Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Insect Mol Biol ; 32(4): 329-339, 2023 08.
Article in English | MEDLINE | ID: mdl-36680546

ABSTRACT

Ticks are important vectors of pathogenic viruses, bacteria, and protozoans to humans, wildlife, and domestic animals. Due to their life cycles, ticks face significant challenges related to water homeostasis. When blood-feeding, they must excrete water and ions, but when off-host (for stretches lasting several months), they must conserve water to avoid desiccation. Aquaporins (AQPs), a family of membrane-bound water channels, are key players in osmoregulation in many animals but remain poorly characterized in ticks. Here, we bioinformatically identified AQP-like genes from the deer tick Ixodes scapularis and used phylogenetic approaches to map the evolution of the aquaporin gene family in arthropods. Most arachnid AQP-like sequences (including those of I. scapularis) formed a monophyletic group clustered within aquaglycerolporins (GLPs) from bacteria to vertebrates. This gene family is absent from insects, revealing divergent evolutionary paths for AQPs in different hematophagous arthropods. Next, we sequenced the full-length cDNA of I. scapularis aquaporin 1 (IsAQP1) and expressed it heterologously in Xenopus oocytes to functionally characterize its permeability to water and solutes. Additionally, we examined IsAQP1 expression across different life stages and adult female organs. We found IsAQP1 is an efficient water channel with high expression in salivary glands prior to feeding, suggesting it plays a role in osmoregulation before or during blood feeding. Its functional properties are unique: unlike most GLPs, IsAQP1 has low glycerol permeability, and unlike most AQPs, it is insensitive to mercury. Together, our results suggest IsAQP1 plays an important role in tick water balance physiology and that it may hold promise as a target of novel vector control efforts.


Subject(s)
Ixodes , Lyme Disease , Humans , Female , Animals , Ixodes/genetics , Ixodes/microbiology , Aquaporin 1/genetics , Aquaporin 1/metabolism , Phylogeny , Bacteria , Water/metabolism , Disease Vectors
2.
Cell Microbiol ; 23(2): e13275, 2021 02.
Article in English | MEDLINE | ID: mdl-33006213

ABSTRACT

The peritrophic matrix (PM) is an acellular membrane that covers the gut epithelium in arthropods and physically separates it from the lumen. The structure is thought to play an important role in tick biology. The PM is also known to impact the persistence of tick-borne pathogens like Borrelia burgdorferi, although limited information is available about its molecular constituents or their biological significance. Herein, we characterise a novel PM-associated gut protein in Ixodes scapularis ticks, annotated as Peritrophic Membrane Chitin Binding Protein (PM_CBP), for its role in the integrity and function of the matrix. The PM_CBP displays homology to the chitin deacetylase metalloenzyme, shows upregulation during tick feeding, and is localized at the luminal surface of the gut epithelium. The structural integrity of the PM was impaired both by the knock down of PM_CBP expression via RNA interference and by treatment with anti-PM_CBP antibodies, as revealed by its electron microscopic appearance. Additionally, the duration of tick engorgement on mice and the passage of experimentally-inoculated fluorescent dextran molecules across the PM are affected by the knock down of PM_CBP expression. The transfer of anti-PM_CBP antibodies into the tick gut impacted the overall composition of the resident microbiome, and also influenced B. burgdorferi acquisition in ticks and its transmission to mice. Taken together, these data highlight the biological significance of the Ixodes PM and suggest that the targeting of its molecular constituents may contribute to the development of novel interventions against tick-borne infections.


