Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters











Publication year range
1.
Eur J Immunol ; 51(11): 2607-2617, 2021 11.
Article in English | MEDLINE | ID: mdl-34392531

ABSTRACT

Rearrangement of the actin cytoskeleton is critical for cytotoxic and immunoregulatory functions as well as migration of natural killer (NK) cells. However, dynamic reorganization of actin is a complex process, which remains largely unknown. Here, we investigated the role of the protein Cereblon (CRBN), an E3 ubiquitin ligase complex co-receptor and the primary target of the immunomodulatory drugs, in NK cells. We observed that CRBN partially colocalizes with F-actin in chemokine-treated NK cells and is recruited to the immunological synapse, thus suggesting a role for this protein in cytoskeleton reorganization. Accordingly, silencing of CRBN in NK cells results in a reduced cytotoxicity that correlates with a defect in conjugate and lytic synapse formation. Moreover, CRBN depletion significantly impairs the ability of NK cells to migrate and reduces the enhancing effect of lenalidomide on NK cell migration. Finally, we provided evidence that CRBN is required for activation of the small GTPase Rac1, a critical mediator of cytoskeleton dynamics. Indeed, in CRBN-depleted NK cells, chemokine-mediated or target cell-mediated Rac1 activation is significantly reduced. Altogether our data identify a critical role for CRBN in regulating NK cell functions and suggest that this protein may mediate the stimulatory effect of lenalidomide on NK cells.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Cell Movement/immunology , Cytotoxicity, Immunologic/immunology , Killer Cells, Natural/immunology , Ubiquitin-Protein Ligases/immunology , rac1 GTP-Binding Protein/immunology , Cell Movement/drug effects , Cytotoxicity, Immunologic/drug effects , Humans , Immunomodulating Agents/pharmacology , Killer Cells, Natural/drug effects , Lenalidomide/pharmacology
2.
Trends Immunol ; 41(4): 286-299, 2020 04.
Article in English | MEDLINE | ID: mdl-32122794

ABSTRACT

Sarcoidosis is an enigmatic multisystem disease characterized by the development and accumulation of granulomas: a compact collection of macrophages that have differentiated into epithelioid cells and which are associated with T helper (Th)1 and Th17 cells. Although no single causative factor has been shown to underlie sarcoidosis in humans, its etiology has been related to microbial, environmental, and genetic factors. We examine how these factors play a role in sarcoidosis pathogenesis. Specifically, we propose that dysfunction of mTOR, Rac1, and autophagy-related pathways not only hampers pathogen or nonorganic particle clearance but also participates in T cell and macrophage dysfunction, driving granuloma formation. This concept opens new avenues for potentially treating sarcoidosis and may serve as a blueprint for other granulomatous disorders.


Subject(s)
Autophagy , Sarcoidosis , TOR Serine-Threonine Kinases , rac1 GTP-Binding Protein , Autophagy/genetics , Humans , Macrophages/immunology , Macrophages/pathology , Sarcoidosis/genetics , Sarcoidosis/immunology , TOR Serine-Threonine Kinases/immunology , Th1 Cells/immunology , Th1 Cells/pathology , Th17 Cells/immunology , rac1 GTP-Binding Protein/immunology
3.
Front Immunol ; 11: 553994, 2020.
Article in English | MEDLINE | ID: mdl-33603730

ABSTRACT

Pathogenic intestinal bacteria lead to significant disease in humans. Here we investigated the role of the multifunctional protein, Apurinic/apyrimidinic endonuclease 1 (APE1), in regulating the internalization of bacteria into the intestinal epithelium. Intestinal tumor-cell lines and primary human epithelial cells were infected with Salmonella enterica serovar Typhimurium or adherent-invasive Escherichia coli. The effects of APE1 inhibition on bacterial internalization, the regulation of Rho GTPase Rac1 as well as the epithelial cell barrier function were assessed. Increased numbers of bacteria were present in APE1-deficient colonic tumor cell lines and primary epithelial cells. Activation of Rac1 was augmented following infection but negatively regulated by APE1. Pharmacological inhibition of Rac1 reversed the increase in intracellular bacteria in APE1-deficient cells whereas overexpression of constitutively active Rac1 augmented the numbers in APE1-competent cells. Enhanced numbers of intracellular bacteria resulted in the loss of barrier function and a delay in its recovery. Our data demonstrate that APE1 inhibits the internalization of invasive bacteria into human intestinal epithelial cells through its ability to negatively regulate Rac1. This activity also protects epithelial cell barrier function.


Subject(s)
Colon , DNA-(Apurinic or Apyrimidinic Site) Lyase/immunology , Epithelial Cells , Escherichia coli Infections , Escherichia coli/immunology , Intestinal Mucosa , Salmonella Infections , Salmonella typhimurium/immunology , rac1 GTP-Binding Protein/immunology , Colon/immunology , Colon/microbiology , Colon/pathology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Epithelial Cells/pathology , Escherichia coli Infections/immunology , Escherichia coli Infections/pathology , HT29 Cells , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Salmonella Infections/immunology , Salmonella Infections/pathology
4.
Fish Shellfish Immunol ; 91: 148-158, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31082520

ABSTRACT

The Rho family GTPase Rac1 acts as a molecular switch for signal transduction to regulate various cellular functions. Here, a Rac1 homolog (LcRac1) was identified in large yellow croaker (Larimichthys crocea), one of the most economically important marine fishes. The LcRac1 protein was expressed in Escherichia coli and purified. Subsequently the specific antibody was raised using the purified fusion protein (GST-LcRac1). LcRac1 was ubiquitously expressed in all 12 tissues we examined, with the highest expression in heart and blood and the weakest expression in head-kidney and spleen. Moreover, time course analysis revealed that LcRac1 expression was obviously up-regulated in liver, spleen and head-kidney after immunization with Poly I:C, LPS and Vibrio parahemolyticus. On the other hand, on the basis of protein interaction, it was found that the LcRac1 interacted with Tropomyosin, a crucial protein in the process of phagocytosis. Furthermore, RNAi assays indicated that the phagocytic percentage and phagocytic index were significantly decreased when the LcRac1 gene was silenced by sequence-specific siRNA. Fluorescence microscopy assays revealed FITC-labeled V. parahemolyticus were remarkably decreased after LcRac1 was silenced by sequence-specific siRNA at 24 h. These findings implicate the vital role of LcRac1 in innate immunity in the large yellow croaker.


Subject(s)
Fish Diseases/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Perciformes/genetics , Perciformes/immunology , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/immunology , Amino Acid Sequence , Animals , Base Sequence , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Profiling/veterinary , Lipopolysaccharides/pharmacology , Phagocytosis/genetics , Phylogeny , Poly I-C/pharmacology , Sequence Alignment/veterinary , Tropomyosin , Vibrio Infections/immunology , Vibrio Infections/veterinary , Vibrio parahaemolyticus/physiology , rac1 GTP-Binding Protein/chemistry
5.
J Immunol ; 202(9): 2529-2534, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30936294

ABSTRACT

Systemic lupus erythematosus severity correlates with elevated serum levels of type I IFNs, cytokines produced in large quantities by plasmacytoid dendritic cells (pDC) in response to engagement of TLR7 and TLR9 with endocytosed nucleic acids. B cell adaptor for PI3K (BCAP) promoted many aspects of TLR7-driven lupus-like disease, including Isg15 and Ifit1 expression in blood and an immature pDC phenotype associated with higher IFN production. BCAP-/- mice produced significantly less serum IFN-α than wild-type mice after injection of TLR9 agonist, and BCAP promoted TLR7 and TLR9-induced IFN-α production specifically in pDC. TLR-induced IFN-α production in pDC requires DOCK2-mediated activation of Rac1 leading to activation of IKKα, a mechanism we show was dependent on BCAP. BCAP-/- pDC had decreased actin polymerization and Rac1 activation and reduced IKKα phosphorylation upon TLR9 stimulation. We show a novel role for BCAP in promoting TLR-induced IFN-α production in pDC and in systemic lupus erythematosus pathogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Dendritic Cells/immunology , Interferon-alpha/immunology , Lupus Erythematosus, Systemic/immunology , Membrane Glycoproteins/immunology , Plasma Cells/immunology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 9/immunology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/pathology , Female , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/immunology , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/immunology , Interferon-alpha/genetics , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Male , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Neuropeptides/genetics , Neuropeptides/immunology , Plasma Cells/pathology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Toll-Like Receptor 7/genetics , Toll-Like Receptor 9/genetics , Ubiquitins/genetics , Ubiquitins/immunology , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/immunology
6.
Front Immunol ; 10: 415, 2019.
Article in English | MEDLINE | ID: mdl-30930895

ABSTRACT

Growing evidence indicate that large antigen-containing particles induce potent T cell-dependent high-affinity antibody responses. These responses require large particle internalization after recognition by the B cell receptor (BCR) on B cells. However, the molecular mechanisms governing BCR-mediated internalization remain unclear. Here we use a high-throughput quantitative image analysis approach to discriminate between B cell particle binding and internalization. We systematically show, using small molecule inhibitors, that human B cells require a SYK-dependent IgM-BCR signaling transduction via PI3K to efficiently internalize large anti-IgM-coated particles. IgM-BCR-mediated activation of PI3K involves both the adaptor protein NCK and the co-receptor CD19. Interestingly, we here reveal a strong NCK-dependence without profound requirement of the co-receptor CD19 in B cell responses to large particles. Furthermore, we demonstrate that the IgM-BCR/NCK signaling event facilitates RAC1 activation to promote actin cytoskeleton remodeling necessary for particle engulfment. Thus, we establish NCK/PI3K/RAC1 as an attractive IgM-BCR signaling axis for biological intervention to prevent undesired antibody responses to large particles.


Subject(s)
Antigen Presentation/immunology , B-Lymphocytes/immunology , Lymphocyte Activation/immunology , Phagocytosis/immunology , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/immunology , B-Lymphocytes/metabolism , Humans , Immunoglobulin M/immunology , Oncogene Proteins/immunology , Phosphatidylinositol 3-Kinases/immunology , Receptors, Antigen, B-Cell/immunology , rac1 GTP-Binding Protein/immunology
7.
Cell Rep ; 26(6): 1614-1626.e5, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30726742

ABSTRACT

ß2-integrins promote neutrophil recruitment to infected tissues and are crucial for host defense. Neutrophil recruitment is defective in leukocyte adhesion deficiency type-1 (LAD1), a condition caused by mutations in the CD18 (ß2-integrin) gene. Using a model of Citrobacter rodentium (CR)-induced colitis, we show that CD18-/- mice display increased intestinal damage and systemic bacterial burden, compared to littermate controls, ultimately succumbing to infection. This phenotype is not attributed to defective neutrophil recruitment, as it is shared by CXCR2-/- mice that survive CR infection. CR-infected CD18-/- mice feature prominent upregulation of IL-17 and downregulation of IL-22. Exogenous IL-22 administration, but not endogenous IL-17 neutralization, protects CD18-/- mice from lethal colitis. ß2-integrin expression on macrophages is mechanistically linked to Rac1/ROS-mediated induction of noncanonical-NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3) inflammasome-dependent IL-1ß production, which promotes ILC3-derived IL-22. Therefore, ß2-integrins are required for protective IL-1ß-dependent IL-22 responses in colitis, and the identified mechanism may underlie the association of human LAD1 with colitis.


Subject(s)
CD18 Antigens/genetics , Citrobacter rodentium/pathogenicity , Colitis/genetics , Enterobacteriaceae Infections/genetics , Interleukins/genetics , Macrophages/immunology , Animals , CD18 Antigens/deficiency , CD18 Antigens/immunology , Citrobacter rodentium/immunology , Colitis/immunology , Colitis/microbiology , Colitis/mortality , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/mortality , Female , Gene Expression Regulation/immunology , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukins/immunology , Intestines/immunology , Intestines/microbiology , Intestines/pathology , Macrophages/microbiology , Macrophages/pathology , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Neuropeptides/genetics , Neuropeptides/immunology , Neutrophil Infiltration , Neutrophils/immunology , Neutrophils/microbiology , Neutrophils/pathology , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Survival Analysis , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/immunology , Interleukin-22
8.
J Leukoc Biol ; 105(6): 1209-1224, 2019 06.
Article in English | MEDLINE | ID: mdl-30748033

ABSTRACT

Neutrophil chemotaxis is essential in responses to infection and underlies inflammation. In neutrophils, the small GTPase Rac1 has discrete functions at both the leading edge and in the retraction of the trailing structure at the cell's rear (uropod), but how Rac1 is regulated at the uropod is unknown. Here, we identified a mechanism mediated by the trafficking protein synaptotagmin-like 1 (SYTL1 or JFC1) that controls Rac1-GTP recycling from the uropod and promotes directional migration of neutrophils. JFC1-null neutrophils displayed defective polarization and impaired directional migration to N-formyl-methionine-leucyl-phenylalanine in vitro, but chemoattractant-induced actin remodeling, calcium signaling and Erk activation were normal in these cells. Defective chemotaxis was not explained by impaired azurophilic granule exocytosis associated with JFC1 deficiency. Mechanistically, we show that active Rac1 localizes at dynamic vesicles where endogenous JFC1 colocalizes with Rac1-GTP. Super-resolution microscopy (STORM) analysis shows adjacent distribution of JFC1 and Rac1-GTP, which increases upon activation. JFC1 interacts with Rac1-GTP in a Rab27a-independent manner to regulate Rac1-GTP trafficking. JFC1-null cells exhibited Rac1-GTP accumulation at the uropod and increased tail length, and Rac1-GTP uropod accumulation was recapitulated by inhibition of ROCK or by interference with microtubule remodeling. In vivo, neutrophil dynamic studies in mixed bone marrow chimeric mice show that JFC1-/- neutrophils are unable to move directionally toward the source of the chemoattractant, supporting the notion that JFC1 deficiency results in defective neutrophil migration. Our results suggest that defective Rac1-GTP recycling from the uropod affects directionality and highlight JFC1-mediated Rac1 trafficking as a potential target to regulate chemotaxis in inflammation and immunity.


Subject(s)
Chemotaxis/immunology , Guanosine Triphosphate/immunology , Membrane Proteins/immunology , Neuropeptides/immunology , Neutrophils/immunology , Pseudopodia/immunology , Vesicular Transport Proteins/immunology , rac1 GTP-Binding Protein/immunology , Animals , Chemotaxis/genetics , Guanosine Triphosphate/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Membrane Proteins/genetics , Mice , Mice, Knockout , Neuropeptides/genetics , Neutrophils/pathology , Pseudopodia/genetics , Pseudopodia/pathology , Vesicular Transport Proteins/genetics , rab27 GTP-Binding Proteins/genetics , rab27 GTP-Binding Proteins/immunology , rac1 GTP-Binding Protein/genetics
9.
Fish Shellfish Immunol ; 84: 998-1006, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30399403

ABSTRACT

Rac1 and Rac2, belonging to the small Rho GTPase family, play an important role during the immune responses. In this study, a Rac1 homolog (CsRac1) and a Rac2 homolog (CsRac2) were cloned from the Cynoglossus semilaevis. The full-length of CsRac1 and CsRac2 cDNA was 1219 bp and 1047 bp, respectively. Both CsRac1 and CsRac2 contain a 579 bp open reading frame (ORF) which encoding a 192 amino acids putative protein. The predicted molecular weight of CsRac1 and CsRac2 was 21.41 kDa and 21.35 kDa, and their theoretical pI was 8.50 and 7.91, respectively. Sequence analysis showed that the conserved RHO domain was detected both from amino acid of CsRac1 and CsRac2. Homologous analysis showed that CsRac1 and CsRac2 share high conservation with other counterparts from different species. The CsRac1 and CsRac2 transcript showed wide tissue distribution, in which CsRac1 and CsRac2 exhibit the highest expression level in liver and gill, respectively. The expression level of CsRac1 and CsRac2 fluctuated in the liver and gill tissues at different time points after challenged by Vibrio harveyi. Specifically, CsRac1 and CsRac2 were significantly up-regulated at 48 h and 96 h post injection. Moreover, the knocking down of CsRac1 and CsRac2 in cell line (TSHKC) reduced the expression of CsPAK1, CsIL1-ß and CsTNF-α. The present data suggests that CsRac1 and CsRac2 might play important roles in the innate immunity of half-smooth tongue sole.


Subject(s)
Fish Diseases/immunology , Fish Proteins/genetics , Fish Proteins/immunology , Flatfishes/genetics , Flatfishes/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Amino Acid Sequence , Animals , Base Sequence , Fish Proteins/chemistry , Gene Expression Profiling/veterinary , Phylogeny , Sequence Alignment/veterinary , Vibrio/physiology , Vibrio Infections/immunology , rac GTP-Binding Proteins/chemistry , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/immunology , rac1 GTP-Binding Protein/chemistry , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/immunology , RAC2 GTP-Binding Protein
10.
Cell Physiol Biochem ; 51(6): 2591-2603, 2018.
Article in English | MEDLINE | ID: mdl-30562745

ABSTRACT

BACKGROUND/AIMS: Chronic inflammation contributes to the development of type 2 diabetes mellitus by targeting the insulin receptor substrate protein-1 (IRS-1) signaling pathway. Previous studies showed that Leukemia related protein 16 (LRP16) reduced insulin stimulated glucose uptake in adipocytes by impairing the IRS-1 signaling pathway. We explored the mechanism by which LRP16 promotes the inflammatory response. METHODS: We screened LRP16 induced proteins in the lipopolysaccharide (LPS)-stimulated inflammatory response using liquid chromatography-mass spectrometry (LC-MS) and analyzed the potential biological functions of these proteins using online bioinformatics tools. mRNA expression and protein expression of target genes were measured by real time PCR and Western blot, respectively. RESULTS: A total of 390 differentially expressed proteins were identified. The mitogen-activated protein kinase (MAPK) signaling pathway was the primary activated pathway in LRP16-expressing cells. Overexpression of LRP16 activated ERK1/2 and Rac1, which are two key players related to the MAPK signaling pathway. Furthermore, knock down of endogenous LRP16 by RNA interference (RNAi) reduced Rac1 expression, ERK activation, and inflammatory cytokine expression in human adipocytes stimulated by LPS. The stimulatory effect of LRP16 was diminished by suppressing Rac1 expression and treating the cells with the ERK specific inhibitor, PD98059. CONCLUSION: These findings revealed the functions of LRP16 in promoting the inflammatory response through activating the Rac1-MAPK1/ERK pathway in human adipocytes.


Subject(s)
Inflammation/immunology , Lipopolysaccharides/immunology , MAP Kinase Signaling System , Neoplasm Proteins/immunology , Signal Transduction , rac1 GTP-Binding Protein/immunology , Adipocytes , Carboxylic Ester Hydrolases , Cell Line , Diabetes Mellitus, Type 2/immunology , Humans
11.
J Immunol ; 201(9): 2624-2640, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30282749

