Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 466
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 184(11): 2896-2910.e13, 2021 05 27.
Article in English | MEDLINE | ID: mdl-34048705

ABSTRACT

Damaged mitochondria need to be cleared to maintain the quality of the mitochondrial pool. Here, we report mitocytosis, a migrasome-mediated mitochondrial quality-control process. We found that, upon exposure to mild mitochondrial stresses, damaged mitochondria are transported into migrasomes and subsequently disposed of from migrating cells. Mechanistically, mitocytosis requires positioning of damaged mitochondria at the cell periphery, which occurs because damaged mitochondria avoid binding to inward motor proteins. Functionally, mitocytosis plays an important role in maintaining mitochondrial quality. Enhanced mitocytosis protects cells from mitochondrial stressor-induced loss of mitochondrial membrane potential (MMP) and mitochondrial respiration; conversely, blocking mitocytosis causes loss of MMP and mitochondrial respiration under normal conditions. Physiologically, we demonstrate that mitocytosis is required for maintaining MMP and viability in neutrophils in vivo. We propose that mitocytosis is an important mitochondrial quality-control process in migrating cells, which couples mitochondrial homeostasis with cell migration.


Subject(s)
Membrane Potential, Mitochondrial/physiology , Mitochondria/metabolism , Animals , Biological Transport , Cell Line , Cell Movement/physiology , Cytoplasm/metabolism , Exocytosis/physiology , Female , Homeostasis , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission/methods , Mitochondria/physiology , Mitochondrial Membranes/metabolism , Organelles/metabolism
2.
Immunity ; 57(5): 1056-1070.e5, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38614091

ABSTRACT

A specialized population of mast cells residing within epithelial layers, currently known as intraepithelial mast cells (IEMCs), was originally observed over a century ago, yet their physiological functions have remained enigmatic. In this study, we unveil an unexpected and crucial role of IEMCs in driving gasdermin C-mediated type 2 immunity. During helminth infection, αEß7 integrin-positive IEMCs engaged in extensive intercellular crosstalk with neighboring intestinal epithelial cells (IECs). Through the action of IEMC-derived proteases, gasdermin C proteins intrinsic to the epithelial cells underwent cleavage, leading to the release of a critical type 2 cytokine, interleukin-33 (IL-33). Notably, mast cell deficiency abolished the gasdermin C-mediated immune cascade initiated by epithelium. These findings shed light on the functions of IEMCs, uncover a previously unrecognized phase of type 2 immunity involving mast cell-epithelial cell crosstalk, and advance our understanding of the cellular mechanisms underlying gasdermin C activation.


Subject(s)
Interleukin-33 , Mast Cells , Phosphate-Binding Proteins , Mast Cells/immunology , Mast Cells/metabolism , Animals , Interleukin-33/metabolism , Interleukin-33/immunology , Mice , Phosphate-Binding Proteins/metabolism , Epithelial Cells/immunology , Epithelial Cells/metabolism , Mice, Inbred C57BL , Mice, Knockout , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/immunology , Cell Communication/immunology
3.
Cell ; 175(4): 1059-1073.e21, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30270039

ABSTRACT

Motivated by the clinical observation that interruption of the mevalonate pathway stimulates immune responses, we hypothesized that this pathway may function as a druggable target for vaccine adjuvant discovery. We found that lipophilic statin drugs and rationally designed bisphosphonates that target three distinct enzymes in the mevalonate pathway have potent adjuvant activities in mice and cynomolgus monkeys. These inhibitors function independently of conventional "danger sensing." Instead, they inhibit the geranylgeranylation of small GTPases, including Rab5 in antigen-presenting cells, resulting in arrested endosomal maturation, prolonged antigen retention, enhanced antigen presentation, and T cell activation. Additionally, inhibiting the mevalonate pathway enhances antigen-specific anti-tumor immunity, inducing both Th1 and cytolytic T cell responses. As demonstrated in multiple mouse cancer models, the mevalonate pathway inhibitors are robust for cancer vaccinations and synergize with anti-PD-1 antibodies. Our research thus defines the mevalonate pathway as a druggable target for vaccine adjuvants and cancer immunotherapies.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cancer Vaccines/immunology , Diphosphonates/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mevalonic Acid/metabolism , rab5 GTP-Binding Proteins/antagonists & inhibitors , Animals , Antigen Presentation , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Cell Line, Tumor , Endosomes/drug effects , Female , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Protein Prenylation , rab5 GTP-Binding Proteins/metabolism
4.
Nature ; 620(7976): 1047-1053, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37459895