Subject(s)
Arthropod Proteins/metabolism , Borrelia burgdorferi/physiology , Gastrointestinal Microbiome , Host-Pathogen Interactions , Ixodes/metabolism , Ixodes/microbiology , Lyme Disease/microbiology , Animals , Borrelia burgdorferi/pathogenicity , Carrier Proteins/metabolism , Chitin/metabolism , DNA, Bacterial , Female , Gene Knockdown Techniques , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C3H , Protein Binding , RNA Interference , RNA, Ribosomal, 16S
3.
Proc Natl Acad Sci U S A ; 115(16): E3788-E3797, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29610317

ABSTRACT

Borrelia burgdorferi is one of the few extracellular pathogens capable of establishing persistent infection in mammals. The mechanisms that sustain long-term survival of this bacterium are largely unknown. Here we report a unique innate immune evasion strategy of B. burgdorferi, orchestrated by a surface protein annotated as BBA57, through its modulation of multiple spirochete virulent determinants. BBA57 function is critical for early infection but largely redundant for later stages of spirochetal persistence, either in mammals or in ticks. The protein influences host IFN responses as well as suppresses multiple host microbicidal activities involving serum complement, neutrophils, and antimicrobial peptides. We also discovered a remarkable plasticity in BBA57-mediated spirochete immune evasion strategy because its loss, although resulting in near clearance of pathogens at the inoculum site, triggers nonheritable adaptive changes that exclude detectable nucleotide alterations in the genome but incorporate transcriptional reprograming events. Understanding the malleability in spirochetal immune evasion mechanisms that ensures their host persistence is critical for the development of novel therapeutic and preventive approaches to combat long-term infections like Lyme borreliosis.


Subject(s)
Bacterial Proteins/physiology , Borrelia burgdorferi/immunology , Immune Evasion , Lipoproteins/physiology , Membrane Proteins/physiology , Animals , Antigens, Bacterial/immunology , Antimicrobial Cationic Peptides/biosynthesis , Antimicrobial Cationic Peptides/genetics , Arachnid Vectors/microbiology , Bacterial Proteins/genetics , Borrelia burgdorferi/genetics , Borrelia burgdorferi/pathogenicity , Cells, Cultured , Complement System Proteins/immunology , Cytokines/biosynthesis , Cytokines/genetics , Female , Gene Expression Regulation, Bacterial , Humans , Ixodes/microbiology , Lipoproteins/genetics , Lyme Disease/immunology , Lyme Disease/microbiology , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, SCID , Specific Pathogen-Free Organisms , Virulence
4.
Cell Microbiol ; 21(2): e12885, 2019 02.
Article in English | MEDLINE | ID: mdl-29934966

ABSTRACT

Borrelia burgdorferi is the causative agent of Lyme disease that persists in a complex enzootic life cycle, involving Ixodes ticks and vertebrate hosts. The microbe invades ticks and vertebrate hosts in spite of active immune surveillance and potent microbicidal responses, and establishes long-term infection utilising mechanisms that are yet to be unravelled. The pathogen can cause multi-system disorders when transmitted to susceptible mammalian hosts, including in humans. In the past decades, several studies identified a limited number of B. burgdorferi gene-products critical for pathogen persistence, transmission between the vectors and the host, and host-pathogen interactions. This review will focus on the interactions between B. burgdorferi proteins, as well as between microbial proteins and host components, protein and non-protein components, highlighting their roles in pathogen persistence in the mammalian host. A better understanding of the contributions of protein interactions in the microbial virulence and persistence of B. burgdorferi would support development of novel therapeutics against the infection.


Subject(s)
Bacterial Proteins/metabolism , Borrelia burgdorferi/pathogenicity , Host-Pathogen Interactions/physiology , Lyme Disease/pathology , Virulence Factors/metabolism , Animals , Bacterial Proteins/genetics , Borrelia burgdorferi/drug effects , Borrelia burgdorferi/genetics , Humans , Ixodes/microbiology , Lyme Disease/drug therapy , Lyme Disease/microbiology , Protein Binding/physiology , Protein Interaction Maps , Virulence , Virulence Factors/genetics
5.
J Infect Dis ; 215(6): 1000-1009, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28453837

ABSTRACT

Borrelia burgdorferi genome harbors several paralogous gene families (pgf) that can encode immunogenic proteins of unknown function. Protein-protein interaction assays using a transmission-blocking vaccine candidate, BBA52, as bait identified an interacting partner in spirochetes-a member of pgf 54, annotated as BBI39. We show that BBI39 is a surface-exposed membrane antigen that is immunogenic during spirochete infection, despite the gene being primarily transcribed in the vector with a transient expression in the host only at tick-bite sites. Immunization of rodents with BBI39, or a diverse paralog, BBI36, or their combination impaired pathogen acquisition by the vector, transmission from ticks to hosts, or induction of disease. High-titer BBI39 immunoglobulin G antibodies, which have borreliacidal properties, could be generated through routine subcutaneous or oral immunization, further highlighting use of BBI39 proteins as novel Lyme disease vaccines that can target pathogens in the host or in ticks.


Subject(s)
Antibodies, Bacterial/blood , Bacterial Outer Membrane Proteins/immunology , Borrelia burgdorferi/genetics , Borrelia burgdorferi/immunology , Lyme Disease Vaccines/immunology , Animals , Ankle Joint/pathology , Antigens, Surface/immunology , Host-Pathogen Interactions , Ixodes/immunology , Lyme Disease/prevention & control , Mice , Mice, Inbred C3H , Protein Interaction Mapping , Vaccination
6.
J Infect Dis ; 213(11): 1786-95, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26747708

ABSTRACT

Borrelia burgdorferi harbors a limited set of transmembrane surface proteins, most of which constitute key targets of humoral immune responses. Here we show that BB0405, a conserved membrane-spanning protein of unknown function, fails to evoke detectable antibody responses despite its extracellular exposure. bb0405 is a member of an operon and ubiquitously expressed throughout the rodent-tick infection cycle. The gene product serves an essential function in vivo, as bb0405-deletion mutants are unable to transmit from ticks and establish infection in mammalian hosts. Despite the lack of BB0405-specific immunoglobulin M or immunoglobulin G antibodies during natural infection, mice immunized with a recombinant version of the protein elicited high-titer and remarkably long-lasting antibody responses, conferring significant host protection against tick-borne infection. Taken together, these studies highlight the essential role of an apparently immune-invisible borrelial transmembrane protein in facilitating infection and its usefulness as a target of protective host immunity blocking the transmission of B. burgdorferi.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Borrelia burgdorferi/immunology , Lyme Disease Vaccines/immunology , Lyme Disease/prevention & control , Animals , Antibodies, Bacterial/immunology , Bacterial Outer Membrane Proteins/biosynthesis , Bacterial Outer Membrane Proteins/genetics , Borrelia burgdorferi/genetics , Drug Delivery Systems , Female , Immunity, Humoral , Immunogenicity, Vaccine , Lyme Disease/immunology , Lyme Disease/microbiology , Lyme Disease/pathology , Mice, Inbred C3H , Mutagenesis
7.
Infect Immun ; 84(8): 2372-2381, 2016 08.
Article in English | MEDLINE | ID: mdl-27271745

ABSTRACT

High-temperature requirement protease A (HtrA) represents a family of serine proteases that play important roles in microbial biology. Unlike the genomes of most organisms, that of Borrelia burgdorferi notably encodes a single HtrA gene product, termed BbHtrA. Previous studies identified a few substrates of BbHtrA; however, their physiological relevance could not be ascertained, as targeted deletion of the gene has not been successful. Here we show that BbhtrA transcripts are induced during spirochete growth either in the stationary phase or at elevated temperature. Successful generation of a BbhtrA deletion mutant and restoration by genetic complementation suggest a nonessential role for this protease in microbial viability; however, its remarkable growth, morphological, and structural defects during cultivation at 37°C confirm a high-temperature requirement for protease activation and function. The BbhtrA-deficient spirochetes were unable to establish infection of mice, as evidenced by assessment of culture, PCR, and serology. We show that transcript abundance as well as proteolytic processing of a borrelial protein required for cell fission and infectivity, BB0323, is impaired in BbhtrA mutants grown at 37°C, which likely contributed to their inability to survive in a mammalian host. Together, these results demonstrate the physiological relevance of a unique temperature-regulated borrelial protease, BbHtrA, which further enlightens our knowledge of intriguing aspects of spirochete biology and infectivity.


