Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Infect Immun ; 84(6): 1796-1805, 2016 06.
Article in English | MEDLINE | ID: mdl-27045038

ABSTRACT

Tick saliva contains a number of effector molecules that inhibit host immunity and facilitate pathogen transmission. How tick proteins regulate immune signaling, however, is incompletely understood. Here, we describe that loop 2 of sialostatin L2, an anti-inflammatory tick protein, binds to annexin A2 and impairs the formation of the NLRC4 inflammasome during infection with the rickettsial agent Anaplasma phagocytophilum Macrophages deficient in annexin A2 secreted significantly smaller amounts of interleukin-1ß (IL-1ß) and IL-18 and had a defect in NLRC4 inflammasome oligomerization and caspase-1 activation. Accordingly, Annexin a2-deficient mice were more susceptible to A. phagocytophilum infection and showed splenomegaly, thrombocytopenia, and monocytopenia. Providing translational support to our findings, better binding of annexin A2 to sialostatin L2 in sera from 21 out of 23 infected patients than in sera from control individuals was also demonstrated. Overall, we establish a unique mode of inflammasome evasion by a pathogen, centered on a blood-feeding arthropod.


Subject(s)
Anaplasma phagocytophilum/immunology , Annexin A2/immunology , Apoptosis Regulatory Proteins/immunology , Calcium-Binding Proteins/immunology , Cystatins/immunology , Ehrlichiosis/microbiology , Immune Evasion , Amino Acid Sequence , Anaplasma phagocytophilum/genetics , Animals , Annexin A2/chemistry , Annexin A2/genetics , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Arachnid Vectors/chemistry , Arachnid Vectors/genetics , Arachnid Vectors/immunology , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/genetics , Caspase 1/genetics , Caspase 1/immunology , Caspases/genetics , Caspases/immunology , Caspases, Initiator , Cystatins/chemistry , Cystatins/genetics , Ehrlichiosis/immunology , Ehrlichiosis/pathology , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation , Humans , Inflammasomes/genetics , Inflammasomes/immunology , Interleukin-18/genetics , Interleukin-18/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Ixodes/chemistry , Ixodes/genetics , Ixodes/immunology , Macrophages/immunology , Macrophages/microbiology , Mice , Models, Molecular , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Signal Transduction
2.
Infect Immun ; 82(6): 2553-64, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24686067

ABSTRACT

Saliva from arthropod vectors facilitates blood feeding by altering host inflammation. Whether arthropod saliva counters inflammasome signaling, a protein scaffold that regulates the activity of caspase-1 and cleavage of interleukin-1ß (IL-1ß) and IL-18 into mature molecules, remains elusive. In this study, we provide evidence that a tick salivary protein, sialostatin L2, inhibits inflammasome formation during pathogen infection. We show that sialostatin L2 targets caspase-1 activity during host stimulation with the rickettsial agent Anaplasma phagocytophilum. A. phagocytophilum causes macrophage activation and hemophagocytic syndrome features. The effect of sialostatin L2 in macrophages was not due to direct caspase-1 enzymatic inhibition, and it did not rely on nuclear factor κB or cathepsin L signaling. Reactive oxygen species from NADPH oxidase and the Loop2 domain of sialostatin L2 were important for the regulatory process. Altogether, our data expand the knowledge of immunoregulatory pathways of tick salivary proteins and unveil an important finding in inflammasome biology.


Subject(s)
Anaplasma phagocytophilum/physiology , Caspase 1/metabolism , Ehrlichiosis/microbiology , Salivary Cystatins/physiology , Analysis of Variance , Animals , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Ehrlichiosis/metabolism , Ehrlichiosis/pathology , Inflammasomes/metabolism , Inflammation/physiopathology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Reactive Oxygen Species
3.
Cell Microbiol ; 15(7): 1070-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23433059

ABSTRACT

Ubiquitination (ubiquitylation) is a common protein modification that regulates a multitude of processes within the cell. This modification is typically accomplished through the covalent binding of ubiquitin to a lysine residue onto a target protein and is catalysed by the presence of three enzymes: an activating enzyme (E1), ubiquitin-conjugating enzyme (E2) and ubiquitin-protein ligase (E3). In recent years, ubiquitination has risen as a major signalling regulator of immunity and microbial pathogenesis in the mammalian system. Still, little is known about how ubiquitin relates specifically to vector immunology. Here, we provide a brief overview of ubiquitin biochemistry and describe how ubiquitination regulates immune responses in arthropods of medical relevance. We also discuss scientific gaps in the literature and suggest that, similar to mammals, ubiquitin is a major regulator of immunity in medically important arthropods.


