on B cells from SLE mice was decreased and that stimulation of inflammatory responses through TLR9 decreased the membrane and total CD1d levels of CD1d on B cells. Moreover, inflammation-related microRNA-155 (miR-155) negatively correlated with the expression of CD1d in B cells. miR-155 directly targeted the 3'-untranslated region (3'-UTR) of CD1d upon TLR9 activation in both humans and mice. The inhibitory effects of miR-155 on CD1d expression in B cells impaired their antigen-presenting capacity to iNKT cells. In addition, Ets-1, a susceptibility gene of SLE, also directly regulated the expression of the CD1d gene at the transcriptional level. These findings provide new insight into the mechanism underlying decreased CD1d expression on B cells in SLE, suggesting that inhibition of inflammation may increase CD1d expression in B cells to ameliorate SLE via modulating iNKT cells.


Subject(s)
Antigens, CD1d/biosynthesis , B-Lymphocytes/immunology , Gene Expression Regulation/immunology , Lupus Erythematosus, Systemic/immunology , MicroRNAs/immunology , Proto-Oncogene Protein c-ets-1/immunology , Animals , Antigen Presentation/immunology , Antigens, CD1d/immunology , Blotting, Western , Coculture Techniques , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Inflammation/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Natural Killer T-Cells/immunology , Real-Time Polymerase Chain Reaction , Toll-Like Receptor 9/immunology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...