ABSTRACT

Zygotic genome activation (ZGA) activates the quiescent genome to enable the maternal-to-zygotic transition1,2. However, the identity of transcription factors that underlie mammalian ZGA in vivo remains elusive. Here we show that OBOX, a PRD-like homeobox domain transcription factor family (OBOX1-OBOX8)3-5, are key regulators of mouse ZGA. Mice deficient for maternally transcribed Obox1/2/5/7 and zygotically expressed Obox3/4 had a two-cell to four-cell arrest, accompanied by impaired ZGA. The Obox knockout defects could be rescued by restoring either maternal and zygotic OBOX, which suggests that maternal and zygotic OBOX redundantly support embryonic development. Chromatin-binding analysis showed that Obox knockout preferentially affected OBOX-binding targets. Mechanistically, OBOX facilitated the 'preconfiguration' of RNA polymerase II, as the polymerase relocated from the initial one-cell binding targets to ZGA gene promoters and distal enhancers. Impaired polymerase II preconfiguration in Obox mutants was accompanied by defective ZGA and chromatin accessibility transition, as well as aberrant activation of one-cell polymerase II targets. Finally, ectopic expression of OBOX activated ZGA genes and MERVL repeats in mouse embryonic stem cells. These data thus demonstrate that OBOX regulates mouse ZGA and early embryogenesis.


Subject(s)
Embryonic Development , Gene Expression Regulation, Developmental , Genome , Homeodomain Proteins , Transcription Factors , Zygote , Animals , Mice , Chromatin/genetics , Chromatin/metabolism , Embryonic Development/genetics , Enhancer Elements, Genetic/genetics , Genome/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mouse Embryonic Stem Cells/metabolism , Mutation , Promoter Regions, Genetic/genetics , RNA Polymerase II/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism , Zygote/metabolism
5.
Nat Immunol ; 17(8): 930-7, 2016 08.
Article in English | MEDLINE | ID: mdl-27322654

ABSTRACT

Most of the known regulatory mechanisms that curb inflammatory gene expression target pre-transcription-initiation steps, and evidence for post-initiation regulation of inflammatory gene expression remains scarce. We found that the transcriptional repressor Hes1 suppressed production of CXCL1, a chemokine that is crucial for recruiting neutrophils. Hes1 negatively regulated neutrophil recruitment in vivo in a manner that was dependent on macrophage-produced CXCL1, and it attenuated the severity of inflammatory arthritis. Mechanistically, inhibition of Cxcl1 expression by Hes1 did not involve modification of transcription initiation. Instead, Hes1 inhibited signal-induced recruitment of the positive transcription-elongation complex P-TEFb and thereby prevented phosphorylation of RNA polymerase II at Ser2 and productive elongation. Thus, our results identify Hes1 as a homeostatic suppressor of inflammatory responses that exerts its suppressive function by regulating transcription elongation.


Subject(s)
Arthritis/genetics , Cell Cycle Proteins/metabolism , Inflammation/genetics , Macrophages/immunology , Transcription Factor HES-1/metabolism , Animals , Cell Cycle Proteins/genetics , Cells, Cultured , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Gene Expression Regulation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Neutrophil Infiltration/genetics , Positive Transcriptional Elongation Factor B/genetics , Positive Transcriptional Elongation Factor B/metabolism , RNA Polymerase II/metabolism , Transcription Elongation, Genetic , Transcription Factor HES-1/genetics
6.
Immunity ; 51(2): 201-203, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31433964

ABSTRACT

Formation of memory T cells is coupled with changes of metabolic status, yet how environmental metabolites affect the transition remains largely unknown. In this issue of Immunity, Bachem et al. (2019) report that microbiota-derived butyrate enhances the memory potential of CD8+ T cells via rewiring cellular metabolism.