Subject(s)
Borrelia burgdorferi/physiology , Lyme Disease/microbiology , Serine Endopeptidases/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Disease Models, Animal , Mice , Protein Binding , Proteolysis , Sequence Deletion , Temperature
8.
J Infect Dis ; 211(3): 462-71, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25139020

ABSTRACT

We have shown that Borrelia burgdorferi gene product BB0323 is essential for cell fission and pathogen persistence in vivo. Here we describe characterization of a conserved hypothetical protein annotated as BB0238, which specifically interacts with the N-terminal region of BB0323. We show that BB0238 is a subsurface protein, and similar to BB0323, exists in the periplasm and as a membrane-bound protein. Deletion of bb0238 in infectious B. burgdorferi did not affect microbial growth in vitro or survival in ticks, but the mutant was unable to persist in mice or transmit from ticks--defects that are restored on genetic complementation. Remarkably, BB0238 and BB0323 contribute to mutual posttranslational stability, because deletion of one causes dramatic reduction in the protein level of the other partner. Interference with the function of BB0238 or BB0323 and their interaction may provide novel strategies to combat B. burgdorferi infection.


Subject(s)
Bacterial Proteins/metabolism , Borrelia burgdorferi/metabolism , Lyme Disease/metabolism , Virulence Factors/metabolism , Virulence/genetics , Animals , Bacterial Proteins/genetics , Borrelia burgdorferi/genetics , Gene Deletion , Genetic Complementation Test/methods , Lyme Disease/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C3H , Ticks/microbiology , Virulence Factors/genetics
9.
J Biol Chem ; 289(18): 12813-22, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24662290

ABSTRACT

Ixodes scapularis ticks transmit a wide array of human and animal pathogens including Borrelia burgdorferi; however, how tick immune components influence the persistence of invading pathogens remains unknown. As originally demonstrated in Caenorhabditis elegans and later in Anopheles gambiae, we show here that an acellular gut barrier, resulting from the tyrosine cross-linking of the extracellular matrix, also exists in I. scapularis ticks. This dityrosine network (DTN) is dependent upon a dual oxidase (Duox), which is a member of the NADPH oxidase family. The Ixodes genome encodes for a single Duox and at least 16 potential peroxidase proteins, one of which, annotated as ISCW017368, together with Duox has been found to be indispensible for DTN formation. This barrier influences pathogen survival in the gut, as an impaired DTN in Doux knockdown or in specific peroxidase knockdown ticks, results in reduced levels of B. burgdorferi persistence within ticks. Absence of a complete DTN formation in knockdown ticks leads to the activation of specific tick innate immune pathway genes that potentially resulted in the reduction of spirochete levels. Together, these results highlighted the evolution of the DTN in a diverse set of arthropod vectors, including ticks, and its role in protecting invading pathogens like B. burgdorferi. Further understanding of the molecular basis of tick innate immune responses, vector-pathogen interaction, and their contributions in microbial persistence may help the development of new targets for disrupting the pathogen life cycle.


Subject(s)
Arthropod Proteins/metabolism , Arthropod Vectors/metabolism , Ixodes/metabolism , NADPH Oxidases/metabolism , Peroxidase/metabolism , Tyrosine/analogs & derivatives , Animals , Arthropod Proteins/genetics , Arthropod Vectors/genetics , Arthropod Vectors/microbiology , Borrelia burgdorferi/growth & development , Borrelia burgdorferi/physiology , Gastrointestinal Tract/microbiology , Gene Expression Regulation, Enzymologic , Humans , Ixodes/genetics , Ixodes/microbiology , Lyme Disease/microbiology , Lyme Disease/parasitology , Mice , Mice, Inbred C3H , Microbial Viability/genetics , Microscopy, Confocal , NADPH Oxidases/genetics , Peroxidase/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Tyrosine/genetics , Tyrosine/metabolism
10.
mSystems ; 8(6): e0092723, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-37874165

ABSTRACT

IMPORTANCE: Here, we demonstrate the adaptability of spatial "omics" methods to identify interphylum processes regulated at the vector-host interface of ticks during a mammalian blood meal. This approach enables a better understanding of complex bipartite or tripartite molecular interactions between hosts, arthropod vectors and transmitted pathogens, and contributes toward the development of spatially aware therapeutic target discovery and description.


Subject(s)
Lipidomics , Ticks , Animals , Guinea Pigs , Host-Pathogen Interactions , Mammals , Skin
11.
Science ; 379(6628): eabl3837, 2023 01 13.
Article in English | MEDLINE | ID: mdl-36634189

ABSTRACT

Ancestral signaling pathways serve critical roles in metazoan development, physiology, and immunity. We report an evolutionary interspecies communication pathway involving a central Ixodes scapularis tick receptor termed Dome1, which acquired a mammalian cytokine receptor motif exhibiting high affinity for interferon-gamma (IFN-γ). Host-derived IFN-γ facilitates Dome1-mediated activation of the Ixodes JAK-STAT pathway. This accelerates tick blood meal acquisition and development while upregulating antimicrobial components. The Dome1-JAK-STAT pathway, which exists in most Ixodid tick genomes, regulates the regeneration and proliferation of gut cells-including stem cells-and dictates metamorphosis through the Hedgehog and Notch-Delta networks, ultimately affecting Ixodes vectorial competence. We highlight the evolutionary dependence of I. scapularis on mammalian hosts through cross-species signaling mechanisms that dually influence arthropod immunity and development.


Subject(s)
Arachnid Vectors , Host-Parasite Interactions , Ixodes , Janus Kinases , Receptors, Cytokine , STAT Transcription Factors , Animals , Interferon-gamma/metabolism , Ixodes/genetics , Ixodes/immunology , Janus Kinases/genetics , Janus Kinases/metabolism , Signal Transduction , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Host-Parasite Interactions/immunology , Receptors, Cytokine/metabolism , Arachnid Vectors/immunology
12.
Infect Immun ; 80(1): 82-90, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22025510

ABSTRACT

Borrelia burgdorferi, a tick-borne bacterial pathogen, causes a disseminated infection involving multiple organs known as Lyme disease. Surface proteins can directly participate in microbial virulence by facilitating pathogen dissemination via interaction with host factors. We show here that a fraction of the B. burgdorferi chromosomal gene product BB0337, annotated as enolase or phosphopyruvate dehydratase, is associated with spirochete outer membrane and is surface exposed. B. burgdorferi enolase, either in a recombinant form or as a membrane-bound native antigen, displays enzymatic activities intrinsic to the glycolytic pathway. However, the protein also interacts with host plasminogen, potentially leading to its activation and resulting in B. burgdorferi-induced fibrinolysis. As expected, enolase displayed consistent expression in vivo, however, with a variable temporal and spatial expression during spirochete infection in mice and ticks. Despite an extracellular exposure of the antigen and a potential role in host-pathogen interaction, active immunization of mice with recombinant enolase failed to evoke protective immunity against subsequent B. burgdorferi infection. In contrast, enolase immunization of murine hosts significantly reduced the acquisition of spirochetes by feeding ticks, suggesting that the protein could have a stage-specific role in B. burgdorferi survival in the feeding vector. Strategies to interfere with the function of surface enolase could contribute to the development of novel preventive measures to interrupt the spirochete infection cycle and reduce the incidences of Lyme disease.


