Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.551
Filter
1.
Biomolecules ; 11(7)2021 07 05.
Article in English | MEDLINE | ID: mdl-34356615

ABSTRACT

The gut epithelial barrier provides the first line of defense protecting the internal milieu from the environment. To circumvent the exposure to constant challenges such as pathogenic infections and commensal bacteria, epithelial and immune cells at the gut barrier require rapid and efficient means to dynamically sense and respond to stimuli. Numerous studies have highlighted the importance of proteolysis in maintaining homeostasis and adapting to the dynamic changes of the conditions in the gut environment. Primarily, proteolytic activities that are involved in immune regulation and inflammation have been examined in the context of the lysosome and inflammasome activation. Yet, the key to cellular and tissue proteostasis is the ubiquitin-proteasome system, which tightly regulates fundamental aspects of inflammatory signaling and protein quality control to provide rapid responses and protect from the accumulation of proteotoxic damage. In this review, we discuss proteasome-dependent regulation of the gut and highlight the pathophysiological consequences of the disarray of proteasomal control in the gut, in the context of aberrant inflammatory disorders and tumorigenesis.


Subject(s)
Intestinal Mucosa , Proteasome Endopeptidase Complex , Proteolysis , Signal Transduction/immunology , Animals , Enzyme Activation/immunology , Humans , Inflammation/enzymology , Intestinal Mucosa/enzymology , Intestinal Mucosa/immunology , Lysosomes/enzymology , Lysosomes/immunology , Proteasome Endopeptidase Complex/immunology , Proteasome Endopeptidase Complex/metabolism
2.
Front Immunol ; 12: 660065, 2021.
Article in English | MEDLINE | ID: mdl-34234775

ABSTRACT

Toll-like receptors (TLRs) play a crucial role in the recognition of pathogen-derived components as a first line of defense against infections. It has been suggested that depending on the nature of the pathogens, TLRs activation induce a distinct cytokine profile that may contribute to the polarization of the acquired immune response. Here, we investigated the early MAPK signaling activation via TLR4 and TLR2 receptors and its impact in differential cytokine profile by macrophages. We found that TLR2 ligands activated MAPKs p38 and ERK earlier compared to the TLR4 ligand LPS in macrophages. Higher IL-10/IL-12 and IL-10/TNF-α ratios were also observed at later time points in response to TLR2 ligands compared to LPS. The results also indicate an earlier activation of the phosphatase MKP-1 and that MKP-1 KO macrophages show a prolongation in p38 phosphorylation in response to TLR2 stimulation. Furthermore, p38 is critical for IL-10 expression in response to TLR2 ligands, which triggers the macrophage change to a M2 and regulatory phenotype in contrast to the M1 phenotype induced by TLR4 activation. Therefore, the early TLR2-mediated p38 induction contributes for the high IL-10 production, likely as a virulence strategy to suppress host Th1 response against certain types of pathogens.


Subject(s)
Dual Specificity Phosphatase 1/immunology , Interleukin-10/immunology , Macrophages/immunology , Toll-Like Receptor 2/immunology , p38 Mitogen-Activated Protein Kinases/immunology , Animals , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Diglycerides/pharmacology , Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 1/metabolism , Enzyme Activation/immunology , Gene Expression/drug effects , Gene Expression/immunology , Interleukin-10/genetics , Interleukin-10/metabolism , Lipopeptides/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligopeptides/pharmacology , Phosphorylation/drug effects , RAW 264.7 Cells , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Front Immunol ; 12: 650779, 2021.
Article in English | MEDLINE | ID: mdl-34194428

ABSTRACT

Listeria monocytogenes (L.m) is efficiently controlled by several cells of the innate immunity, including the Mast Cell (MC). MC is activated by L.m inducing its degranulation, cytokine production and microbicidal mechanisms. TLR2 is required for the optimal control of L.m infection by different cells of the immune system. However, little is known about the MC receptors involved in recognizing this bacterium and whether these interactions mediate MC activation. In this study, we analyzed whether TLR2 is involved in mediating different MC activation responses during L.m infection. We found that despite MC were infected with L.m, they were able to clear the bacterial load. In addition, MC degranulated and produced ROS, TNF-α, IL-1ß, IL-6, IL-13 and MCP-1 in response to bacterial infection. Interestingly, L.m induced the activation of signaling proteins: ERK, p38 and NF-κB. When TLR2 was blocked, L.m endocytosis, bactericidal activity, ROS production and mast cell degranulation were not affected. Interestingly, only IL-6 and IL-13 production were affected when TLR2 was inhibited in response to L.m infection. Furthermore, p38 activation depended on TLR2, but not ERK or NF-κB activation. These results indicate that TLR2 mediates only some MC activation pathways during L.m infection, mainly those related to IL-6 and IL-13 production.