Subject(s)
Butyrates , Microbiota , CD8-Positive T-Lymphocytes
7.
Immunity ; 51(2): 272-284.e7, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31399282

ABSTRACT

Macrophage polarization is accompanied by drastic changes in L-arginine metabolism. Two L-arginine catalytic enzymes, iNOS and arginase 1, are well-characterized hallmark molecules of classically and alternatively activated macrophages, respectively. The third metabolic fate of L-arginine is the generation of creatine that acts as a key source of cellular energy reserve, yet little is known about the role of creatine in the immune system. Here, genetic, genomic, metabolic, and immunological analyses revealed that creatine reprogrammed macrophage polarization by suppressing M(interferon-γ [IFN-γ]) yet promoting M(interleukin-4 [IL-4]) effector functions. Mechanistically, creatine inhibited the induction of immune effector molecules, including iNOS, by suppressing IFN-γ-JAK-STAT1 transcription-factor signaling while supporting IL-4-STAT6-activated arginase 1 expression by promoting chromatin remodeling. Depletion of intracellular creatine by ablation of the creatine transporter Slc6a8 altered macrophage-mediated immune responses in vivo. These results uncover a previously uncharacterized role for creatine in macrophage polarization by modulating cellular responses to cytokines such as IFN-γ and IL-4.


Subject(s)
Arginine/metabolism , Creatine/metabolism , Liver Cirrhosis/metabolism , Macrophages/physiology , Membrane Transport Proteins/metabolism , Animals , Cell Differentiation , Cells, Cultured , Cellular Reprogramming , Humans , Immunity, Cellular , Interferon-gamma/metabolism , Liver Cirrhosis/chemically induced , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Signal Transduction , Tetrachloroethylene
9.
Nat Immunol ; 16(8): 838-849, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26147685

ABSTRACT

Interferon-γ (IFN-γ) primes macrophages for enhanced microbial killing and inflammatory activation by Toll-like receptors (TLRs), but little is known about the regulation of cell metabolism or mRNA translation during this priming. We found that IFN-γ regulated the metabolism and mRNA translation of human macrophages by targeting the kinases mTORC1 and MNK, both of which converge on the selective regulator of translation initiation eIF4E. Physiological downregulation of mTORC1 by IFN-γ was associated with autophagy and translational suppression of repressors of inflammation such as HES1. Genome-wide ribosome profiling in TLR2-stimulated macrophages showed that IFN-γ selectively modulated the macrophage translatome to promote inflammation, further reprogram metabolic pathways and modulate protein synthesis. These results show that IFN-γ-mediated metabolic reprogramming and translational regulation are key components of classical inflammatory macrophage activation.


Subject(s)
Interferon-gamma/immunology , Macrophage Activation/immunology , Macrophages/immunology , Protein Biosynthesis/immunology , RNA, Messenger/immunology , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Cells, Cultured , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/immunology , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Profiling , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Homeodomain Proteins/metabolism , Humans , Interferon-gamma/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Macrophage Activation/drug effects , Macrophage Activation/genetics , Macrophages/drug effects , Macrophages/metabolism , Mechanistic Target of Rapamycin Complex 1 , MicroRNAs/genetics , Microscopy, Fluorescence , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , Multiprotein Complexes/metabolism , Protein Biosynthesis/drug effects , Protein Biosynthesis/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/metabolism , Transcription Factor HES-1
10.
Plant Cell ; 35(7): 2570-2591, 2023 06 26.
Article in English | MEDLINE | ID: mdl-37040621

ABSTRACT

SALT OVERLY SENSITIVE1 (SOS1) is a key component of plant salt tolerance. However, how SOS1 transcription is dynamically regulated in plant response to different salinity conditions remains elusive. Here, we report that C-type Cyclin1;1 (CycC1;1) negatively regulates salt tolerance by interfering with WRKY75-mediated transcriptional activation of SOS1 in Arabidopsis (Arabidopsis thaliana). Disruption of CycC1;1 promotes SOS1 expression and salt tolerance in Arabidopsis because CycC1;1 interferes with RNA polymerase II recruitment by occupying the SOS1 promoter. Enhanced salt tolerance of the cycc1;1 mutant was completely compromised by an SOS1 mutation. Moreover, CycC1;1 physically interacts with the transcription factor WRKY75, which can bind to the SOS1 promoter and activate SOS1 expression. In contrast to the cycc1;1 mutant, the wrky75 mutant has attenuated SOS1 expression and salt tolerance, whereas overexpression of SOS1 rescues the salt sensitivity of wrky75. Intriguingly, CycC1;1 inhibits WRKY75-mediated transcriptional activation of SOS1 via their interaction. Thus, increased SOS1 expression and salt tolerance in cycc1;1 were abolished by WRKY75 mutation. Our findings demonstrate that CycC1;1 forms a complex with WRKY75 to inactivate SOS1 transcription under low salinity conditions. By contrast, under high salinity conditions, SOS1 transcription and plant salt tolerance are activated at least partially by increased WRKY75 expression but decreased CycC1;1 expression.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis/metabolism , Salt Tolerance/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Gene Expression Regulation, Plant/genetics , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism
11.
Trends Immunol ; 44(2): 93-100, 2023 02.
Article in English | MEDLINE | ID: mdl-36586780

ABSTRACT

Cytokine release syndrome (CRS) is a severe clinical syndrome marked by drastic elevation of inflammatory cytokines such as interleukin (IL)-1, IL-6, and tumor necrosis factor (TNF). Despite the current empirical therapeutic strategies, prediction of CRS onset and identification of high-risk individuals are not satisfactory due to poor understanding of the mechanisms underlying CRS-related immune dysfunction and risk factors for CRS. Recent studies have suggested that conditions such as stress, obesity, diabetes, and hypertension may contribute to the development of CRS. Here, we discuss potential connections between these conditions and CRS pathogenesis, with a focus on stress hormone catecholamine-mediated effects, hoping that the design of CRS therapeutic approaches ensues from a renewed perspective.


Subject(s)
Catecholamines , Cytokine Release Syndrome , Humans , Catecholamines/therapeutic use , Cytokines , Risk Factors
12.
PLoS Pathog ; 19(1): e1011108, 2023 01.
Article in English | MEDLINE | ID: mdl-36656870

ABSTRACT

Mounting evidence suggests that the gut microbiota plays an important role in the pathogenesis of mastitis, an important disease affecting the health of lactating women and the development of the dairy industry. However, the effect of the regulation of the gut microbiota by dietary components on mastitis development remains unknown. In this study, we found that a fiber-enriched diet alleviated Staphylococcus aureus (S. au)-induced mastitis in mice, which was dependent on the gut microbiota as depletion of the gut microbiota by antibiotics abolished this protective effect. Likewise, fecal microbiota transplantation (FMT) from high-inulin (HI)-treated mice (HIF) to recipient mice improved S. au-induced mastitis in mice. Consumption of an HI diet and HIF increased fecal short-chain fatty acid (SCFA) levels compared with the control group. Moreover, treatment with SCFAs, especially butyrate, alleviated S. au-induced mastitis in mice. Mechanistically, consumption of an HI diet enhanced the host antimicrobial program in macrophages through inhibiting histone deacetylase 3 by the production of butyrate. Collectively, our results suggest that modulation of the gut microbiota and its metabolism by dietary components is a potential strategy for mastitis intervention and serve as a basis for other infectious diseases.


Subject(s)
Butyrates , Mastitis , Animals , Female , Mice , Anti-Bacterial Agents/pharmacology , Diet , Lactation , Macrophages , Mastitis/therapy , Staphylococcus aureus , Dietary Fiber
13.
PLoS Pathog ; 19(11): e1011764, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37948460

ABSTRACT

Subacute ruminal acidosis (SARA) has been demonstrated to promote the development of mastitis, one of the most serious diseases in dairy farming worldwide, but the underlying mechanism is unclear. Using untargeted metabolomics, we found hexadecanamide (HEX) was significantly reduced in rumen fluid and milk from cows with SARA-associated mastitis. Herein, we aimed to assess the protective role of HEX in Staphylococcus aureus (S. aureus)- and SARA-induced mastitis and the underlying mechanism. We showed that HEX ameliorated S. aureus-induced mastitis in mice, which was related to the suppression of mammary inflammatory responses and repair of the blood-milk barrier. In vitro, HEX depressed S. aureus-induced activation of the NF-κB pathway and improved barrier integrity in mouse mammary epithelial cells (MMECs). In detail, HEX activated PPARα, which upregulated SIRT1 and subsequently inhibited NF-κB activation and inflammatory responses. In addition, ruminal microbiota transplantation from SARA cows (S-RMT) caused mastitis and aggravated S. aureus-induced mastitis, while these changes were reversed by HEX. Our findings indicate that HEX effectively attenuates S. aureus- and SARA-induced mastitis by limiting inflammation and repairing barrier integrity, ultimately highlighting the important role of host or microbiota metabolism in the pathogenesis of mastitis and providing a potential strategy for mastitis prevention.