Subject(s)
Borrelia burgdorferi/enzymology , Borrelia burgdorferi/physiology , Host-Pathogen Interactions , Microbial Viability , Phosphopyruvate Hydratase/metabolism , Plasminogen/metabolism , Ticks/microbiology , Animals , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/metabolism , Borrelia burgdorferi/metabolism , Borrelia burgdorferi/pathogenicity , Female , Fibrinolysis , Gene Expression Profiling , Mice , Mice, Inbred C3H , Protein Binding , Protein Interaction Mapping
14.
Methods Mol Biol ; 1690: 105-114, 2018.
Article in English | MEDLINE | ID: mdl-29032540

ABSTRACT

Borrelia burgdorferi is maintained in nature by a tick-rodent infection cycle where it traverses and colonizes a variety of host and vector tissues. A tick-borne murine model has been developed to study Lyme disease in the laboratory, which has a substantial impact in advancing our knowledge of spirochete infectivity and pathogenesis. Here, we detail a microinjection-based method for rapid and efficient infection of ticks with B. burgdorferi. While laboratory generation of B. burgdorferi-infected nymphs via natural larval engorgement on infected hosts and subsequent molting could take several weeks to months, the microinjection-based infection procedure requires only a few hours to generate infected ticks and allows introduction of defined quantities of spirochetes, including mutant isolates that are attenuated for infection in mice and thus cannot be naturally acquired by ticks. We also describe a quantitative PCR-based protocol for the measurement of B. burgdorferi in tick and murine hosts targeting spirochete RNA that is highly efficient, reproducible, and a better surrogate of active infection.


Subject(s)
Borrelia burgdorferi/isolation & purification , Lyme Disease/microbiology , Lyme Disease/transmission , Microinjections/methods , Ticks/microbiology , Animals , Borrelia burgdorferi/genetics , Borrelia burgdorferi/physiology , DNA, Complementary/genetics , Disease Models, Animal , Lyme Disease/pathology , Mice , RNA, Bacterial/genetics , Real-Time Polymerase Chain Reaction/methods , Reverse Transcriptase Polymerase Chain Reaction/methods
15.
Methods Mol Biol ; 1690: 259-277, 2018.
Article in English | MEDLINE | ID: mdl-29032550

ABSTRACT

The proteome of Borrelia burgdorferi undergoes dynamic alterations as the microbe cycles through and persists in diverse host or vector environments. Therefore, studies of B. burgdorferi proteome and protein-protein interactions, which play central roles in biological processes in diverse organisms, are critical in understanding biology and infectivity of spirochetes. Here, we describe the proteomic analysis of B. burgdorferi by two-dimensional (2-D) gel electrophoresis followed by protein identification via liquid chromatography-mass spectrometry and database searching. We also describe assays for studying the interaction between borrelial proteins: a novel high-throughput luciferase assay, yeast two-hybrid assay, and a far-Western assay that are routinely used in our laboratories.


Subject(s)
Bacterial Proteins/metabolism , Borrelia burgdorferi/metabolism , Lyme Disease/microbiology , Protein Interaction Mapping/methods , Protein Interaction Maps , Proteomics/methods , Bacterial Proteins/analysis , Blotting, Far-Western/methods , Borrelia burgdorferi/chemistry , Electrophoresis, Gel, Two-Dimensional/methods , Electrophoresis, Polyacrylamide Gel/methods , HEK293 Cells , Humans , Mass Spectrometry/methods , Proteome/analysis , Proteome/metabolism , Two-Hybrid System Techniques
16.
Sci Rep ; 7(1): 2932, 2017 06 07.
Article in English | MEDLINE | ID: mdl-28592866

ABSTRACT

Two Borrelia burgdorferi interacting proteins, BB0238 and BB0323, play distinct roles in pathogen biology and infectivity although a significance of their interaction remained enigmatic. Here we identified the polypeptide segment essential for BB0238-BB0323 interaction and examined how it supports spirochete infectivity. We show that the interaction region in BB0323 requires amino acid residues 22-200, suggesting that the binding encompasses discontinuous protein segments. In contrast, the interaction region in BB0238 spans only 11 amino acids, residues 120-130. A deletion of these 11 amino acids neither alters the overall secondary structure of the protein, nor affects its stability or oligomerization property, however, it reduces the post-translational stability of the binding partner, BB0323. Mutant B. burgdorferi isolates producing BB0238 lacking the 11-amino acid interaction region were able to persist in ticks but failed to transmit to mice or to establish infection. These results suggest that BB0238-BB0323 interaction is critical for post-translational stability of BB0323, and that this interaction is important for mammalian infectivity and transmission of B. burgdorferi. We show that saturation or inhibition of BB0238-BB0323 interaction could be studied in a luciferase assay, which could be amenable for future identification of small molecule inhibitors to combat B. burgdorferi infection.


Subject(s)
Bacterial Proteins/metabolism , Borrelia burgdorferi/physiology , Host-Pathogen Interactions , Lyme Disease/metabolism , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Disease Models, Animal , Lyme Disease/microbiology , Mice , Protein Binding , Protein Interaction Domains and Motifs
17.
Nat Commun ; 8: 14401, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28195158

ABSTRACT

The insect immune deficiency (IMD) pathway resembles the tumour necrosis factor receptor network in mammals and senses diaminopimelic-type peptidoglycans present in Gram-negative bacteria. Whether unidentified chemical moieties activate the IMD signalling cascade remains unknown. Here, we show that infection-derived lipids 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoglycerol (POPG) and 1-palmitoyl-2-oleoyl diacylglycerol (PODAG) stimulate the IMD pathway of ticks. The tick IMD network protects against colonization by three distinct bacteria, that is the Lyme disease spirochete Borrelia burgdorferi and the rickettsial agents Anaplasma phagocytophilum and A. marginale. Cell signalling ensues in the absence of transmembrane peptidoglycan recognition proteins and the adaptor molecules Fas-associated protein with a death domain (FADD) and IMD. Conversely, biochemical interactions occur between x-linked inhibitor of apoptosis protein (XIAP), an E3 ubiquitin ligase, and the E2 conjugating enzyme Bendless. We propose the existence of two functionally distinct IMD networks, one in insects and another in ticks.


Subject(s)
Arthropods/immunology , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/veterinary , Ixodes/immunology , Lipids/adverse effects , Lipids/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Anaplasma marginale/immunology , Anaplasma marginale/pathogenicity , Anaplasma phagocytophilum/immunology , Anaplasma phagocytophilum/pathogenicity , Animals , Arthropods/metabolism , Borrelia burgdorferi/immunology , Borrelia burgdorferi/pathogenicity , Carrier Proteins , Disease Models, Animal , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Escherichia coli/genetics , Fas-Associated Death Domain Protein , Gene Silencing , HEK293 Cells , Humans , Ixodes/metabolism , Lyme Disease/immunology , Phosphatidylglycerols/immunology , RNA, Small Interfering/metabolism , Recombinant Proteins , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism
18.
Cell Host Microbe ; 20(1): 91-8, 2016 Jul 13.
Article in English | MEDLINE | ID: mdl-27374407

ABSTRACT

Evolution of hematophagy in blood-sucking parasites likely involves communication with their hosts. We find that Ixodes ticks are responsive to IFNγ acquired in a blood meal from mice infected with the Lyme disease-causing bacteria Borrelia burgdorferi, leading to induction of antimicrobial responses. Ixodes ticks parasitizing B. burgdorferi-infected mice upregulated an I. scapularis Rho-like GTPase (IGTPase). IGTPase knockdown enhanced B. burgdorferi levels in post-fed ticks, suggesting this protein controls spirochete survival. Notably, IGTPase was only induced during pathogen acquisition from mice and not upon transmission to naive hosts. Microinjection of ticks with IFNγ induced IGTPase, and ticks parasitizing IFNγ knockout mice, failed to upregulate IGTPase. Additionally, ticks lacking the transcription factor STAT, which signals downstream of IFNγ, did not induce IGTPase. IGTPase expression induced antimicrobial peptides, including Dae2, previously shown to inhibit B. burgdorferi. These results identify an interspecies signaling cascade allowing ticks to detect invading bacteria and mount microbicidal responses.


Subject(s)
Blood/immunology , Borrelia burgdorferi/immunology , Feeding Behavior , Immunity, Innate , Interferons/metabolism , Ixodes/physiology , Signal Transduction , Animals , Blood/microbiology , Gene Knockdown Techniques , Ixodes/immunology , Ixodes/microbiology , Mice, Inbred C3H , Mice, Knockout , Monomeric GTP-Binding Proteins/metabolism
19.
Article in English | MEDLINE | ID: mdl-25202684

ABSTRACT

Ixodes scapularis, commonly known as the deer tick, transmits a wide array of human and animal pathogens including Borrelia burgdorferi. Despite substantial advances in our understanding of immunity in model arthropods, including other disease vectors, precisely how I. scapularis immunity functions and influences persistence of invading pathogens remains largely unknown. This review provides a comprehensive analysis of the recently sequenced I. scapularis genome for the occurrence of immune-related genes and related pathways. We will also discuss the potential influence of immunity-related genes on the persistence of tick-borne pathogens with an emphasis on the Lyme disease pathogen B. burgdorferi. Further enhancement of our knowledge of tick immune responses is critical to understanding the molecular basis of the persistence of tick-borne pathogens and development of novel interventions against the relevant infections.


Subject(s)
Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immunity/genetics , Ixodes/genetics , Ixodes/immunology , Animals , Antimicrobial Cationic Peptides/metabolism , Arachnid Vectors , Genome, Insect , Genomics , Humans , Ixodes/metabolism , Ixodes/microbiology , Phagocytosis/genetics , Phagocytosis/immunology , Reactive Oxygen Species/metabolism , Signal Transduction , Tick-Borne Diseases/microbiology , Tick-Borne Diseases/transmission
20.
Pathog Dis ; 70(2): 176-84, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24376161

ABSTRACT

OspC is produced by all species of the Borrelia burgdorferi sensu lato complex and is required for infectivity in mammals. To test the hypothesis that the conserved C-terminal motif (C10) of OspC is required for function in vivo, a mutant B. burgdorferi strain (B31::ospCΔC10) was created in which ospC was replaced with an ospC gene lacking the C10 motif. The ability of the mutant to infect mice was investigated using tick transmission and needle inoculation. Infectivity was assessed by cultivation, qRT-PCR, and measurement of IgG antibody responses. B31::ospCΔC10 retained the ability to infect mice by both needle and tick challenge and was competent to survive in ticks after exposure to the blood meal. To determine whether recombinant OspC protein lacking the C-terminal 10 amino acid residues (rOspCΔC10) can bind plasminogen, the only known mammalian-derived ligand for OspC, binding analyses were performed. Deletion of the C10 motif resulted in a statistically significant decrease in plasminogen binding. Although deletion of the C10 motif influenced plasminogen binding, it can be concluded that the C10 motif is not required for OspC to carry out its critical in vivo functions in tick to mouse transmission.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/metabolism , Borrelia burgdorferi Group/physiology , Lyme Disease/transmission , Virulence Factors/metabolism , Amino Acid Motifs , Animals , Antigens, Bacterial/genetics , Bacterial Adhesion , Bacterial Outer Membrane Proteins/genetics , Borrelia burgdorferi Group/genetics , DNA Mutational Analysis , Mice, Inbred C3H , Plasminogen/metabolism , Sequence Deletion , Ticks
SELECTION OF CITATIONS
SEARCH DETAIL