Subject(s)
Interleukin-13/immunology , Interleukin-6/immunology , Listeria monocytogenes/immunology , Mast Cells/immunology , Toll-Like Receptor 2/immunology , Animals , Cell Degranulation/immunology , Cell Degranulation/physiology , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Enzyme Activation/immunology , Host-Pathogen Interactions/immunology , Interleukin-13/metabolism , Interleukin-6/metabolism , Listeria monocytogenes/physiology , Mast Cells/microbiology , Mast Cells/physiology , Mice, Inbred C57BL , NF-kappa B/immunology , NF-kappa B/metabolism , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Toll-Like Receptor 2/metabolism , p38 Mitogen-Activated Protein Kinases/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Biochem Soc Trans ; 49(3): 1311-1324, 2021 06 30.
Article in English | MEDLINE | ID: mdl-34060593

ABSTRACT

Innate immune responses are tightly regulated by various pathways to control infections and maintain homeostasis. One of these pathways, the inflammasome pathway, activates a family of cysteine proteases called inflammatory caspases. They orchestrate an immune response by cleaving specific cellular substrates. Canonical inflammasomes activate caspase-1, whereas non-canonical inflammasomes activate caspase-4 and -5 in humans and caspase-11 in mice. Caspases are highly specific enzymes that select their substrates through diverse mechanisms. During inflammation, caspase activity is responsible for the secretion of inflammatory cytokines and the execution of a form of lytic and inflammatory cell death called pyroptosis. This review aims to bring together our current knowledge of the biochemical processes behind inflammatory caspase activation, substrate specificity, and substrate signalling.


Subject(s)
Caspases/immunology , Cytokines/immunology , Inflammasomes/immunology , Inflammation/immunology , Signal Transduction/immunology , Animals , Caspases/metabolism , Cytokines/metabolism , Enzyme Activation/immunology , Humans , Inflammasomes/metabolism , Inflammation/metabolism , Pyroptosis/immunology , Substrate Specificity
5.
Sci Rep ; 11(1): 1458, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33446766

ABSTRACT

T cell activation is intimately linked to metabolism, as distinct metabolic requirements support the functional and phenotypical differences between quiescent and activated T cells. Metabolic transition from mitochondrial oxidative phosphorylation to aerobic glycolysis is crucial for a proper T cell activation. However, the role of tricarboxylic acid cycle (TCA), and in particular succinate dehydrogenase (SDH) in activated T cells needs further elucidation. Here we show that inhibition of SDH during activation of T cells results in strong impairment of proliferation, expression of activation markers, and production of key inflammatory cytokines, despite a concomitant increase in glycolytic metabolic activity. Similar effect of SDH inhibition were demonstrated in pre-activated T cell. Interestingly, itaconic acid, an endogenous SDH inhibitor released from activated macrophages and dendritic cells, had no immunomodulator effect. Taken together, our findings demonstrate that SDH enzyme fitness is critical for mounting and maintaining appropriate activation and function of human T cells.


Subject(s)
Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Lymphocyte Activation/drug effects , Succinate Dehydrogenase/antagonists & inhibitors , T-Lymphocytes/immunology , Cytokines/biosynthesis , Cytokines/immunology , Enzyme Activation/drug effects , Enzyme Activation/immunology , Gene Expression Regulation/immunology , Humans , Succinate Dehydrogenase/immunology
6.
Dev Comp Immunol ; 117: 103986, 2021 04.
Article in English | MEDLINE | ID: mdl-33359739

ABSTRACT

Entomopathogenic fungi have high potential for controlling insect pests, although the slow killing speed has blocked their widespread application. To increase the virulence of entomopathogenic fungi, genetic modification can be employed. Egf1.0 is an immunosuppressive protein encoded by polydnavirus, carried by parasitoid wasp Microplitis demolitor, which blocks the prophenoloxidase (PPO) activation response of host insects. In this study, we explored the feasibility of genetically modifying entomopathogenic fungi with increased virulence by expressing Egf1.0. In comparison with the wild-type parents, the median lethal concentration (LC50) of Beauveria bassiana expressing Egf1.0 against Helicoverpa armigera was reduced by 2.7-fold, and the median lethal time (LT50) was reduced by 22.8%. In vitro assay showed that recombinant Egf1.0 was able to inhibit the PPO activation response of H. armigera. In vivo assay revealed that the expression of Egf1.0 in B. bassiana caused a higher degree of suppression to PPO activation response of H. armigera. These assays suggested that the increased virulence of the transgenic fungi is due to the increased ability to suppress the host insect's immune response. Moreover, colony growth, conidia yield, and germination assays revealed that the expression of Egf1.0 in B. bassiana had no effect on its growth and development. In conclusion, the expression of Egf1.0 can significantly enhance the pathogenicity of B. bassiana against host insects.


Subject(s)
Beauveria/immunology , Insect Proteins/immunology , Monophenol Monooxygenase/immunology , Moths/immunology , Transgenes/immunology , Viral Proteins/immunology , Animals , Base Sequence , Beauveria/genetics , Beauveria/pathogenicity , Enzyme Activation/genetics , Enzyme Activation/immunology , Gene Expression Regulation/immunology , Host-Pathogen Interactions/immunology , Insect Proteins/genetics , Insect Proteins/metabolism , Monophenol Monooxygenase/genetics , Monophenol Monooxygenase/metabolism , Moths/metabolism , Moths/microbiology , Transgenes/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Virulence/genetics , Virulence/immunology
7.
Int Immunol ; 33(3): 149-160, 2021 03 01.
Article in English | MEDLINE | ID: mdl-32986079

ABSTRACT

Innate lymphoid cells (ILCs) are a family of developmentally related leukocytes that rapidly secrete polarized sets of cytokines to combat infection and promote tissue repair at mucosal barriers. Among them, group 3 ILCs (ILC3s) play an important role in maintenance of the gut homeostasis by producing IL-22, and their development and function critically depend on the transcription factor RORγt. Although recent evidence indicates that RORγt+ ILC3s are reduced in the gut in the absence of the Cdc42 activator DOCK8 (dedicator of cytokinesis 8), the underlying mechanism remains unclear. We found that genetic deletion of Dock8 in RORγt+-lineage cells markedly reduced ILC3s in the lamina propria of the small intestine. By analyzing BrdU incorporation, it was revealed that DOCK8 deficiency did not affect the cell proliferation. Furthermore, when lineage marker-negative (Lin-) α4ß7+ CD127+ RORγt- fetal liver cells were cultured with OP9 stromal cells in the presence of stem cell factor (SCF) and IL-7 in vitro, RORγt+ ILC3s normally developed irrespective of DOCK8 expression. However, DOCK8-deficient ILC3s exhibited a severe defect in survival of ILC3s under the condition with or without IL-7. Similar defects were observed when we analyzed Dock8VAGR mice having mutations in the catalytic center of DOCK8, thereby failing to activate Cdc42. Thus, DOCK8 acts in cell-autonomous manner to control survival of ILC3s in the gut through Cdc42 activation.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Intestinal Mucosa/cytology , Lymphocytes/metabolism , cdc42 GTP-Binding Protein/metabolism , Animals , Catalytic Domain/genetics , Cell Line , Cell Proliferation/genetics , Cell Survival/genetics , Cytokines/metabolism , Enzyme Activation/immunology , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , Interleukin-7/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Stem Cell Factor/metabolism
8.
Front Immunol ; 12: 801781, 2021.
Article in English | MEDLINE | ID: mdl-35003129

ABSTRACT

Marek's disease virus (MDV), an avian alphaherpesvirus, infects chickens, transforms CD4+ T cells, and induces immunosuppression early during infection. However, the exact mechanisms involved in MDV-induced immunosuppression are yet to be identified. Here, our results demonstrate that MDV infection in vitro and in vivo induces activation of cyclooxygenase-2 (COX-2) and production of prostaglandin E2 (PGE2). This exerts its inhibitory effects on T cell proliferation at day 21 post infection via PGE2 receptor 2 (EP2) and receptor 4 (EP4). Impairment of the MDV-induced T cell proliferation was associated with downregulation of IL-2 and transferrin uptake in a COX-2/PGE2 dependent manner in vitro. Interestingly, oral administration of a COX-2 inhibitor, meloxicam, during MDV infection inhibited COX-2 activation and rescued T cell proliferation at day 21 post infection. Taken together, our results reveal a novel mechanism that contributes to immunosuppression in the MDV-infected chickens.


Subject(s)
Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Immune Tolerance/immunology , Marek Disease/immunology , T-Lymphocytes/immunology , Animals , Cell Proliferation/physiology , Chickens , Enzyme Activation/immunology , Herpesvirus 2, Gallid , Lymphocyte Activation/immunology , Marek Disease/metabolism , Marek Disease/virology
9.
Front Immunol ; 12: 790258, 2021.
Article in English | MEDLINE | ID: mdl-35069570