Subject(s)
Mastitis , Staphylococcus aureus , Humans , Female , Animals , Mice , Cattle , Staphylococcus aureus/metabolism , NF-kappa B/metabolism , Milk , Mastitis/metabolism
14.
Hepatology ; 79(2): 425-437, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-37611260

ABSTRACT

BACKGROUND AND AIMS: The predominantly progressive, indeterminate, and predominantly regressive (P-I-R) classification extends beyond staging and provides information on dynamic changes of liver fibrosis. However, the prognostic implication of P-I-R classification is not elucidated. Therefore, in the present research, we investigated the utility of P-I-R classification in predicting the on-treatment clinical outcomes. APPROACH AND RESULTS: In an extension study on a randomized controlled trial, we originally enrolled 1000 patients with chronic hepatitis B and biopsy-proven histological significant fibrosis, and treated them for more than 7 years with entecavir-based therapy. Among the 727 patients with a second biopsy at treatment week 72, we compared P-I-R classification and Ishak score changes in 646 patients with adequate liver sections for the histological evaluation. Progressive, indeterminate, and regressive cases were observed in 70%, 17%, and 13% of patients before treatments and 20%, 14%, and 64% after 72-week treatment, respectively, which could further differentiate the histological outcomes of patients with stable Ishak scores. The 7-year cumulative incidence of HCC was 1.5% for the regressive cases, 4.3% for the indeterminate cases, and 22.8% for the progressive cases ( p <0.001). After adjusting for age, treatment regimen, platelet counts, cirrhosis, Ishak fibrosis score changes, and Laennec staging, the posttreatment progressive had a HR of 17.77 (vs. posttreatment regressive; 95% CI: 5.55-56.88) for the incidence of liver-related events (decompensation, HCC, and death/liver transplantation). CONCLUSIONS: The P-I-R classification can be a meaningful complement to the Ishak fibrosis score not only in evaluating the histological changes but also in predicting the clinical outcomes.


Subject(s)
Carcinoma, Hepatocellular , Hepatitis B, Chronic , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/pathology , Antiviral Agents/therapeutic use , Liver Neoplasms/pathology , Liver Cirrhosis/pathology , Liver/pathology , Hepatitis B, Chronic/complications , Hepatitis B, Chronic/drug therapy , Hepatitis B, Chronic/pathology , Fibrosis , Biopsy/adverse effects
15.
FASEB J ; 38(2): e23383, 2024 01 31.
Article in English | MEDLINE | ID: mdl-38197892

ABSTRACT

Mastitis is the most frequent disease of cows and has well-recognized detrimental effects on animal wellbeing and dairy farm profitability. With the advent of the postantibiotic era, alternative antibiotic agents, especially probiotics, have received increasing attention in the treatment of mastitis. Based on research showing that Lactobacillus reuteri (L. reuteri) has anti-inflammatory effects, this study explored the protective effects and mechanisms of L. reuteri against mastitis induced by Staphylococcus aureus (S. aureus) in mice. First, mice with S. aureus-induced mastitis were orally administered L. reuteri, and the inflammatory response in the mammary gland was observed. The results showed that L. reuteri significantly inhibited S. aureus-induced mastitis. Moreover, the concentration of oxytocin (OT) and protein expression of oxytocin receptor (OTR) were measured, and inhibition of OTR or vagotomy reversed the protective effect of L. reuteri or its culture supernatant (LCS) on S. aureus-induced mastitis. In addition, in mouse mammary epithelial cells (MMECs), OT inhibited the inflammation induced by S. aureus by inhibiting the protein expression of OTR. It was suggested that L. reuteri protected against S. aureus-induced mastitis by releasing OT. Furthermore, microbiological analysis showed that the composition of the microbiota was altered, and the relative abundance of Lactobacillus was significantly increased in gut and mammary gland after treatment with L. reuteri or LCS. In conclusion, our study found the L. reuteri inhibited the mastitis-induced by S. aureus via promoting the release of OT, and treatment with L. reuteri increased the abundance of Lactobacillus in both gut and mammary gland.


Subject(s)
Gastrointestinal Microbiome , Limosilactobacillus reuteri , Mastitis , Staphylococcal Infections , Female , Humans , Animals , Cattle , Mice , Oxytocin/pharmacology , Oxytocin/therapeutic use , Staphylococcus aureus , Mastitis/therapy , Receptors, Oxytocin , Lactobacillus
16.
J Immunol ; 211(9): 1340-1347, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37756541

ABSTRACT

TNF plays a crucial role in inflammation and bone resorption in various inflammatory diseases, including rheumatoid arthritis (RA). However, its direct ability to drive macrophages to differentiate into osteoclasts is limited. Although RBP-J is recognized as a key inhibitor of TNF-mediated osteoclastogenesis, the precise mechanisms that restrain TNF-induced differentiation of macrophages into osteoclasts are not fully elucidated. In this study, we identified that the Notch ligand Jagged1 is a previously unrecognized RBP-J target. The expression of Jagged1 is significantly induced by TNF mainly through RBP-J. The TNF-induced Jagged1 in turn functions as a feedback inhibitory regulator of TNF-mediated osteoclastogenesis. This feedback inhibition of osteoclastogenesis by Jagged1 does not exist in RANKL-induced mouse osteoclast differentiation, as RANKL does not induce Jagged1 expression. The Jagged1 level in peripheral blood monocytes/osteoclast precursors is decreased in RA compared with the nonerosive inflammatory disease systemic lupus erythematosus, suggesting a mechanism that contributes to increased osteoclast formation in RA. Moreover, recombinant Jagged1 suppresses human inflammatory osteoclastogenesis. Our findings identify Jagged1 as an RBP-J direct target that links TNF and Notch signaling pathways and restrains TNF-mediated osteoclastogenesis. Given that Jagged1 has no effect on TNF-induced expression of inflammatory genes, its use may present a new complementary therapeutic approach to mitigate inflammatory bone loss with little impact on the immune response in disease conditions.


Subject(s)
Arthritis, Rheumatoid , Bone Resorption , Humans , Animals , Mice , Osteogenesis , Feedback , Osteoclasts/metabolism , Macrophages , Arthritis, Rheumatoid/metabolism , RANK Ligand/metabolism , Cell Differentiation , Tumor Necrosis Factor-alpha/metabolism
17.
Small ; 20(10): e2306736, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37880862

ABSTRACT

Superionic halides have aroused interests in field of solid electrolytes such as Li2 ZrCl6 . However, they are still facing challenges including poor air stability which lacks in-depth investigation. Here, moisture instability of Li2 ZrCl6 is demonstrated and decomposition mechanism in air is clearly revealed. Li2 ZrCl6 decomposes into Li2 ZrO3 , ZrOCl2 ·xH2 O and LiCl during initial stage as halides upon moisture exposure. Later, these side products evolve into LiCl(H2 O) and Li6 Zr2 O7 after longer time exposure. More importantly, structure of destroyed halides cannot be recovered after postheating. Later, Indium is doped into Li2 ZrCl6 (9.7 × 10-5 S cm-1 ) to explore its effect on structure and properties. Crystal structure of ball-milled In-doped Li2 ZrCl6 electrolytes is converted from the Li3 YCl6 -like to Li3 InCl6 -like with increasing In content and ionic conductivity can also be enhanced (0.768-1.13) × 10-3 S cm-1 ). More importantly, good air stability of optimal Li2.8 Zr0.2 In0.8 Cl6 is achieved since halide hydrates are formed after air exposure instead of total decomposition and the hydrates can be restored to Li2.8 Zr0.2 In0.8 Cl6 after postheating. Moreover, reheated Li2.8 Zr0.2 In0.8 Cl6 after air exposure is successfully applied in solid-state LiNi0.8 Co0.1 Mn0.1 O2 /halides/Li6 PS5 Cl/Li-In battery. The results in this work can provide insights into air instability of Li2 ZrCl6 and effective strategy to regulate air stability of halides.