Subject(s)
Mammals/immunology , Protein Processing, Post-Translational , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Arthropods/immunology , Humans , Ubiquitination
4.
J Infect Dis ; 208(11): 1830-40, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-23901084

ABSTRACT

Ubiquitination is a posttranslational modification that regulates protein degradation and signaling in eukaryotes. Although it is acknowledged that pathogens exploit ubiquitination to infect mammalian cells, it remains unknown how microbes interact with the ubiquitination machinery in medically relevant arthropods. Here, we show that the ubiquitination machinery is present in the tick Ixodes scapularis and demonstrate that the E3 ubiquitin ligase named x-linked inhibitor of apoptosis protein (XIAP) restricts bacterial colonization of this arthropod vector. We provide evidence that xiap silencing significantly increases tick colonization by the bacterium Anaplasma phagocytophilum, the causative agent of human granulocytic anaplasmosis. We also demonstrate that (i) XIAP polyubiquitination is dependent on the really interesting new gene (RING) catalytic domain, (ii) XIAP polyubiquitination occurs via lysine (K)-63 but not K-48 residues, and (iii) XIAP-dependent K-63 polyubiquitination requires zinc for catalysis. Taken together, our data define a role for ubiquitination during bacterial colonization of disease vectors.


Subject(s)
Anaplasma phagocytophilum/physiology , Arachnid Vectors/enzymology , Ehrlichiosis/microbiology , Ixodes/enzymology , Ubiquitin-Protein Ligases/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , Animals , Arachnid Vectors/microbiology , Catalytic Domain , Humans , Ixodes/microbiology , RNA Interference , Signal Transduction , Ubiquitin-Protein Ligases/genetics , Ubiquitination , X-Linked Inhibitor of Apoptosis Protein/genetics
5.
Infect Immun ; 80(9): 3194-205, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22753375

ABSTRACT

Anaplasma phagocytophilum is a tick-borne rickettsial pathogen that provokes an acute inflammatory response during mammalian infection. The illness caused by A. phagocytophilum, human granulocytic anaplasmosis, occurs irrespective of pathogen load and results instead from host-derived immunopathology. Thus, characterizing A. phagocytophilum genes that affect the inflammatory process is critical for understanding disease etiology. By using an A. phagocytophilum Himar1 transposon mutant library, we showed that a single transposon insertion into the A. phagocytophilum dihydrolipoamide dehydrogenase 1 gene (lpda1 [APH_0065]) affects inflammation during infection. A. phagocytophilum lacking lpda1 revealed enlargement of the spleen, increased splenic extramedullary hematopoiesis, and altered clinicopathological abnormalities during mammalian colonization. Furthermore, LPDA1-derived immunopathology was independent of neutrophil infection and correlated with enhanced reactive oxygen species from NADPH oxidase and nuclear factor (NF)-κB signaling in macrophages. Taken together, these findings suggest the presence of different signaling pathways in neutrophils and macrophages during A. phagocytophilum invasion and highlight the importance of LPDA1 as an immunopathological molecule.


Subject(s)
Anaplasma phagocytophilum/enzymology , Dihydrolipoamide Dehydrogenase/immunology , Dihydrolipoamide Dehydrogenase/metabolism , Ehrlichiosis/immunology , Ehrlichiosis/pathology , Virulence Factors/immunology , Virulence Factors/metabolism , Adult , Anaplasma phagocytophilum/immunology , Anaplasma phagocytophilum/pathogenicity , Animals , Ehrlichiosis/microbiology , Female , Gene Deletion , Humans , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mutagenesis, Insertional , Neutrophils/immunology , Neutrophils/microbiology , Spleen/microbiology , Spleen/pathology
6.
Tissue Barriers ; 3(1-2): e1004975, 2015.
Article in English | MEDLINE | ID: mdl-25838974

ABSTRACT

M cells are a subset of mucosal epithelial cells with specialized capability to transport antigens across the mucosal barrier, but there is limited information on antigen transfer in the subepithelial zone due to the challenges in tracking microparticles and antigens that are transcytosed by this unique cell. Using transgenic reporter mice expressing dsRed in the cytoplasm of M cells and EGFP in myeloid cells, we observed that the M cell basolateral pocket hosts a close interaction between B lymphocytes and dendritic cells. Interestingly, we identified a population of previously undescribed M cell-derived vesicles (MCM) that are constitutively shed into the subepithelial space and readily taken up by CX3CR1(+)CD11b(+) CD11c(+) dendritic cells. These MCM are characterized by their cytoplasmic dsRed confirming their origin from the M cell cytoplasm. MCM showed preferential colocalization in dendritic cells with transcytosed bacteria but not transcytosed polystyrene beads, indicating a selective sorting of cargo fate in the subepithelial zone. The size and number of MCM were found to be upregulated by bacterial transcytosis and soluble toll-like receptor 2 (TLR2) agonist, further pointing to dynamic regulation of this mechanism. These results suggest that MCM provide a unique function by delivering to dendritic cells, various materials such as M cell-derived proteins, effector proteins, toxins, and particles found in the M cell cytoplasm during infection or surveillance.