ABSTRACT

Coordination among multiple signaling pathways ensures an appropriate immune response, where a signaling pathway may impair or augment another signaling pathway. Here, we report a negative feedback regulation of signaling through the key innate immune mediator MyD88 by inflammasome-activated caspase-1. NLRP3 inflammasome activation impaired agonist- or infection-induced TLR signaling and cytokine production through the proteolytic cleavage of MyD88 by caspase-1. Site-specific mutagenesis was used to identify caspase-1 cleavage site within MyD88 intermediary segment. Different cleavage site location within MyD88 defined the functional consequences of MyD88 cleavage between mouse and human cells. LPS/monosodium urate-induced mouse inflammation model corroborated the physiological role of this mechanism of regulation, that could be reversed by chemical inhibition of NLRP3. While Toll/interleukin-1 receptor (TIR) domain released by MyD88 cleavage additionally contributed to the inhibition of signaling, Waldenström's macroglobulinemia associated MyD88L265P mutation is able to evade the caspase-1-mediated inhibition of MyD88 signaling through the ability of its TIRL265P domain to recruit full length MyD88 and facilitate signaling. The characterization of this mechanism reveals an additional layer of innate immunity regulation.


Subject(s)
Caspase 1/immunology , Immunity, Innate , Inflammasomes/immunology , Myeloid Differentiation Factor 88/immunology , Signal Transduction/immunology , Animals , Caspase 1/genetics , Enzyme Activation/immunology , HEK293 Cells , Humans , Inflammasomes/genetics , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Signal Transduction/genetics , THP-1 Cells
10.
Int Rev Cell Mol Biol ; 357: 35-55, 2020.
Article in English | MEDLINE | ID: mdl-33234244

ABSTRACT

The Tyro3, Axl, and MerTK (TAM) receptors are three homologous Type I Receptor Tyrosine Kinases that have important homeostatic functions in multicellular organisms by regulating the clearance of apoptotic cells (efferocytosis). Pathologically, TAM receptors are overexpressed in a wide array of human cancers, and often associated with aggressive tumor grade and poor overall survival. In addition to their expression on tumor cells, TAMs are also expressed on infiltrating myeloid-derived cells in the tumor microenvironment, where they appear to act akin to negative immune checkpoints that impair host anti-tumor immunity. The ligands for TAMs are two endogenous proteins, Growth Arrest-Specific 6 (Gas6) and Protein S (Pros1), that function as bridging molecules between externalized phosphatidylserine (PtdSer) on apoptotic cells and the TAM ectodomains. One interesting feature of TAMs biology is that their ligand proteins require specific post-translational modifications to acquire activities. This chapter summarized these important modifications and explained the molecular mechanisms behind such phenomenon. Current evidences suggest that these modifications help Gas6/Pros1 to achieve optimal PtdSer-binding capacities. In addition, this chapter included recent discovery of regulating machineries of PtdSer dynamic across the plasma membrane, as well as their potential impacts in the tumor microenvironment. Taken together, this review highlights the importance of the upstream PtdSer and Gas6 in regulating TAMs' function and hope to provide researchers with new perspectives to inspire future studies of TAM receptors in human disease models.


Subject(s)
Neoplasms/immunology , Protein Processing, Post-Translational/immunology , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Tumor Microenvironment/immunology , c-Mer Tyrosine Kinase/immunology , Animals , Apoptosis/immunology , Enzyme Activation/immunology , Humans , Intercellular Signaling Peptides and Proteins/immunology , Neoplasms/pathology , Protein S/immunology , Axl Receptor Tyrosine Kinase
11.
ACS Synth Biol ; 9(10): 2828-2839, 2020 10 16.
Article in English | MEDLINE | ID: mdl-32927940

ABSTRACT

Synthetic biology and metabolic engineering offer potentially green and attractive routes to the production of high value compounds. The provision of high-quality parts and pathways is crucial in enabling the biosynthesis of chemicals using synthetic biology. While a number of regulatory parts that provide control at the transcriptional and translational level have been developed, relatively few exist at the protein level. Single domain antibodies (sdAb) such as camelid heavy chain variable fragments (VHH) possess binding characteristics which could be exploited for their development and use as novel parts for regulating metabolic pathways at the protein level in microbial cell factories. Here, a platform for the use of VHH as tools in Escherichia coli is developed and subsequently used to modulate linalool production in E. coli. The coproduction of a Design of Experiments (DoE) optimized pBbE8k His6-VHHCyDisCo system alongside a heterologous linalool production pathway facilitated the identification of anti-bLinS VHH that functioned as modulators of bLinS. This resulted in altered product profiles and significant variation in the titers of linalool, geraniol, nerolidol, and indole obtained. The ability to alter the production levels of high value terpenoids, such as linalool, in a tunable manner at the protein level could represent a significant step forward for the development of improved microbial cell factories. This study serves as a proof of principle indicating that VHH can be used to modulate enzyme activity in engineered pathways within E. coli. Given their almost limitless binding potential, we posit that single domain antibodies could emerge as powerful regulatory parts in synthetic biology applications.


Subject(s)
Acyclic Monoterpenes/metabolism , Bacterial Proteins/immunology , Escherichia coli/genetics , Escherichia coli/metabolism , Hydro-Lyases/immunology , Immunoglobulin Heavy Chains/immunology , Metabolic Engineering/methods , Single-Domain Antibodies/immunology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bioreactors , Camelids, New World/immunology , Codon , Enzyme Activation/immunology , Hydro-Lyases/genetics , Hydro-Lyases/metabolism , Streptomyces/enzymology , Streptomyces/genetics , Synthetic Biology/methods
12.
Infection ; 48(5): 665-669, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32737833

ABSTRACT

Novel severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) became pandemic by the end of March 2020. In contrast to the 2002-2003 SARS-CoV outbreak, which had a higher pathogenicity and lead to higher mortality rates, SARSCoV-2 infection appears to be much more contagious. Moreover, many SARS-CoV-2 infected patients are reported to develop low-titer neutralizing antibody and usually suffer prolonged illness, suggesting a more effective SARS-CoV-2 immune surveillance evasion than SARS-CoV. This paper summarizes the current state of art about the differences and similarities between the pathogenesis of the two coronaviruses, focusing on receptor binding domain, host cell entry and protease activation. Such differences may provide insight into possible intervention strategies to fight the pandemic.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/epidemiology , Pandemics , Pneumonia, Viral/epidemiology , Severe Acute Respiratory Syndrome/epidemiology , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Spike Glycoprotein, Coronavirus/immunology , Angiotensin-Converting Enzyme 2 , Antibodies, Viral/biosynthesis , Betacoronavirus/immunology , COVID-19 , Cathepsins/genetics , Cathepsins/immunology , Coronavirus Infections/enzymology , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Enzyme Activation/immunology , Humans , Immune Evasion , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/immunology , Pneumonia, Viral/enzymology , Pneumonia, Viral/immunology , Pneumonia, Viral/pathology , Protein Binding , Protein Domains , Severe acute respiratory syndrome-related coronavirus/immunology , SARS-CoV-2 , Serine Endopeptidases/genetics , Serine Endopeptidases/immunology , Severe Acute Respiratory Syndrome/enzymology , Severe Acute Respiratory Syndrome/immunology , Severe Acute Respiratory Syndrome/pathology , Severity of Illness Index , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Virus Internalization , Virus Replication
13.
J Immunol ; 205(6): 1564-1579, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32747502

ABSTRACT

Middle East respiratory syndrome coronavirus (MERS-CoV) is a highly pathogenic human coronavirus causing severe disease and mortality. MERS-CoV infection failed to elicit robust IFN response, suggesting that the virus might have evolved strategies to evade host innate immune surveillance. In this study, we identified and characterized type I IFN antagonism of MERS-CoV open reading frame (ORF) 8b accessory protein. ORF8b was abundantly expressed in MERS-CoV-infected Huh-7 cells. When ectopically expressed, ORF8b inhibited IRF3-mediated IFN-ß expression induced by Sendai virus and poly(I:C). ORF8b was found to act at a step upstream of IRF3 to impede the interaction between IRF3 kinase IKKε and chaperone protein HSP70, which is required for the activation of IKKε and IRF3. An infection study using recombinant wild-type and ORF8b-deficient MERS-CoV further confirmed the suppressive role of ORF8b in type I IFN induction and its disruption of the colocalization of HSP70 with IKKε. Ectopic expression of HSP70 relieved suppression of IFN-ß expression by ORF8b in an IKKε-dependent manner. Enhancement of IFN-ß induction in cells infected with ORF8b-deficient virus was erased when HSP70 was depleted. Taken together, HSP70 chaperone is important for IKKε activation, and MERS-CoV ORF8b suppresses type I IFN expression by competing with IKKε for interaction with HSP70.


Subject(s)
Enzyme Activation/immunology , I-kappa B Kinase/immunology , Interferon Type I/immunology , Middle East Respiratory Syndrome Coronavirus/immunology , Viral Proteins/immunology , Betacoronavirus , COVID-19 , Cell Line , Coronavirus Infections , HSP70 Heat-Shock Proteins/immunology , HSP70 Heat-Shock Proteins/metabolism , Humans , I-kappa B Kinase/metabolism , Interferon Type I/metabolism , Middle East Respiratory Syndrome Coronavirus/metabolism , Pandemics , Pneumonia, Viral , SARS-CoV-2 , Viral Proteins/metabolism
14.
PLoS Pathog ; 16(5): e1008572, 2020 05.
Article in English | MEDLINE | ID: mdl-32413093

ABSTRACT

The apicomplexan Toxoplasma gondii induces strong protective immunity dependent upon recognition by Toll-like receptors (TLR)11 and 12 operating in conjunction with MyD88 in the murine host. However, TLR11 and 12 proteins are not present in humans, inspiring us to investigate MyD88-independent pathways of resistance. Using bicistronic IL-12-YFP reporter mice on MyD88+/+ and MyD88-/- genetic backgrounds, we show that CD11c+MHCII+F4/80- dendritic cells, F4/80+ macrophages, and Ly6G+ neutrophils were the dominant cellular sources of IL-12 in both wild type and MyD88 deficient mice after parasite challenge. Parasite dense granule protein GRA24 induces p38 MAPK activation and subsequent IL-12 production in host macrophages. We show that Toxoplasma triggers an early and late p38 MAPK phosphorylation response in MyD88+/+ and MyD88-/- bone marrow-derived macrophages. Using the uracil auxotrophic Type I T. gondii strain cps1-1, we demonstrate that the late response does not require active parasite proliferation, but strictly depends upon GRA24. By i. p. inoculation with cps1-1 and cps1-1:Δgra24, we identified unique subsets of chemokines and cytokines that were up and downregulated by GRA24. Finally, we demonstrate that cps1-1 triggers a strong host-protective GRA24-dependent Th1 response in the absence of MyD88. Our data identify GRA24 as a major mediator of p38 MAPK activation, IL-12 induction and protective immunity that operates independently of the TLR/MyD88 cascade.


Subject(s)
Interleukin-12/immunology , MAP Kinase Signaling System/immunology , Macrophages/immunology , Myeloid Differentiation Factor 88/immunology , Protozoan Proteins/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , p38 Mitogen-Activated Protein Kinases/immunology , Animals , Enzyme Activation/genetics , Enzyme Activation/immunology , Interleukin-12/genetics , MAP Kinase Signaling System/genetics , Macrophages/parasitology , Macrophages/pathology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Protozoan Proteins/genetics , Toxoplasma/genetics , Toxoplasmosis/genetics , Toxoplasmosis/pathology , p38 Mitogen-Activated Protein Kinases/genetics
15.
Eur J Immunol ; 50(6): 809-821, 2020 06.
Article in English | MEDLINE | ID: mdl-32022930

ABSTRACT

Terminal differentiation of B cells into antibody-secreting cells is the foundation of humoral immune response. B-1 cells, which are different from B-2 cells, preferentially differentiate into plasma cells. CMTM7 is a MARVEL-domain-containing membrane protein predominantly expressed in B cells that plays an important role in B-1a cell development. The present study assessed CMTM7 function in response to antigen stimulation. Following immunization with T cell-dependent and T cell-independent antigens, Cmtm7-deficient mice exhibited decreased IgM but normal IgG responses in vivo. In vitro stimulation with LPSs induced Cmtm7-/- B-1 cell activation, whereas proliferation was marginally reduced. Notably, Cmtm7 deficiency markedly suppressed plasma cell differentiation in response to TLR agonists, accompanied by a decrease in IgM and IL-10 production. At the molecular level, loss of Cmtm7 repressed the downregulation of Pax5 and the upregulation of Xbp1, Irf4, and Prdm1. Furthermore, p38 phosphorylation was inhibited in Cmtm7-/- B-1 cells. Experiments using a p38 inhibitor revealed that p38 activation was essential for the terminal differentiation of B-1 cells, suggesting that Cmtm7 contributes to B-1 cell differentiation by maintaining p38 activation. Overall, the data reveal the crucial functions of CMTM7 in TLR-induced terminal differentiation and p38 activation in B-1 cells.


Subject(s)
B-Lymphocyte Subsets/immunology , Cell Differentiation/immunology , Chemokines/immunology , MAP Kinase Signaling System/immunology , MARVEL Domain-Containing Proteins/immunology , Plasma Cells/immunology , Toll-Like Receptors/immunology , p38 Mitogen-Activated Protein Kinases/immunology , Animals , B-Lymphocyte Subsets/cytology , Cell Differentiation/genetics , Chemokines/genetics , Enzyme Activation/genetics , Enzyme Activation/immunology , Immunoglobulin M/genetics , Immunoglobulin M/immunology , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Interleukin-10/genetics , Interleukin-10/immunology , MAP Kinase Signaling System/genetics , MARVEL Domain-Containing Proteins/genetics , Mice , Mice, Knockout , Plasma Cells/cytology , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/immunology , Toll-Like Receptors/genetics , p38 Mitogen-Activated Protein Kinases/genetics
16.
J Immunol ; 204(5): 1322-1333, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31996460

ABSTRACT

With the development of liver surgery, ischemia-reperfusion (IR) injury has received increasing attention. Roquin-1 has been shown to play an important role in innate immune and immune balance. We demonstrate that Roquin-1 expression increased at 1 h after IR and then decreased in C57B/L mice. The immunofluorescence double-label showed that Roquin-1 was mainly expressed in macrophages (mø). Furthermore, we used clodronate liposomes to remove mø, and injected the bone marrow-derived mø (BMDM) through the tail vein in 1 h before IR. We found that liver IR injury was aggravated by Roquin-1 interference. The results of PCR and ELISA suggested that after interference with Roquin-1, mø increased toward M1 and decreased toward M2. Then, interference with Roquin-1 promoted the polarization of mø to M1 and inhibited the polarization of M2. By Western blot technology and AMPKα and mTOR inhibitors, we found that Roquin-1 promotes the phosphorylation of mTOR and STAT3 by inhibiting the phosphorylation of AMPKα. We used AICAR to activate AMPKα in mø and found that the level of ubiquitination of AMPKα was decreased after activation of AMPKα. Furthermore, by bioinformatics methods, we identified potential ubiquitination sites on AMPKα. By the point mutation experiments in vitro, we confirmed that the ubiquitination of these sites is regulated by Roquin-1. Meanwhile, Roquin-1 interference inhibited the activation and function of AMPKα. This topic describes the protection of liver IR injury by Roquin-1 and discusses its main mechanism for regulating AMPKα activity through ubiquitination and affecting the polarization of mø.


Subject(s)
Liver Diseases/immunology , Liver/immunology , Macrophages/immunology , Reperfusion Injury/immunology , Ubiquitin-Protein Ligases/immunology , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/immunology , Animals , Enzyme Activation/immunology , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Liver/pathology , Liver Diseases/genetics , Liver Diseases/pathology , Macrophages/pathology , Male , Mice , Point Mutation , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics , Ubiquitination/immunology
17.
Front Immunol ; 11: 617404, 2020.
Article in English | MEDLINE | ID: mdl-33633735

ABSTRACT

The role of PI3K-mTOR pathway in regulating NK cell development has been widely reported. However, it remains unclear whether NK cell development depends on the protein kinase B (PKB), which links PI3K and mTOR, perhaps due to the potential redundancy of PKB. PKB has two phosphorylation sites, threonine 308 (T308) and serine 473 (S473), which can be phosphorylated by phosphoinositide-dependent protein kinase-1 (PDK1) and mTORC2, respectively. In this study, we established a mouse model in which PKB was inactivated through the deletion of PDK1 and Rictor, a key component of mTORC2, respectively. We found that the single deletion of PDK1 or Rictor could lead to a significant defect in NK cell development, while combined deletion of PDK1 and Rictor severely hindered NK cell development at the early stage. Notably, ectopic expression of myristoylated PKB significantly rescued this defect. In terms of mechanism, in PDK1/Rictor-deficient NK cells, E4BP4, a transcription factor for NK cell development, was less expressed, and the exogenous supply of E4BP4 could alleviate the developmental defect of NK cell in these mice. Besides, overexpression of Bcl-2 also helped the survival of PDK1/Rictor-deficient NK cells, suggesting an anti-apoptotic role of PKB in NK cells. In summary, complete phosphorylation of PKB at T308 and S473 by PDK1 and mTORC2 is necessary for optimal NK cell development, and PKB regulates NK cell development by promoting E4BP4 expression and preventing cell apoptosis.


Subject(s)
Killer Cells, Natural/immunology , Mechanistic Target of Rapamycin Complex 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Animals , Cell Differentiation/immunology , Enzyme Activation/immunology , Killer Cells, Natural/metabolism , Mechanistic Target of Rapamycin Complex 2/immunology , Mice , Phosphorylation , Proto-Oncogene Proteins c-akt/immunology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/immunology , Signal Transduction/immunology
18.
Immunity ; 52(1): 109-122.e6, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31882361

ABSTRACT

Recent work suggests that cholesterol metabolism impacts innate immune responses against infection. However, the key enzymes or the natural products and mechanisms involved are not well elucidated. Here, we have shown that upon DNA and RNA viral infection, macrophages reduced 7-dehydrocholesterol reductase (DHCR7) expression. DHCR7 deficiency or treatment with the natural product 7-dehydrocholesterol (7-DHC) could specifically promote phosphorylation of IRF3 (not TBK1) and enhance type I interferon (IFN-I) production in macrophages. We further elucidated that viral infection or 7-DHC treatment enhanced AKT3 expression and activation. AKT3 directly bound and phosphorylated IRF3 at Ser385, together with TBK1-induced phosphorylation of IRF3 Ser386, to achieve IRF3 dimerization. Deletion of DHCR7 and the DHCR7 inhibitors including AY9944 and the chemotherapy drug tamoxifen promoted clearance of Zika virus and multiple viruses in vitro or in vivo. Taken together, we propose that the DHCR7 inhibitors and 7-DHC are potential therapeutics against emerging or highly pathogenic viruses.


Subject(s)
Dehydrocholesterols/metabolism , Interferon Regulatory Factor-3/metabolism , Interferon Type I/biosynthesis , Macrophages/immunology , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Vesicular Stomatitis/immunology , A549 Cells , Animals , Cell Line , Cholesterol/metabolism , Enzyme Activation/immunology , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Oxidoreductases Acting on CH-CH Group Donors/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RAW 264.7 Cells , RNA Interference , RNA, Small Interfering/genetics , Vesicular stomatitis Indiana virus/immunology
19.
Front Immunol ; 10: 2673, 2019.
Article in English | MEDLINE | ID: mdl-31824484

ABSTRACT

It is known that Trypanosoma congolense infection in mice is associated with increased production of proinflammatory cytokines by macrophages and monocytes. However, the intracellular signaling pathways leading to the production of these cytokines still remain unknown. In this paper, we have investigated the innate receptors and intracellular signaling pathways that are associated with T. congolense-induced proinflammatory cytokine production in macrophages. We show that the production of IL-6, IL-12, and TNF-α by macrophages in vitro and in vivo following interaction with T. congolense is dependent on phosphorylation of mitogen-activated protein kinase (MAPK) including ERK, p38, JNK, and signal transducer and activation of transcription (STAT) proteins. Specific inhibition of MAPKs and STATs signaling pathways significantly inhibited T. congolense-induced production of proinflammatory cytokines in macrophages. We further show that T. congolense-induced proinflammatory cytokine production in macrophages is mediated via Toll-like receptor 2 (TLR2) and involves the adaptor molecule, MyD88. Deficiency of MyD88 and TLR2 leads to impaired cytokine production by macrophages in vitro and acute death of T. congolense-infected relatively resistant mice. Collectively, our results provide insight into T. congolense-induced activation of the immune system that leads to the production of proinflammatory cytokines and resistance to the infection.


Subject(s)
Myeloid Differentiation Factor 88/metabolism , Toll-Like Receptor 2/metabolism , Trypanosomiasis, African/immunology , Trypanosomiasis, African/metabolism , Adenylate Kinase/immunology , Adenylate Kinase/metabolism , Animals , Cytokines/biosynthesis , Enzyme Activation/immunology , Female , Macrophages/immunology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/immunology , STAT Transcription Factors/immunology , STAT Transcription Factors/metabolism , Toll-Like Receptor 2/immunology , Trypanosoma congolense/immunology
20.
Int J Mol Sci ; 20(22)2019 Nov 12.
Article in English | MEDLINE | ID: mdl-31726690

ABSTRACT

Interferon (IFN)-γ is mainly secreted by CD4+ T helper 1 (Th1), natural killer (NK) and NKT cells after skin injury. Although IFN-γ is well known regarding its inhibitory effects on collagen synthesis by fibroblasts in vitro, information is limited regarding its role in wound healing in vivo. In the present study, we analyzed how the defect of IFN-γ affects wound healing. Full-thickness wounds were created on the backs of wild type (WT) C57BL/6 and IFN-γ-deficient (KO) mice. We analyzed the percent wound closure, wound breaking strength, accumulation of leukocytes, and expression levels of COL1A1, COL3A1, and matrix metalloproteinases (MMPs). IFN-γKO mice exhibited significant attenuation in wound closure on Day 10 and wound breaking strength on Day 14 after wound creation, characteristics that are associated with prolonged neutrophil accumulation. Expression levels of COL1A1 and COL3A1 mRNA were lower in IFN-γKO than in WT mice, whereas expression levels of MMP-2 (gelatinase) mRNA were significantly greater in IFN-γKO than in WT mice. Moreover, under neutropenic conditions created with anti-Gr-1 monoclonal antibodies, wound closure in IFN-γKO mice was recovered through low MMP-2 expression levels. These results suggest that IFN-γ may be involved in the proliferation and maturation stages of wound healing through the regulation of neutrophilic inflammatory responses.


Subject(s)
Gene Expression Regulation, Enzymologic/immunology , Interferon-gamma/deficiency , Matrix Metalloproteinase 2/immunology , Neutrophils/immunology , Wound Healing/immunology , Animals , Collagen Type I/genetics , Collagen Type I/immunology , Collagen Type I, alpha 1 Chain , Collagen Type III/genetics , Collagen Type III/immunology , Enzyme Activation/genetics , Enzyme Activation/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interferon-gamma/immunology , Matrix Metalloproteinase 2/genetics , Mice , Mice, Knockout , Neutrophils/pathology , Wound Healing/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...