18.
Small ; 20(2): e2306746, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37658491

ABSTRACT

The development of organic materials that deliver room-temperature phosphorescence (RTP) is highly interesting for potential applications such as anticounterfeiting, optoelectronic devices, and bioimaging. Herein, a molecular chaperone strategy for controlling isolated chromophores to achieve high-performance RTP is demonstrated. Systematic experiments coupled with theoretical evidence reveal that the host plays a similar role as a molecular chaperone that anchors the chromophores for limited nonradiative decay and directs the proper conformation of guests for enhanced intersystem crossing through noncovalent interactions. For deduction of structure-property relationships, various structure-related descriptors that correlate with the RTP performance are identified, thus offering the possibility to quantitatively design and predict the phosphorescent behaviors of these systems. Furthermore, application in thermal printing is well realized for these RTP materials. The present work discloses an effective strategy for efficient construction of organic RTP materials, delivering a modular model which is expected to help expand the diversity of desirable RTP systems.

19.
Nat Immunol ; 13(7): 642-50, 2012 May 20.
Article in English | MEDLINE | ID: mdl-22610140

ABSTRACT

Emerging concepts suggest that the functional phenotype of macrophages is regulated by transcription factors that define alternative activation states. We found that RBP-J, the main nuclear transducer of signaling via Notch receptors, augmented Toll-like receptor 4 (TLR4)-induced expression of key mediators of classically activated M1 macrophages and thus of innate immune responses to Listeria monocytogenes. Notch-RBP-J signaling controlled expression of the transcription factor IRF8 that induced downstream M1 macrophage-associated genes. RBP-J promoted the synthesis of IRF8 protein by selectively augmenting kinase IRAK2-dependent signaling via TLR4 to the kinase MNK1 and downstream translation-initiation control through eIF4E. Our results define a signaling network in which signaling via Notch-RBP-J and TLRs is integrated at the level of synthesis of IRF8 protein and identify a mechanism by which heterologous signaling pathways can regulate the TLR-induced inflammatory polarization of macrophages.


Subject(s)
Immunoglobulin J Recombination Signal Sequence-Binding Protein/immunology , Inflammation/immunology , Interferon Regulatory Factors/immunology , Macrophages/immunology , Receptors, Notch/immunology , Animals , Cell Polarity/immunology , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation/immunology , Interferon Regulatory Factors/biosynthesis , Interleukin-1 Receptor-Associated Kinases/immunology , Listeriosis/immunology , Macrophage Activation/immunology , Male , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Transcription Factors/metabolism
20.
Microb Pathog ; 187: 106509, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38185451

ABSTRACT

BACKGROUND: Mastitis is a serious disease which affects animal husbandry, particularly in cow breeding. The etiology of mastitis is complex and its pathological mechanism is not yet fully understood. Our previous research in clinical investigation has revealed that subclinical ketosis can increase the number of somatic cell counts (SCC) in milk, although the underlying mechanism remains unclear. Recent studies have further confirmed the significant role of mastitis. RESULTS: In this study, we aimed to examine the SCC, rumen microbiota, and metabolites in the milkmen of cows with subclinical ketosis. Additionally, we conducted a rumen microbiota transplant into mice to investigate the potential association between rumen microbiota disturbance and mastitis induced by subclinical ketosis in dairy cows. The study has found that cows with subclinical ketosis have a higher SCC in their milk compared to healthy cows. Additionally, there were significant differences in the rumen microbiota and the level of volatile fatty acid (VFA) between cows with subclinical ketosis and healthy cows. Moreover, transplanting the rumen microbiota from subclinical ketosis and mastitis cows into mice can induce mammary inflammation and liver function damage than transplanting the rumen flora from healthy dairy cows. CONCLUSIONS: In addition to the infection of mammary gland by pathogenic microorganisms, there is also an endogenous therapeutic pathway mediated by rumen microbiota. Targeted rumen microbiota modulation may be an effective way to prevent and control mastitis in dairy cows.


Subject(s)
Ketosis , Mastitis, Bovine , Microbiota , Female , Animals , Cattle , Mice , Humans , Mastitis, Bovine/pathology , Rumen/metabolism , Ketosis/metabolism , Ketosis/veterinary , Milk , Lactation
SELECTION OF CITATIONS
SEARCH DETAIL