ABSTRACT

Endosomal traffic of TCR and signaling molecules regulates immunological synapse formation and T cell activation. We recently showed that Rab11 endosomes regulate the subcellular localization of the tyrosine kinase Lck and of the GTPase Rac1 and control their functions in TCR signaling and actin cytoskeleton remodeling. HIV-1 infection of T cells alters their endosomal traffic, activation capacity, and actin cytoskeleton organization. The viral protein Nef is pivotal for these modifications. We hypothesized that HIV-1 Nef could jointly alter Lck and Rac1 endosomal traffic and concomitantly modulate their functions. In this study, we show that HIV-1 infection of human T cells sequesters both Lck and Rac1 in a pericentrosomal compartment in an Nef-dependent manner. Strikingly, the Nef-induced Lck compartment contains signaling-competent forms (phosphorylated on key Tyr residues) of Lck and some of its downstream effectors, TCRζ, ZAP70, SLP76, and Vav1, avoiding the proximal LAT adaptor. Importantly, Nef-induced concentration of signaling molecules was concomitant with the upregulation of several early and late T cell activation genes. Moreover, preventing the concentration of the Nef-induced Lck compartment by depleting the Rab11 effector FIP3 counteracted Nef-induced gene expression upregulation. In addition, Nef extensively sequesters Rac1 and downregulates Rac1-dependent actin cytoskeleton remodeling, thus reducing T cell spreading. Therefore, by modifying their endosomal traffic, Nef hijacks signaling and actin cytoskeleton regulators to dually modulate their functional outputs. Our data shed new light into the molecular mechanisms that modify T cell physiology during HIV-1 infection.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV Infections/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , nef Gene Products, Human Immunodeficiency Virus/immunology , rac1 GTP-Binding Protein/metabolism , Actin Cytoskeleton/immunology , Actin Cytoskeleton/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Endosomes/immunology , Endosomes/metabolism , Endosomes/virology , HIV Infections/metabolism , Humans , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/immunology , Protein Transport/immunology , Signal Transduction/immunology , rac1 GTP-Binding Protein/immunology
12.
J Cell Mol Med ; 22(9): 4550-4554, 2018 09.
Article in English | MEDLINE | ID: mdl-29993181

ABSTRACT

Sphingosine-1-phosphate (S1P) is a crucial regulator in vascular inflammation. Our recent study found that under pathophysiological concentration in active anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV), S1P participated in MPO-ANCA-positive IgG-induced glomerular endothelial cell (GEnC) activation via a S1P receptor (S1PR)-dependent way. However, the downstream signalling pathways are not fully clear yet. In this study, we demonstrated that Rho guanosine triphosphatases (GTPases) signalling pathways, RhoA and Rac1 in particular, were implicated in MPO-ANCA-positive IgG-mediated GEnCs activation enhanced by pathophysiological concentration of S1P in AAV. These results provide mechanistic insights into vascular barrier dysfunction in AAV, which may facilitate the development of effective therapies.


Subject(s)
Antibodies, Antineutrophil Cytoplasmic/genetics , Endothelial Cells/drug effects , Immunoglobulin G/genetics , Lysophospholipids/pharmacology , Sphingosine/analogs & derivatives , rac1 GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/genetics , Antibodies, Antineutrophil Cytoplasmic/biosynthesis , Endothelial Cells/cytology , Endothelial Cells/immunology , Gene Expression Regulation , Humans , Immunoglobulin G/biosynthesis , Kidney Glomerulus/cytology , Kidney Glomerulus/immunology , Peroxidase/antagonists & inhibitors , Peroxidase/genetics , Peroxidase/immunology , Primary Cell Culture , Signal Transduction , Sphingosine/pharmacology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology , rac1 GTP-Binding Protein/immunology , rhoA GTP-Binding Protein/immunology
13.
Front Immunol ; 9: 679, 2018.
Article in English | MEDLINE | ID: mdl-29686674

ABSTRACT

Bacterial pathogens are associated with severe infections (e.g., sepsis) and exacerbation of debilitating conditions such as chronic obstructive pulmonary disease (COPD). The interactions of bacterial pathogens with macrophages, a key component of innate immunity and host defense, are not clearly understood and continue to be intensively studied. Having previously demonstrated a role of Wnt5A signaling in phagocytosis, we proceeded to decipher the connection of Wnt5A signaling with infection by pathogenic bacteria, namely Pseudomonas aeruginosa (PA) and Streptococcus pneumoniae (SP), which are related with the progression of COPD and sepsis. We found that during the initial hours of infection with PA and SP, there is decrease in the steady state levels of the Wnt5A protein in macrophages. Suppression of Wnt5A signaling, moreover, impairs macrophage clearance of the bacterial infection both in vitro and in vivo. Activation of Wnt5A signaling, on the other hand, enhances clearance of the infection. Macrophage-mediated containment of bacterial infection in our study is dependant on Wnt5A-induced Rac1/Disheveled activation and cytochalasin D inhibitable actin assembly, which is associated with ULK1 kinase activity and LC3BII accumulation. Our experimental findings are consistent with Wnt5A signaling-dependent induction of autophagic killing (xenophagy) of PA and SP, as further substantiated by transmission electron microscopy. Overall, our study unveils the prevalence of a Wnt5A-Rac1-Disheveled-mediated actin-associated autophagy circuit as an important component of innate immunity in host macrophages that may turn out crucial for restricting infection by leading bacterial pathogens.


Subject(s)
Dishevelled Proteins/immunology , Host-Pathogen Interactions/immunology , Neuropeptides/immunology , Pneumococcal Infections/immunology , Pseudomonas Infections/immunology , Wnt-5a Protein/immunology , rac1 GTP-Binding Protein/immunology , Animals , Cell Line , Macrophages/immunology , Mice, Inbred BALB C , Peritonitis/immunology , Pseudomonas aeruginosa , Respiratory Tract Infections/immunology , Streptococcus pneumoniae
14.
Cell Physiol Biochem ; 45(6): 2351-2368, 2018.
Article in English | MEDLINE | ID: mdl-29554649

ABSTRACT

BACKGROUND/AIMS: Recent researches highlighted the protective potential of pioglitazone, a PPAR-γ agonist, in the progression of cerebral ischemia-reperfusion injury. However, there has been no study on the application of pioglitazone in treating ischemic stroke through mechanisms involving pyroptosis. METHODS: The cerebral injury was established by middle cerebral artery occlusion (MCAO). in vitro ischemia in primary cultured astrocytes was induced by the oxygen-glucose deprivation (OGD). ELISA and Western Blot analysis were employed to the levels of PPAR-γ, pyroptosis-related biomarkers and cytoplasmic translocation of HMGB-1 and RAGE expression as well as Rac1 activity, respectively. RESULTS: We demonstrated that repeated intraperitoneal administration of pioglitazone remarkably reduced the infarct volume, improved neurological deficits and suppressed the Rac1 activity with significant reduction of excessive ROS in rat model of middle cerebral artery occlusion (MCAO). Moreover, pioglitazone alleviated the up-regulation of pyroptosis-related biomarkers and the increased cytoplasmic translocation of HMGB-1 and RAGE expression in cerebral penumbra cortex. Similarly, the protective effects of pioglitazone on cultured astrocytes were characterized by reduced Rac1 activity, pyroptosis related protein expressions and lactate dehydrogenase (LDH) release. However, these protective effects of pioglitazone were neutralized with the use of GW9662, a PPAR-γ inhibitor. Interestingly, Rac1 knockdown in lentivirus with the Rac1 small hair RNA (shRNA) could inhibit the OGD-induced pyroptosis of primary cultured astrocytes. Furthermore, the combination of Rac1-shRNA and pioglitazone can further strengthen the inhibitory effects on pyroptosis induced by OGD. CONCLUSION: The neuroprotection of pioglitazone was attributable to the alleviated ischemia/hypoxia-induced pyroptosis and was also associated with the PPARγ-mediated suppression of HGMB-1/RAGE signaling pathway. Moreover, the inhibition of Rac1 promoted this function.


Subject(s)
Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/therapeutic use , Pyroptosis/drug effects , Signal Transduction/drug effects , Thiazolidinediones/therapeutic use , Animals , Astrocytes/drug effects , Astrocytes/immunology , Astrocytes/pathology , Cells, Cultured , Glucose/immunology , HMGB1 Protein/immunology , Infarction, Middle Cerebral Artery/immunology , Infarction, Middle Cerebral Artery/pathology , Male , Neuroprotection/drug effects , PPAR gamma/immunology , Pioglitazone , Rats, Sprague-Dawley , Reactive Oxygen Species/immunology , Receptor for Advanced Glycation End Products/immunology , Reperfusion Injury/drug therapy , Reperfusion Injury/immunology , Reperfusion Injury/pathology , rac1 GTP-Binding Protein/immunology
15.
ChemMedChem ; 13(7): 754-761, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29359495

ABSTRACT

Cytotoxic necrotizing factor 1 (CNF1) is a toxin produced by pathogenic strains of Escherichia coli responsible for extra-intestinal infections. CNF1 deamidates Rac1, thereby triggering its permanent activation and worsening inflammatory reactions. Activated Rac1 is prone to proteasomal degradation. There is no targeted therapy against CNF1, despite its clinical relevance. In this work we developed a fluorescent cell-based immunoassay to screen for inhibitors of CNF1-induced Rac1 degradation among 1120 mostly approved drugs. Eleven compounds were found to prevent CNF1-induced Rac1 degradation, and five also showed a protective effect against CNF1-induced multinucleation. Finally, lasalocid, monensin, bepridil, and amodiaquine protected cells from both diphtheria toxin and CNF1 challenges. These data highlight the potential for drug repurposing to fight several bacterial infections and Rac1-based diseases.


Subject(s)
Bacterial Toxins/antagonists & inhibitors , Escherichia coli Proteins/antagonists & inhibitors , Small Molecule Libraries/pharmacology , rac1 GTP-Binding Protein/metabolism , Amodiaquine/pharmacology , Bacterial Toxins/adverse effects , Bacterial Toxins/metabolism , Bepridil/pharmacology , Diphtheria Toxin/adverse effects , Drug Repositioning , Escherichia coli/chemistry , Escherichia coli Proteins/adverse effects , Escherichia coli Proteins/metabolism , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Immunoassay , Lasalocid/pharmacology , Monensin/pharmacology , rac1 GTP-Binding Protein/chemistry , rac1 GTP-Binding Protein/immunology
16.
Biochem Biophys Res Commun ; 495(3): 2209-2213, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29269293

ABSTRACT

Various physiologically active effects of polymerized polyphenols have been reported. In this study, we synthesized a polymerized polyphenol (mL2a-pCA) by polymerizing caffeic acid using mutant Agaricus brasiliensis laccase and analyzed its physiological activity and mechanism of action. We found that mL2a-pCA induced morphological changes and the production of cytokines and chemokines in C3H/HeN mouse-derived resident peritoneal macrophages in vitro. The mechanisms of action of polymerized polyphenols on in vitro mouse resident peritoneal cells have not been characterized in detail previously. Herein, we report that the mL2a-pCA-induced production of interleukin-6 (IL-6) and monocyte chemotactic protein-1 (MCP-1) in C3H/HeN mouse-derived resident peritoneal cells was inhibited by treatment with the Rac1 inhibitor NSC23766 trihydrochloride. In addition, we found that mL2a-pCA activated the phosphorylation Rac1. Taken together, the results show that mL2a-pCA induced macrophage activation via Rac1 phosphorylation-dependent pathways.


Subject(s)
Laccase/chemistry , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/pathology , Neuropeptides/immunology , Polyphenols/administration & dosage , Polyphenols/chemistry , rac1 GTP-Binding Protein/immunology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Activation , Macrophage Activation/drug effects , Macrophages/drug effects , Male , Mice , Mice, Inbred C3H , Phosphorylation/drug effects
17.
J Immunol ; 199(9): 3176-3186, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28947541

ABSTRACT

In elderly patients, bacterial infection often causes severe complications and sepsis. Compared to younger patients, older patients are more susceptible to sepsis caused by respiratory infection. Macrophage (Mϕ) phagocytosis of bacteria plays a critical role in the clearance of pathogens and the initiation of immune responses. It has been suggested that Mϕ exhibit age-related functional alterations, including reduced chemotaxis, phagocytosis, antibacterial defense, and the ability to generate reactive oxygen species. However, the mechanisms behind these changes remain unclear. The present study sought to determine changes in bacterial phagocytosis in aging alveolar Mϕ (AMϕ) and the underlying mechanisms. We show that bacteria initiate cytoskeleton remodeling in AMϕ through interaction with macrophage receptor with collagenous structure (MARCO), a bacterial scavenger receptor. This remodeling, in turn, promotes enhanced cell surface expression of MARCO and bacterial phagocytosis. We further demonstrate that Rac1-GTP mediates MARCO signaling and activates actin-related protein-2/3 complex, an F-actin nucleator, thereby inducing F-actin polymerization, filopodia formation, and increased cell surface expression of MARCO, all of which are essential for the execution of bacteria phagocytosis. However, AMϕ isolated from aging mice exhibit suppressed Rac1 mRNA and protein expression, which resulted in decreases in Rac1-GTP levels and actin-related protein-2/3 activation, as well as subsequent attenuation of F-actin polymerization, filopodia formation, and cell surface expression of MARCO. As a result, bacterial phagocytosis in aging AMϕ is decreased. This study highlights a previously unidentified mechanism by which aging impairs Mϕ phagocytosis of bacteria. Targeting these pathways may improve outcomes of bacterial infection in elderly patients.


Subject(s)
Actin Cytoskeleton/immunology , Aging/immunology , Escherichia coli K12/immunology , Macrophages, Alveolar/immunology , Phagocytosis/physiology , Actin Cytoskeleton/genetics , Aging/genetics , Animals , Humans , Male , Mice , Mice, Knockout , Neuropeptides/genetics , Neuropeptides/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/immunology
18.
Mol Med Rep ; 16(5): 6346-6353, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28849230

ABSTRACT

The endogenously activated rheumatoid arthritis (RA) synovial fibroblasts (RSFs) are likely to be the key to curing the disease. RSFs express Toll­like receptors (TLRs) rendering them prone to activation by exogenous and endogenous TLR ligands, resulting in the production of chemokines and cytokines Germline deletion of tumor necrosis factor­α­induced protein­8 like 2 (TIPE2, also known as TNFAIP8L2) results in fatal inflammation and hypersensitivity to TLR and T cell receptor stimulation. The present study demonstrates an inverse association between TIPE2 and cytokine gene expression in RSFs following lipopolysaccharide (LPS) stimulation. Enhanced TIPE2 expression decreased Ras­related C3 botulinum toxin substrate (Rac) activation and interferon regulatory factor 3 phosphorylation, and phosphoinositide 3­kinase and Rac inhibition significantly diminished LPS­induced cytokine gene expression in RSFs. In conclusion, the findings of the present study demonstrate that TIPE2 serves a negative role in activating the Rac signaling pathway and in the initiation of the immune response by decreasing the activity of proinflammatory cytokines. These results may be useful in designing novel strategies for the prevention and treatment of RA.


Subject(s)
Arthritis, Experimental/genetics , Fibroblasts/immunology , Interferon Regulatory Factor-3/genetics , Intracellular Signaling Peptides and Proteins/genetics , Synoviocytes/immunology , rac1 GTP-Binding Protein/genetics , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Cell Line , Fibroblasts/pathology , Gene Expression Regulation , Interferon Regulatory Factor-3/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-1/genetics , Interleukin-1/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Intracellular Signaling Peptides and Proteins/immunology , Lipopolysaccharides , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , Phosphorylation , Rats , Signal Transduction , Synovial Membrane/immunology , Synovial Membrane/pathology , Synoviocytes/pathology , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , rac1 GTP-Binding Protein/immunology
19.
Biochem Biophys Res Commun ; 490(4): 1274-1281, 2017 09 02.
Article in English | MEDLINE | ID: mdl-28687489

ABSTRACT

Enhanced turnover of subchondral trabecular bone is a hallmark of rheumatoid arthritis (RA) and it results from an imbalance between bone resorption and bone formation activities. To investigate the formation and activation of osteoclasts which mediate bone resorption, a Fas-deficient MRL/lpr mouse model which spontaneously develops autoimmune arthritis and exhibits decreased bone mass was studied. Various assays were performed on subchondral trabecular bone of the temporomandibular joint (TMJ) from MRL/lpr mice and MRL+/+ mice. Initially, greater osteoclast production was observed in vitro from bone marrow macrophages obtained from MRL/lpr mice due to enhanced phosphorylation of NF-κB, as well as Akt and MAPK, to receptor activator of nuclear factor-κB ligand (RANKL). Expression of sphingosine 1-phosphate receptor 1 (S1P1) was also significantly upregulated in the condylar cartilage. S1P1 was found to be required for S1P-induced migration of osteoclast precursor cells and downstream signaling via Rac1. When SN50, a synthetic NF-κB-inhibitory peptide, was applied to the MRL/lpr mice, subchondral trabecular bone loss was reduced and both production of osteoclastogenesis markers and sphingosine kinase (Sphk) 1/S1P1 signaling were reduced. Thus, the present results suggest that Fas/S1P1 signaling via activation of NF-κB in osteoclast precursor cells is a key factor in the pathogenesis of RA in the TMJ.


Subject(s)
Arthritis, Rheumatoid/immunology , Bone Resorption/immunology , NF-kappa B/immunology , Osteoclasts/drug effects , Receptors, Lysosphingolipid/immunology , Temporomandibular Joint/immunology , fas Receptor/immunology , Animals , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Autoimmunity , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Bone Resorption/genetics , Bone Resorption/pathology , Bone Resorption/prevention & control , Cell Differentiation , Disease Models, Animal , Female , Gene Expression Regulation , Lysophospholipids/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred MRL lpr , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/immunology , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Neuropeptides/genetics , Neuropeptides/immunology , Osteoclasts/immunology , Osteoclasts/pathology , Osteogenesis/drug effects , Osteogenesis/immunology , Peptides/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/immunology , Primary Cell Culture , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , RANK Ligand/genetics , RANK Ligand/immunology , Receptors, Lysosphingolipid/genetics , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/immunology , Temporomandibular Joint/drug effects , Temporomandibular Joint/pathology , fas Receptor/genetics , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/immunology
20.
J Cell Sci ; 130(15): 2530-2540, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28600322

ABSTRACT

Lamellipodia are sheet-like cell protrusions driven by actin polymerization mainly through Rac1, a GTPase molecular switch. In Fcγ receptor-mediated phagocytosis of IgG-opsonized erythrocytes (IgG-Es), Rac1 activation is required for lamellipodial extension along the surface of IgG-Es. However, the significance of Rac1 deactivation in phagosome formation is poorly understood. Our live-cell imaging and electron microscopy revealed that RAW264 macrophages expressing a constitutively active Rac1 mutant showed defects in phagocytic cup formation, while lamellipodia were formed around IgG-Es. Because activated Rac1 reduced the phosphorylation levels of myosin light chains, failure of the cup formation is probably due to inhibition of actin/myosin II contractility. Reversible photo-manipulation of the Rac1 switch in macrophages fed with IgG-Es could phenocopy two lamellipodial motilities: outward-extension and cup-constriction by Rac1 ON and OFF, respectively. In conjunction with fluorescence resonance energy transfer imaging of Rac1 activity, we provide a novel mechanistic model of phagosome formation spatiotemporally controlled by Rac1 switching within a phagocytic cup.


Subject(s)
Macrophages/immunology , Models, Immunological , Neuropeptides/immunology , Phagosomes/immunology , Receptors, IgG/immunology , rac1 GTP-Binding Protein/immunology , Animals , Mice , Phosphorylation/immunology , RAW 264.7 Cells
SELECTION OF CITATIONS
SEARCH DETAIL