7.
Front Microbiol ; 4: 308, 2013.
Article in English | MEDLINE | ID: mdl-24155744

ABSTRACT

Arthropod saliva possesses anti-hemostatic, anesthetic, and anti-inflammatory properties that facilitate feeding and, inadvertently, dissemination of pathogens. Vector-borne diseases caused by these pathogens affect millions of people each year. Many studies address the impact of arthropod salivary proteins on various immunological components. However, whether and how arthropod saliva counters Nod-like (NLR) sensing remains elusive. NLRs are innate immune pattern recognition molecules involved in detecting microbial molecules and danger signals. Nod1/2 signaling results in activation of the nuclear factor-κB and the mitogen-activated protein kinase pathways. Caspase-1 NLRs regulate the inflammasome~- a protein scaffold that governs the maturation of interleukin (IL)-1ß and IL-18. Recently, several vector-borne pathogens have been shown to induce NLR activation in immune cells. Here, we provide a brief overview of NLR signaling and discuss clinically relevant vector-borne pathogens recognized by NLR pathways. We also elaborate on possible anti-inflammatory effects of arthropod saliva on NLR signaling and microbial pathogenesis for the purpose of exchanging research perspectives.

8.
PLoS One ; 8(9): e75911, 2013.
Article in English | MEDLINE | ID: mdl-24058709

ABSTRACT

Mammals and plants share pathogen-sensing systems named nod-like receptors (NLRs). Some NLRs form the inflammasome, a protein scaffold that regulates the secretion of interleukin (IL)-1ß and IL-18 by cleaving catalytically inactive substrates into mature cytokines. Here, we show an immune conservation between plant and mammalian NLRs and demonstrate that the murine nuclear receptor binding SET domain protein 1 (NSD1), a protein that bears similarity to the NLR regulator enhanced downy mildew 2 (EDM2) in Arabidopsis, diminishes caspase-1 activity during extracellular stimulation with Listeria monocytogenes listeriolysin O (LLO). EDM2 is known to regulate plant developmental processes, whereas NSD1 is associated with developmental disorders. We observed that NSD1 neither affects nuclear factor (NF)-κB signaling nor regulates NLRP3 inflammasome gene expression at the chromatin, transcriptional or translational level during LLO stimulation of macrophages. Silencing of Nsd1 followed by LLO stimulation led to increased caspase-1 activation, enhanced post-translational maturation of IL-1ß and IL-18 and elevated pyroptosis, a form of cell death associated with inflammation. Furthermore, treatment of macrophages with LLO(W492A), which lacks hemolytic activity due to a tryptophan to alanine substitution in the undecapeptide motif, indicates the importance of functional LLO for NSD1 regulation of the NLRP3 inflammasome. Taken together, our results indicate that NLR signaling in plants may be used for gene discovery in mammals.


Subject(s)
Bacterial Toxins/pharmacology , Carrier Proteins/metabolism , Caspase 1/metabolism , Heat-Shock Proteins/pharmacology , Hemolysin Proteins/pharmacology , Macrophages/metabolism , Nuclear Proteins/metabolism , Signal Transduction/drug effects , Animals , Carrier Proteins/genetics , Caspase 1/genetics , Cell Death/drug effects , Cell Death/genetics , Cell Line , Enzyme Activation/drug effects , Enzyme Activation/genetics , Histone-Lysine N-Methyltransferase , Inflammasomes/genetics , Inflammasomes/metabolism , Interleukin-18/genetics , Interleukin-18/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Listeria monocytogenes , Macrophages/pathology , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Nuclear Proteins/genetics , Signal Transduction/genetics
9.
Parasit Vectors ; 5: 229, 2012 Oct 10.
Article in English | MEDLINE | ID: mdl-23050849

ABSTRACT

BACKGROUND: Ixodes scapularis saliva enables the transmission of infectious agents to the mammalian host due to its immunomodulatory, anesthetic and anti-coagulant properties. However, how I. scapularis saliva influences host cytokine secretion in the presence of the obligate intracellular rickettsial pathogen Anaplasma phagocytophilum remains elusive. METHODS: Bone marrow derived macrophages (BMDMs) were stimulated with pathogen associated molecular patterns (PAMPs) and A. phagocytophilum. Cytokine secretion was measured in the presence and absence of I. scapularis saliva. Human peripheral blood mononuclear cells (PBMCs) were also stimulated with Tumor Necrosis Factor (TNF)-α in the presence and absence of I. scapularis saliva and interleukin (IL)-8 was measured. RESULTS: I. scapularis saliva inhibits inflammatory cytokine secretion by macrophages during stimulation of Toll-like (TLR) and Nod-like receptor (NLR) signaling pathways. The effect of I. scapularis saliva on immune cells is not restricted to murine macrophages because decreasing levels of interleukin (IL)-8 were observed after TNF-α stimulation of human peripheral blood mononuclear cells. I. scapularis saliva also mitigates pro-inflammatory cytokine response by murine macrophages during challenge with A. phagocytophilum. CONCLUSIONS: These findings suggest that I. scapularis may inhibit inflammatory cytokine secretion during rickettsial transmission at the vector-host interface.


Subject(s)
Anaplasma phagocytophilum/immunology , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Ixodes/immunology , Saliva/immunology , Adult , Animals , Humans , Immunologic Factors/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL