Advertisement
Molecular Bases of Disease| Volume 289, ISSUE 18, P12813-12822, May 02, 2014

A Dityrosine Network Mediated by Dual Oxidase and Peroxidase Influences the Persistence of Lyme Disease Pathogens within the Vector*

  • Xiuli Yang
    Footnotes
    Affiliations
    Department of Veterinary Medicine and Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, Maryland 20742
    Search for articles by this author
  • Alexis A. Smith
    Footnotes
    Affiliations
    Department of Veterinary Medicine and Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, Maryland 20742
    Search for articles by this author
  • Mark S. Williams
    Affiliations
    Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland 21201
    Search for articles by this author
  • Utpal Pal
    Correspondence
    To whom correspondence should be addressed: Dept. of Veterinary Medicine, Bldg. 795, Rm. 1341, University of Maryland, College Park, MD 20742. Tel.: 301-314-2118; Fax: 301-314-6855
    Affiliations
    Department of Veterinary Medicine and Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, Maryland 20742
    Search for articles by this author
  • Author Footnotes
    * This work was supported, in whole or in part, by National Institutes of Health Grant R01AI080615 (to U. P.).
    This article contains supplemental Table S1.
    1 Both authors contributed equally to this work.
Open AccessPublished:March 24, 2014DOI:https://doi.org/10.1074/jbc.M113.538272
      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.

      Introduction

      Lyme disease, the most prevalent tick-borne infection in the United States, is caused by the spirochete bacterium, Borrelia burgdorferi (
      • Stanek G.
      • Wormser G.P.
      • Gray J.
      • Strle F.
      Lyme borreliosis.
      ,
      • Steere A.C.
      Lyme disease.
      ). The pathogen thrives in nature through an intricate infectious cycle, including an arthropod vector, Ixodes scapularis, and a vertebrate host, primarily wild rodents (
      • Radolf J.D.
      • Caimano M.J.
      • Stevenson B.
      • Hu L.T.
      Of ticks, mice and men: understanding the dual-host lifestyle of Lyme disease spirochaetes.
      ). I. scapularis engorges upon the host dermis, usually for several days, and during this feeding process, pathogens like B. burgdorferi can efficiently transit between the arthropod and mammalian hosts (
      • Piesman J.
      • Oliver J.R.
      • Sinsky R.J.
      Growth kinetics of the Lyme disease spirochete (Borrelia burgdorferi) in vector ticks (Ixodes dammini).
      ,
      • Tyson K.R.
      • Piesman J.
      Lyme disease spirochete-tick-host interactions.
      ). Once acquired by ticks, the pathogen remains in the vector gut throughout the intermolt period until the next blood meal, when a fraction of the spirochetes exit the gut, enter the hemocoel, and finally invade the salivary glands (
      • de Silva A.M.
      • Tyson K.R.
      • Pal U.
      Molecular characterization of the tick-Borrelia interface.
      ,
      • Pal U.
      • Fikrig E.
      Tick interactions.
      ). Once in the glands, the bacteria can then be transmitted to a new host. Thus, maintenance of the pathogen in the enzootic cycle requires its successful persistence through multiple developmental stages of the arthropod, as well as its coordinated dissemination through tick tissues to a new host.
      The gut environment of blood-feeding arthropods including that of the feeding tick (
      • Dong Y.
      • Aguilar R.
      • Xi Z.
      • Warr E.
      • Mongin E.
      • Dimopoulos G.
      Anopheles gambiae immune responses to human and rodent Plasmodium parasite species.
      • Munderloh U.G.
      • Kurtti T.J.
      Cellular and molecular interrelationships between ticks and prokaryotic tick-borne pathogens.
      ,
      • Yassine H.
      • Osta M.A.
      Anopheles gambiae innate immunity.
      ,
      • Sonenshine D.E.
      ), which pathogens like B. burgdorferi encounters upon its arrival from an infected host, is likely to be hostile. As spirochetes adapt to survive in the luminal spaces of the gut, the bacteria must avoid the intense digestive activities of gut epithelial cells and at the same time bypass innate immune defense mechanisms (
      • Kopácek P.
      • Hajdusek O.
      • Buresová V.
      • Daffre S.
      Tick innate immunity.
      ,
      • Sonenshine D.E.
      • Hynes W.L.
      • Ceraul S.M.
      • Mitchell R.
      • Benzine T.
      Host blood proteins and peptides in the midgut of the tick Dermacentor variabilis contribute to bacterial control.
      ). A number of studies in model arthropods illustrated the existence of diverse and potent pathogen recognition molecules, which in turn induce the Toll (against bacterial, viral, and fungal pathogens) or immune deficiency (against Gram bacteria) pathways (
      • Govind S.
      • Nehm R.H.
      Innate immunity in fruit flies: a textbook example of genomic recycling.
      • Hoffmann J.A.
      • Reichhart J.M.
      Drosophila innate immunity: an evolutionary perspective.
      ,
      • Tanji T.
      • Hu X.
      • Weber A.N.
      • Ip Y.T.
      Toll and IMD pathways synergistically activate an innate immune response in Drosophila melanogaster.
      ,
      • Tanji T.
      • Ip Y.T.
      Regulators of the Toll and Imd pathways in the Drosophila innate immune response.
      ,
      • Valanne S.
      • Wang J.H.
      • Rämet M.
      The Drosophila Toll signaling pathway.
      ). How these pathways operate in I. scapularis, especially their activation following B. burgdorferi invasion, however, remains obscure. Nevertheless, ticks likely produce classical antimicrobial peptides in the gut (
      • Hynes W.L.
      • Ceraul S.M.
      • Todd S.M.
      • Seguin K.C.
      • Sonenshine D.E.
      A defensin-like gene expressed in the black-legged tick, Ixodes scapularis.
      ), although their role in tick immunity remains enigmatic; for example, although a defensin-like gene is up-regulated in Ixodes ticks following infection with B. burgdorferi, it does not clear the infection (
      • Rudenko N.
      • Golovchenko M.
      • Edwards M.J.
      • Grubhoffer L.
      Differential expression of Ixodes ricinus tick genes induced by blood feeding or Borrelia burgdorferi infection.
      ). Earlier studies also reported the development of annotated catalogs of organ-specific transcripts (
      • Anderson J.M.
      • Sonenshine D.E.
      • Valenzuela J.G.
      Exploring the mialome of ticks: an annotated catalogue of midgut transcripts from the hard tick, Dermacentor variabilis (Acari: Ixodidae).
      ,
      • Ribeiro J.M.
      • Alarcon-Chaidez F.
      • Francischetti I.M.
      • Mans B.J.
      • Mather T.N.
      • Valenzuela J.G.
      • Wikel S.K.
      An annotated catalog of salivary gland transcripts from Ixodes scapularis ticks.
      ); these studies will have a significant impact on our understanding of tick biology and vector-pathogen interaction. Nevertheless, despite these studies, the precise mechanism by which B. burgdorferi survives in the gut or evades the tick innate immune system requires further investigation.
      Replication of pathogens or commensal bacteria within the gut during blood meal engorgement by arthropods could activate epithelial immunity; thus, arthropods must develop a strategy to maintain the gut homeostasis. A previous study demonstrated that a molecular barrier, termed the dityrosine network (DTN),
      The abbreviations used are: DTN
      dityrosine network
      Duox
      dual oxidase
      ROS
      reactive oxygen species
      dsRNA
      double-stranded RNA
      qRT-PCR
      quantitative RT-PCR
      NOS
      nitric-oxide synthase.
      is formed within the gut of a blood-sucking arthropod, Anopheles gambiae, during the uptake of a blood meal and surrounds the gut epithelial layer (
      • Kumar S.
      • Molina-Cruz A.
      • Gupta L.
      • Rodrigues J.
      • Barillas-Mury C.
      A peroxidase/dual oxidase system modulates midgut epithelial immunity in Anopheles gambiae.
      ). The formation of the DTN decreases the gut permeability to immune elicitors, thereby protecting the beneficial gut microbiota, which in turn supports the invading pathogens like Plasmodium. The DTN in mosquitoes is dependent upon two separate enzymes, dual oxidase (Duox) and a heme peroxidase, which catalyze the cross-linking of tyrosines (
      • Kumar S.
      • Molina-Cruz A.
      • Gupta L.
      • Rodrigues J.
      • Barillas-Mury C.
      A peroxidase/dual oxidase system modulates midgut epithelial immunity in Anopheles gambiae.
      ). Dual oxidase is a prominent member of the NADPH oxidase family and a transmembrane protein that has previously been shown to be a source of local reactive oxygen species (ROS), which play an important role in maintenance of mucosal immunity (
      • Donkó A.
      • Péterfi Z.
      • Sum A.
      • Leto T.
      • Geiszt M.
      Dual oxidases.
      ). In Drosophila, targeted depletion of Duox and the subsequent decrease in ROS production leads to uncontrolled proliferation of gut bacteria and also renders the fly more susceptible to microbial infection (
      • Ha E.M.
      • Oh C.T.
      • Bae Y.S.
      • Lee W.J.
      A direct role for dual oxidase in Drosophila gut immunity.
      ). Similarly, expression of Duox in airway epithelial cells promotes antimicrobial defenses (
      • Geiszt M.
      • Witta J.
      • Baffi J.
      • Lekstrom K.
      • Leto T.L.
      Dual oxidases represent novel hydrogen peroxide sources supporting mucosal surface host defense.
      ).
      A search in the I. scapularis genome database indicated that the arthropod encodes for a single Duox and at least 16 potential peroxidases. Considering an important yet less understood role of Duox and specific peroxidase enzymes in formation of an epithelial barrier influencing the host-microbe homeostasis, including controlling proliferation of symbiotic or nonsymbiotic bacterial communities in the gut during feeding (
      • Kumar S.
      • Molina-Cruz A.
      • Gupta L.
      • Rodrigues J.
      • Barillas-Mury C.
      A peroxidase/dual oxidase system modulates midgut epithelial immunity in Anopheles gambiae.
      ,
      • Ha E.M.
      • Oh C.T.
      • Bae Y.S.
      • Lee W.J.
      A direct role for dual oxidase in Drosophila gut immunity.
      ,
      • Bae Y.S.
      • Choi M.K.
      • Lee W.J.
      Dual oxidase in mucosal immunity and host-microbe homeostasis.
      ), we sought to investigate how these enzymes affect persistence of a major tick-borne pathogen, B. burgdorferi. These studies could contribute to a better understanding of specific aspects of tick immunity, as well as help in the development of new strategies that interfere with pathogen persistence in an enzootic infectious cycle.

      EXPERIMENTAL PROCEDURES

      B. burgdorferi, Mice, and Ticks

      A low passage infectious isolate of B. burgdorferi, clone B31-A3, was used in this study (
      • Elias A.F.
      • Stewart P.E.
      • Grimm D.
      • Caimano M.J.
      • Eggers C.H.
      • Tilly K.
      • Bono J.L.
      • Akins D.R.
      • Radolf J.D.
      • Schwan T.G.
      • Rosa P.
      Clonal polymorphism of Borrelia burgdorferi strain B31 MI: implications for mutagenesis in an infectious strain background.
      ). Four- to six-week-old C3H/HeN mice were purchased from the National Institutes of Health. Animal experiments were performed in accordance with the guidelines of the Institutional Animal Care and Use Committee and Institutional Biosafety Committee. I. scapularis ticks used in this study originated from a colony that is maintained in our laboratory.

      RNA Isolation and PCR

      The oligonucleotide primer sequences for the quantitative RT-PCR analysis of tick gene expression or measurement of bacterial load are indicated in supplemental Table S1. For analysis of Duox and peroxidase expression, the naïve nymphal ticks or nymphs microinjected with dsRNA were allowed to feed on naïve or B. burgdorferi-infected C3H/HeN mice (25 ticks/mouse) as detailed (
      • Kariu T.
      • Coleman A.S.
      • Anderson J.F.
      • Pal U.
      Methods for rapid transfer and localization of Lyme disease pathogens within the tick gut.
      ,
      • Kariu T.
      • Smith A.
      • Yang X.
      • Pal U.
      A chitin deacetylase-like protein is a predominant constituent of tick peritrophic membrane that influences the persistence of Lyme disease pathogens within the vector.
      ). The ticks were collected at various time points of feeding or as postfed nymphs. Individual ticks were processed separately. Total RNA was extracted from ticks using TRIzol (Invitrogen), treated with RNase-free DNaseI (Qiagen), and then reverse transcribed to cDNA using the AffinityScript cDNA synthesis kit (Stratagene). The relative transcript levels of the target genes in ticks were assessed by quantitative RT-PCR (qRT-PCR) (
      • Yang X.
      • Coleman A.S.
      • Anguita J.
      • Pal U.
      A chromosomally encoded virulence factor protects the Lyme disease pathogen against host-adaptive immunity.
      ). To test the efficiency and exclude nonspecific amplification of the primer pairs, the qRT-PCR amplification in each well was followed by melt-curve analysis. The amplification was performed in an iQ5 real time thermal cycler (Bio-Rad) using SYBR Green Supermix (Bio-Rad) as detailed. Transcript levels of individual genes were calculated using the 2−ΔΔCt method and further normalized against tick β-actin transcripts (
      • Yang X.
      • Coleman A.S.
      • Anguita J.
      • Pal U.
      A chromosomally encoded virulence factor protects the Lyme disease pathogen against host-adaptive immunity.
      ). For assessment of levels of B. burgdorferi or gut bacterial numbers in ticks, the flaB or 16S transcript levels, respectively, were quantitated using qPCR as detailed (
      • Yang X.
      • Coleman A.S.
      • Anguita J.
      • Pal U.
      A chromosomally encoded virulence factor protects the Lyme disease pathogen against host-adaptive immunity.
      ).

      RNA Interference

      RNA interference of target tick genes was accomplished using our published procedures (
      • Kariu T.
      • Smith A.
      • Yang X.
      • Pal U.
      A chitin deacetylase-like protein is a predominant constituent of tick peritrophic membrane that influences the persistence of Lyme disease pathogens within the vector.
      ,
      • Pal U.
      • Li X.
      • Wang T.
      • Montgomery R.R.
      • Ramamoorthi N.
      • Desilva A.M.
      • Bao F.
      • Yang X.
      • Pypaert M.
      • Pradhan D.
      • Kantor F.S.
      • Telford S.
      • Anderson J.F.
      • Fikrig E.
      TROSPA, an Ixodes scapularis receptor for Borrelia burgdorferi.
      ). Tick cDNA was prepared as described above and used as a template to amplify various fragments of the open reading frames of the corresponding target genes: Duox (accession number ISCW007865) or peroxidases (accession numbers ISCW017368 and ISCW002528, referred to herein as peroxidase 002528 and peroxidase 017368). A fragment of the GFP gene was also amplified as a control. The primers used to amplify the specific DNA fragments are listed in supplemental Table S1. The Duox, peroxidase, and GFP amplicons were separately cloned into the corresponding restriction sites of the L4440 double T7 script vector, and the dsRNA was synthesized and purified using a commercial kit (MEGAscript RNAi kit; Ambion, Inc.). Five μl of the dsRNA (2 μg/μl) was loaded into capillary tubes and microinjected into the gut of unfed nymphs (25 ticks/group) as described. The injected nymphs were kept overnight in an incubator and used for feeding on mice, and repleted ticks were individually processed for assessment of gene silencing and pathogen levels using qRT-PCR analysis. Primers that bind further upstream and downstream of the target cDNA encompassing the dsRNA sequence were used for the qRT-PCR, as indicated in supplemental Table S1.

      Infection Studies

      For an assessment of whether the RNA interference-mediated reduction of Duox or peroxidase expression affected B. burgdorferi acquisition by nymphal ticks, C3H mice were infected with spirochetes (105 spirochetes/mouse). Following 14 days of infection, the corresponding Duox, peroxidase, or GFP dsRNA preparations were administrated to nymphs, and the microinjected ticks were placed on B. burgdorferi-infected mice (25 ticks/mouse). Groups of ticks were forcibly detached from the mice following 48 h of feeding, whereas the others were allowed to feed to repletion and then collected. The flaB transcripts were detected using qRT-PCR as detailed earlier. At least three independent experiments were performed, and 6–10 ticks were recovered from each mouse. Each tick was analyzed individually for measurement of target transcripts or pathogen burden using qRT-PCR.

      Confocal Immunofluorescence Microscopy

      For confocal microscopy (
      • Kariu T.
      • Smith A.
      • Yang X.
      • Pal U.
      A chitin deacetylase-like protein is a predominant constituent of tick peritrophic membrane that influences the persistence of Lyme disease pathogens within the vector.
      ), tissues from engorged ticks were fixed for 24 h at 4 °C in 4% (w/v) paraformaldehyde in PBS followed by infiltration with 30% sucrose. Cryosectioning was performed at −20 °C using a cryotome, and 5-μm-thick sections were incubated in PBS with 5% goat serum and 0.5% Tween 20, as detailed (
      • Kariu T.
      • Smith A.
      • Yang X.
      • Pal U.
      A chitin deacetylase-like protein is a predominant constituent of tick peritrophic membrane that influences the persistence of Lyme disease pathogens within the vector.
      ). The sections were then labeled with mouse monoclonal antibodies against dityrosine (Genox Corporation), rabbit anti-peroxidase antibodies (Sigma), or isotype control (mouse or rabbit IgG; Sigma) followed by Alexa 488-labeled goat anti-mouse or Alexa 568-labeled goat anti-rabbit IgG (Invitrogen). The slides were then washed using PBS with 0.05% Tween 20, stained with DAPI (Invitrogen), and imaged by a LSM 510 laser confocal microscope (Zeiss) as described (
      • Kariu T.
      • Smith A.
      • Yang X.
      • Pal U.
      A chitin deacetylase-like protein is a predominant constituent of tick peritrophic membrane that influences the persistence of Lyme disease pathogens within the vector.
      ).

      Assays for Nitric-oxide Synthase Activity

      The NOS activity of the tick tissues was analyzed using a commercial kit (Ultrasensitive colorimetric assay kit; Oxford Biomedical Research) as described (
      • Ghigo D.
      • Riganti C.
      • Gazzano E.
      • Costamagna C.
      • Bosia A.
      Cycling of NADPH by glucose 6-phosphate dehydrogenase optimizes the spectrophotometric assay of nitric oxide synthase activity in cell lysates.
      ). Briefly, whole tick samples were homogenized in PBS under the ice and centrifuged briefly, and protein concentration in the collected supernatant was measured. The NOS activity was detected using an equal amount of protein (40 μg) from groups of Duox, peroxidase, or GFP knockdown ticks as detailed, and the OD values at 540 nm were calculated using a standard curve consisting of serial dilutions of nitrite from 0 to 100 μm.

      Bioinformatics and Statistical Analysis

      Unless indicated otherwise, protein annotation and searches were executed using the VectorBase database. Analyses of protein families and domains were performed using the Pfam and CDD databases. Sequence alignment analysis was performed using Cobalt Constraint-based Multiple Protein Alignment Tool. The results are expressed as the means ± S.E. Statistical significance of differences observed between experimental and control groups were analyzed using GraphPad Prism version 4.0 (GraphPad Software). A two-tailed Student's t test was utilized to compare the mean values, and p < 0.05 was considered significant.

      RESULTS

      Expression of Dual Oxidase during Early Tick Feeding Coincides with the Replication of Bacteria in the Gut, which Features a Distinct Dityrosine Network

      Ixodid ticks are known to harbor a diverse set of gut bacteria (
      • van Overbeek L.
      • Gassner F.
      • van der Plas C.L.
      • Kastelein P.
      • Nunes-da Rocha U.
      • Takken W.
      Diversity of Ixodes ricinus tick-associated bacterial communities from different forests.
      ,
      • Moreno C.X.
      • Moy F.
      • Daniels T.J.
      • Godfrey H.P.
      • Cabello F.C.
      Molecular analysis of microbial communities identified in different developmental stages of Ixodes scapularis ticks from Westchester and Dutchess Counties, New York.
      ). To further study the role of the Duox in the genesis of a DTN and protection of invading pathogens like B. burgdorferi, we first assessed replication of the gut bacterial population during feeding. To accomplish this, naïve mice (3 animals/group) were parasitized with nymphal ticks (15 ticks/mouse), and partially fed ticks were removed at 6–48 h of feeding. Batches of harvested ticks were subjected to total RNA or DNA isolation. Total bacterial numbers were examined by qPCR analyses using tick DNA as a template and generic bacteria-specific 16 S primers, whereas expression of Duox was determined using qRT-PCR. The results showed that bacterial numbers in the feeding tick gut peaks at ∼20 h during engorgement (Fig. 1A). Although up-regulation of Duox occurred at multiple time points during feeding, the earliest peak is recorded at 20 h (Fig. 1B), which temporally coincides with the greatest bacterial numbers in feeding ticks. These data suggest that the induction of Duox during the initial hours of feeding potentially in response to gut microbe replication. We then perform a similar tick feeding study to assess whether B. burgdorferi presence within the gut also induces Duox expression. The results show that the ticks that parasitized B. burgdorferi-infected mice contained at least 3-fold higher levels of Duox transcripts in comparison with the ticks that fed on naïve mice (Fig. 1C). We next employed confocal immunofluorescence microscopy to assess whether a DTN structure could also be localized in feeding ticks. To accomplish this, samples of gut and a control organ (salivary gland) were removed from 24–48 h fed ticks, and frozen tissue sections were labeled with a mouse monoclonal antibody specifically against dityrosine or isotype control antibodies. The data indicated specific binding of dityrosine antibodies to the tick gut, indicating the presence of a DTN, whereas no reactivity was recorded for the salivary glands (Fig. 1D) or in gut tissues incubated with isotype control antibodies.
      Figure thumbnail gr1
      FIGURE 1.Proliferation of bacteria in feeding ticks coincides with expression of Duox in the gut, which contained a detectable dityrosine network. A, proliferation of gut bacterial population within feeding ticks. Naïve nymphal ticks were allowed to parasitize naïve mice and were collected at different times after placement on the mice. Bacterial numbers were measured by qPCR detection of 16S rRNA levels. B, expression of Duox. Naïve nymphal ticks were collected at different times of feeding on mice or as fully engorged postfed ticks, and Duox transcript levels were measured by qRT-PCR. Means ± S.E. of three independent infection experiments are presented. C, B. burgdorferi infection in ticks further enhances duox transcript levels. Naïve nymphal ticks were allowed to parasitize uninfected or B. burgdorferi (Bb)-infected mouse and collected at 48 h after placement on mice, and Duox transcript levels were measured by qRT-PCR. D, the existence of a DTN within the tick gut. Feeding ticks were collected at 24 h following their placement on mice, and confocal immunofluorescence staining of tick tissue sections (gut and salivary glands) was performed using isotype control IgG or dityrosine antibodies (labeled green). The nuclei in tick tissues were labeled with DAPI (blue). EC, epithelial cell; L, luminal space. The arrow indicates the DTN in the lumen adjacent to gut cells.

      Duox Knockdown Reduces B. burgdorferi Persistence in Ticks

      Because our studies revealed that the replication of gut microbes peaks at 20 h of feeding, which also coincides with Duox expression and formation of the DTN in the gut, we next examined whether RNAi-mediated knockdown of Duox could interfere with the occurrence of a DTN and affect the persistence of B. burgdorferi in the gut. Because Duox is a relatively large gene (5,418 bp), to identify the region(s) that are most susceptible to RNAi-mediated knockdown, five different dsRNA preparations that encompass various regions of the gene were generated (Fig. 2A) and empirically tested for their ability to silence target gene expression. One of them (dsRNA2) was selected based on its efficiency in knocking down Duox expression significantly (Fig. 2B, p < 0.01) and used in all subsequent studies. Next we examined whether reduction of Duox expression affects formation of the DTN and whether this influences B. burgdorferi persistence using dsRNA-injected ticks that fed on B. burgdorferi-infected mice. We found that the appearance of the DTN formation was less obvious in Duox knockdown ticks (Fig. 2C), which also retained significantly less B. burgdorferi burden when compared with control (GFP dsRNA-injected) ticks (Fig. 2D, p < 0.01). Similarly, qPCR analysis using 16 S rRNA levels of total gut bacteria also shows an apparent decrease in Duox knockdown ticks (data not shown). Together, these results suggest that the tick Duox plays a beneficial role for the Lyme disease pathogen while feeding ticks acquire it from infected hosts.
      Figure thumbnail gr2
      FIGURE 2.Knockdown of Duox impairs DTN formation and influences persistence of B. burgdorferi in feeding ticks. A, schematic representation of Duox open reading frame showing regions targeted for RNA interference. Nucleotide positions of domains as identified by NCBI conserved domain database search (gray boxes), regions encompassing dsRNA constructs (dsR1–dsR5, red), and detection primers (blue) are shown. Of the dsRNA constructs tested, dsRNA2 (dsR2, spanning nucleotide positions 960–1440, shown in bold type) was most effective and used for subsequent studies. Primers spanning 5′ and 3′ regions of Duox (shown in blue text) were used for detection of transcripts using RT-PCR analysis using agarose gel electrophoresis and quantitative RT-PCR, respectively. B, knockdown of Duox transcripts induced by RNA interference. Nymphal ticks (25/group) were injected with Duox dsRNA or control GFP dsRNA, fed on naïve mice, and collected at 48 h after onset of feeding; isolated guts were processed for measurement of Duox transcripts. The left panel shows RT-PCR analyses using primers spanning either 5′ peroxidase domain region of Duox or a loading control gene, tick β-actin, which are expected to amplify same-sized (400 base pairs) products. The right panel shows quantitative RT-PCR analysis using primers targeting 3′ FAD domain region (A), and the results were normalized against corresponding β-actin transcript levels. Each circle represents an individual tick that was processed and analyzed separately. Compared with ticks microinjected with GFP dsRNA, ticks injected with Duox dsRNA experienced significant down-regulation of Duox expression (p < 0.01). C, Duox knockdown interferes with the formation of the DTN. After 60 h of feeding, ticks were collected, and the guts were dissected. DTN formation was detected using anti-dityrosine antibodies (green) and a nuclear stain, DAPI (blue), and imaged under a confocal microscope. D, knockdown of Duox expression reduces B. burgdorferi persistence in engorged ticks. Ticks were microinjected with Duox dsRNA or GFP dsRNA and allowed to parasitize mice that were infected with B. burgdorferi. After feeding, ticks were collected, and B. burgdorferi burden in ticks was detected by measuring flaB transcripts and normalized with tick β-actin. Each dot represents an individual tick sample. Compared with ticks microinjected with GFP dsRNA, B. burgdorferi burden in ticks was significantly reduced after injection of Duox dsRNA (p < 0.01).

      Identification of a Gut Peroxidase Involved in DTN Formation

      Because previous studies suggested that DTN formation could require participation of a specific gut peroxidase (
      • Kumar S.
      • Molina-Cruz A.
      • Gupta L.
      • Rodrigues J.
      • Barillas-Mury C.
      A peroxidase/dual oxidase system modulates midgut epithelial immunity in Anopheles gambiae.
      ), we next labeled the tick gut using an anti-peroxidase antibody. The results show that peroxidase labeling in the gut readily co-localizes with the DTN labeling (Fig. 3A). We further searched for the existence of a gut peroxidase that is potentially involved in DTN formation. Because the Ixodes genome encodes for at least 16 potential peroxidase proteins, we studied their expression levels in our attempt to shortlist potential candidates. RT-PCR analyses indicated that 11 of 16 peroxidases showed dramatic yet variable levels of transcripts in feeding ticks (Fig. 3B). We further assessed whether their expression is modulated during spirochete infection by comparing corresponding mRNA levels in ticks that fed on B. burgdorferi-infected versus naïve mice. The data show that although expression of a majority of these peroxidases remains unaltered in the presence of B. burgdorferi (data not shown), two of them (annotated as ISCW017368 and ISCW002528) displayed dramatic induction when spirochetes are present in feeding ticks (Fig. 3C, p < 0.01). Subsequently, a measurement of transcript levels of both of these genes suggested their induction at various different time points of tick feeding (Fig. 3, D and E); although mRNA levels of ISCW002528 peak between 6 and 12 h, ISCW017368 is most dramatically expressed between 24 and 36 h.
      Figure thumbnail gr3
      FIGURE 3.Localization and expression of gut peroxidases. A, co-localization of peroxidase and DTN in fed tick gut. Confocal immunofluorescence staining of tick gut at 60 h of feeding was performed using dityrosine antibodies (green), peroxidase antibodies (red), and DAPI. The arrow indicates DTN. EC, epithelial cell; L, luminal space. B, expression of 16 annotated tick peroxidases during feeding. Groups of ticks were collected at 48 h of feeding, and expression of the following 16 peroxidase genes in feeding ticks was detected by RT-PCR: ISCW013767 (lane 1), ISCW018825 (lane 2), ISCW017070 (lane 3), ISCW005828 (lane 4), ISCW022517 (lane 5), ISCW013159 (lane 6), ISCW008495 (lane 7), ISCW002528 (lane 8), ISCW001759 (lane 9), ISCW017368 (lane 10), ISCW022537 (lane 11), ISCW015098 (lane 12), ISCW019584 (lane 13), ISCW024650 (lane 14), ISCW020571 (lane 15), and ISCW020569 (lane 16). C, identification of two peroxidases that are up-regulated during B. burgdorferi infection in ticks. Ticks were allowed to feed on naïve mice or B. burgdorferi-infected mice, collected at 48 h of feeding, and processed for qRT-PCR using gene-specific primers, as indicated in . Although expression of most of the peroxidases did display detectable alteration (data not shown), peroxidase ISCW017368 and peroxidase ISCW002528 were significantly up-regulated in ticks that fed on B. burgdorferi-infected mice, compared with those that fed on naïve mice (p < 0.01). D and E, kinetics of peroxidase expression. Naïve nymphal ticks were collected at different times of feeding on mice or as fully engorged postfed (PF) ticks, and transcript levels of ISCW017368 (D) and ISCW002528 (E) were measured by qRT-PCR. The means ± S.E. of three independent infection experiments are presented.

      Selective Silencing of Peroxidase ISCW017368 Influences DTN Formation and B. burgdorferi Acquisition by Ticks

      Because two of the 16 potential peroxidase enzymes, ISCW017368 and ISCW002528, were induced upon spirochete infection of ticks, we used an RNAi-mediated knockdown strategy to assess their relative importance in DTN formation and the subsequent persistence of B. burgdorferi. To accomplish this, dsRNA preparations targeting each gene were constructed. Injected ticks displayed dramatic down-regulation of corresponding genes as compared with control (GFP dsRNA-injected) ticks (Fig. 4A). Unlike ISCW002528-silenced ticks or control ticks, ISCW017368 knockdown ticks reflected an obvious impairment of DTN integrity (Fig. 4B). Accordingly, silencing of ISCW017368 significantly influenced levels of B. burgdorferi in ticks (Fig. 4C, p < 0.01), whereas ISCW002528 knockdown had no apparent effects (Fig. 4C, p > 0.05), in comparison with corresponding controls. Together, these data suggest that the peroxidase ISCW017368, as well as Duox, plays an important role in the formation of the DTN and influences the persistence of Lyme disease pathogens within ticks.
      Figure thumbnail gr4
      FIGURE 4.Knockdown of peroxidase ISCW017368 but not ISCW02528 impairs DTN formation and influences persistence of B. burgdorferi in feeding ticks. A, RNA interference-mediated knockdown of peroxidase transcripts in feeding ticks. Nymphal ticks (25/group) were injected with dsRNA from the genes corresponding to ISCW017368 or ISCW002528 (indicated as Per017368 and Per002528, respectively) or control (GFP), fed on naïve mice, and collected at 48 h after the onset of feeding. Isolated guts were processed for quantitative RT-PCR using detection primers against target genes, and the results were normalized against tick β-actin. Each circle represents an individual tick that was processed and analyzed separately. Compared with their respective controls (GFP dsRNA), expression of both peroxidases was significantly down-regulated (p < 0.01). B, knockdown of peroxidase Per0017368 impaired formation of DTN. The network was detected in dsRNA-microinjected feeding ticks as detailed in B using anti-dityrosine (green) and DAPI (blue) for labeling tick cell nuclei. The DTN structures were more obvious in ticks injected with dsRNA for peroxidase ISCW02528 or GFP, compared with those injected with peroxidase ISCW017368 dsRNA. EC, epithelial cell; L, luminal space. C, knockdown of peroxidase ISCW017368 transcript levels reduces B. burgdorferi persistence in feeding ticks. Ticks were microinjected with dsRNA for ISCW017368, ISCW002528, or control (GFP) and allowed to parasitize mice that were infected with B. burgdorferi. After feeding, ticks were collected, and B. burgdorferi burden in ticks was detected by measuring flaB transcripts and normalized with tick β-actin. Each dot represents an individual tick sample. No significant differences in B. burgdorferi burdens between ticks injected with peroxidase ISCW02528 and GFP dsRNA were observed (p > 0.05) (right panel). However, compared with ticks microinjected with control (GFP dsRNA), those injected with ISCW017368 dsRNA experienced a significant reduction in B. burgdorferi burden (p < 0.01) (left panel).

      Induction of Nitric-oxide Synthase Potentially Contributes to the Reduction of B. burgdorferi in Ticks

      Because disruption of the DTN barrier could result in a strong pathogen-specific immune response, we sought to assess whether the impaired DTN in Duox knockdown ticks and observed reduction of spirochete levels is due to activation of specific tick innate immune pathway(s) or component(s). We compared the transcript levels of representative potential immune genes in Duox knockdown and control (GFP dsRNA-injected) ticks. Analysis of a selected set of putative immune genes grouped under non-self-recognition, signal transduction, and anti-microbial peptides indicated that none of the tested genes showed significant induction in Duox knockdown ticks; however, induction of a gene encoding nitric-oxide synthase (NOS) was observed (Fig. 5A, p < 0.05). Because B. burgdorferi responsiveness to NO-mediated killing (
      • Ma Y.
      • Seiler K.P.
      • Tai K.F.
      • Yang L.
      • Woods M.
      • Weis J.J.
      Outer surface lipoproteins of Borrelia burgdorferi stimulate nitric oxide production by the cytokine-inducible pathway.
      ,
      • Modolell M.
      • Schaible U.E.
      • Rittig M.
      • Simon M.M.
      Killing of Borrelia burgdorferi by macrophages is dependent on oxygen radicals and nitric oxide and can be enhanced by antibodies to outer surface proteins of the spirochete.
      ) and the anti-parasitic effects of NOS has been documented in DTN-silenced mosquitoes (
      • Kumar S.
      • Molina-Cruz A.
      • Gupta L.
      • Rodrigues J.
      • Barillas-Mury C.
      A peroxidase/dual oxidase system modulates midgut epithelial immunity in Anopheles gambiae.
      ), the I. scapularis NOS gene was further confirmed for its up-regulation in additional groups of B. burgdorferi-infected Duox or peroxidase ISCW017368 knockdown ticks (Fig. 5B), which also reflected an augmentation of total NOS enzymatic activity (Fig. 5C, p < 0.05). Together, these results indicate that the enhancement of potential NO production by NOS could contribute to the destruction of B. burgdorferi following impairment of the DTN in ticks.
      Figure thumbnail gr5
      FIGURE 5.Knockdown of Duox and peroxidase ISCW017368 induces nitric-oxide synthase. A, expression of representative genes potentially related to innate immune responses in Duox knockdown ticks. Naïve ticks were microinjected with GFP dsRNA (white bars) or Duox dsRNA (black bars), as detailed in . Transcript levels for selected genes were measured using qRT-PCR analysis at 48 h of feeding. Although mRNA levels of most genes remained unaltered (p > 0.05), expression of the NOS gene was significantly up-regulated compared with that of corresponding control (GFP dsRNA) (p < 0.05). B, effects of Duox or peroxidase ISCW017368 knockdown on NOS gene expression during tick feeding on B. burgdorferi-infected mouse. Knockdown of corresponding Duox or peroxidase ISCW017368 gene expression was performed as detailed in A and significantly induced NOS mRNA expression in ticks (p < 0.05). C, enzymatic activity of NOS. Activity of NOS was measured in ticks microinjected with either Duox dsRNA (left panel) or peroxidase ISCW017368 dsRNA (right panel), reflecting significant enhancement of NOS enzyme activity compared with ticks injected with GFP dsRNA (p < 0.05).

      DISCUSSION

      B. burgdorferi thrives in nature though an intricate tick-mammal infection cycle (
      • Radolf J.D.
      • Caimano M.J.
      • Stevenson B.
      • Hu L.T.
      Of ticks, mice and men: understanding the dual-host lifestyle of Lyme disease spirochaetes.
      ,
      • Pal U.
      • Fikrig E.
      Adaptation of Borrelia burgdorferi in the vector and vertebrate host.
      ). Notwithstanding an intense research focus on Lyme borreliosis, precisely how the pathogen successfully persists in and is transmitted through ticks, despite a potentially rigorous arthropod innate immune response against invading microorganisms (
      • Govind S.
      • Nehm R.H.
      Innate immunity in fruit flies: a textbook example of genomic recycling.
      • Hoffmann J.A.
      • Reichhart J.M.
      Drosophila innate immunity: an evolutionary perspective.
      ,
      • Tanji T.
      • Hu X.
      • Weber A.N.
      • Ip Y.T.
      Toll and IMD pathways synergistically activate an innate immune response in Drosophila melanogaster.
      ,
      • Tanji T.
      • Ip Y.T.
      Regulators of the Toll and Imd pathways in the Drosophila innate immune response.
      ,
      • Valanne S.
      • Wang J.H.
      • Rämet M.
      The Drosophila Toll signaling pathway.
      ), remains enigmatic. Here, we present evidence that a molecular barrier, termed dityrosine network (DTN) exists in ticks that could represent a protective strategy to preserve homeostasis of gut commensals that replicate during feeding. Homologous to peritrophic matrix described in the gut of most insects including ticks (
      • Kariu T.
      • Smith A.
      • Yang X.
      • Pal U.
      A chitin deacetylase-like protein is a predominant constituent of tick peritrophic membrane that influences the persistence of Lyme disease pathogens within the vector.
      ,
      • Zhu Z.
      • Gern L.
      • Aeschlimann A.
      The peritrophic membrane of Ixodes ricinus.
      ), this molecular wall is normally synthesized in response to blood feeding and creates a protective barrier between luminal contents and underlying gut epithelial cells. In Drosophila, the matrix is part of a protective innate immune response because in its absence, increased susceptibility to bacterial toxins is observed (
      • Ha E.M.
      • Oh C.T.
      • Bae Y.S.
      • Lee W.J.
      A direct role for dual oxidase in Drosophila gut immunity.
      ). In contrast, as shown in A. gambiae earlier (
      • Kumar S.
      • Molina-Cruz A.
      • Gupta L.
      • Rodrigues J.
      • Barillas-Mury C.
      A peroxidase/dual oxidase system modulates midgut epithelial immunity in Anopheles gambiae.
      ), formation of the DTN in ticks also likely decreases the gut permeability to immune elicitors, thereby inadvertently also protecting pathogens like B. burgdorferi. We show that DTN formation in I. scapularis ticks required participation of an NADPH oxidase, a tick Duox, along with a specific gut peroxidase. Together, our results highlight the paradigm of a less well known yet critical function of a NADPH oxidase system in protein cross-linking activity related to diverse cellular functions that specifically range from cross-linking of the extracellular matrix or cuticular proteins in Caenorhabditis elegans to eggshell hardening or epithelial barrier (DTN) functions in insects.
      Originally identified in the thyroid, Duox enzymes have been shown to promote formation of dityrosine linkages (
      • Donkó A.
      • Péterfi Z.
      • Sum A.
      • Leto T.
      • Geiszt M.
      Dual oxidases.
      ). In mammals, formation of these bonds relies upon heme peroxidase activity, such as thyroperoxidase or myeloperoxidase, utilizing hydrogen peroxide often formed from NADPH oxidase activity. However, Duox contains an extracellular peroxidase domain, which appears to bind heme and serve as a functional peroxidase in C. elegans (
      • Edens W.A.
      • Sharling L.
      • Cheng G.
      • Shapira R.
      • Kinkade J.M.
      • Lee T.
      • Edens H.A.
      • Tang X.
      • Sullards C.
      • Flaherty D.B.
      • Benian G.M.
      • Lambeth J.D.
      Tyrosine cross-linking of extracellular matrix is catalyzed by Duox, a multidomain oxidase/peroxidase with homology to the phagocyte oxidase subunit gp91phox.
      ,
      • Meitzler J.L.
      • Ortiz de Montellano P.R.
      Caenorhabditis elegans and human dual oxidase 1 (DUOX1) “peroxidase” domains: insights into heme binding and catalytic activity.
      ). Mutations in the peroxidase domain of C. elegans Duox lead to a blistering phenotype caused by loss of protein cross-linking, without impaired ROS generation and host defense dependent upon Duox (
      • Chávez V.
      • Mohri-Shiomi A.
      • Garsin D.A.
      Ce-Duox1/BLI-3 generates reactive oxygen species as a protective innate immune mechanism in Caenorhabditis elegans.
      ). The same domain in human Duox, however, does not bind heme nor exert peroxidase or tyrosine cross-linking activity (
      • Meitzler J.L.
      • Ortiz de Montellano P.R.
      Caenorhabditis elegans and human dual oxidase 1 (DUOX1) “peroxidase” domains: insights into heme binding and catalytic activity.
      ). Our sequence analysis data (Fig. 6) suggest that Duox enzymes in mosquito and tick are also well conserved; however, interestingly, they lack a key Glu (Glu-238) residue that, when mutated in C. elegans, impairs heme binding (
      • Meitzler J.L.
      • Brandman R.
      • Ortiz de Montellano P.R.
      Perturbed heme binding is responsible for the blistering phenotype associated with mutations in the6 Caenorhabditis elegans dual oxidase 1 (DUOX1) peroxidase domain.
      ). Notably, that mutated protein still has residual peroxidase activity and can mediate tyrosine cross-linking in C. elegans (
      • Meitzler J.L.
      • Brandman R.
      • Ortiz de Montellano P.R.
      Perturbed heme binding is responsible for the blistering phenotype associated with mutations in the6 Caenorhabditis elegans dual oxidase 1 (DUOX1) peroxidase domain.
      ). Therefore, we speculate that arthropod Duox may retain some peroxidase/cross-linking function, and low levels of dityrosine immunoreactivity remains in ISCW017368 knockdown ticks. Nevertheless, it appears that both Duox and a separate heme peroxidase are required for proper DTN formation in both mosquito and tick gut, although a direct link between these two enzymes is not established in our study. However, as our expression analysis and gene silencing studies indicated, among the 11 peroxidases that are expressed in feeding ticks and that are induced during B. burgdorferi infection, only one (ISCW017368) participated in DTN formation. Therefore, we speculate that although I. scapularis produces several peroxidases that are involved in unknown aspects of tick physiology, only one with potential heme peroxidase activity might have co-evolved with Duox to participate in the formation of DTN.
      Figure thumbnail gr6
      FIGURE 6.Amino acid sequence alignment of potential peroxidase domain of I. scapularis Duox with orthologs from other model nematode and arthropod species. Partial amino acid sequences (annotations are shown according to NCBI protein sequences) from I. scapularis (EEC10543.1), C. elegans (O61213.2), Drosophila melanogaster (AAF51201.2), and A. gambiae (XP_319115.4) were aligned using Cobalt Constraint-based Multiple Protein Alignment Tool (NCBI). The key residues for heme binding and peroxidase function are identified based upon previous studies on Duox peroxidase domains (PMID 20947510; PMID 19460756), and their conservations or deviations are indicated with colored letters and boxes. Alteration of a key glutamic acid residue (Glu-238) in C. elegans that is important of heme binding (
      • Meitzler J.L.
      • Ortiz de Montellano P.R.
      Caenorhabditis elegans and human dual oxidase 1 (DUOX1) “peroxidase” domains: insights into heme binding and catalytic activity.
      ,
      • Meitzler J.L.
      • Brandman R.
      • Ortiz de Montellano P.R.
      Perturbed heme binding is responsible for the blistering phenotype associated with mutations in the6 Caenorhabditis elegans dual oxidase 1 (DUOX1) peroxidase domain.
      ) in I. scapularis or other arthropods is indicated in red text.
      The Ixodes genome is relatively large and highly redundant; although it encodes a single Duox, there are at least 16 potential peroxidases, 8 of which are annotated as heme peroxidases, 4 as glutathione peroxidases, 3 as phospholipid-hydroperoxide glutathione, 1 as thioredoxin peroxidase, and 1 as cAMP-regulated phosphoprotein/endosulfine. Although each of these enzymes could play individual and potentially important roles in tick physiology and immunity, 11 are dramatically expressed in feeding ticks. Considering further up-regulation of two of these in the presence of B. burgdorferi, the functions of peroxidase ISCW002528 and peroxidase ISCW017368 could certainly be related to vector immunity. In fact, our data indicate that the latter, which is annotated as a heme peroxidase, is an indispensible partner of the tick Duox system essential for DTN formation. Interestingly, although the molecular basis of DTN formation remains unknown, in mosquitoes, the participation of a heme peroxidase is also required (
      • Kumar S.
      • Molina-Cruz A.
      • Gupta L.
      • Rodrigues J.
      • Barillas-Mury C.
      A peroxidase/dual oxidase system modulates midgut epithelial immunity in Anopheles gambiae.
      ). Thus, our results underscore the importance of this specific class of peroxidases in tyrosine cross-linking critical for DTN formation. The blistering phenotype of peroxidase domain mutants in C. elegans Duox suggests the lack of additional peroxidase partner contribution to protein cross-linking in the nematode (
      • Edens W.A.
      • Sharling L.
      • Cheng G.
      • Shapira R.
      • Kinkade J.M.
      • Lee T.
      • Edens H.A.
      • Tang X.
      • Sullards C.
      • Flaherty D.B.
      • Benian G.M.
      • Lambeth J.D.
      Tyrosine cross-linking of extracellular matrix is catalyzed by Duox, a multidomain oxidase/peroxidase with homology to the phagocyte oxidase subunit gp91phox.
      ,
      • Chávez V.
      • Mohri-Shiomi A.
      • Garsin D.A.
      Ce-Duox1/BLI-3 generates reactive oxygen species as a protective innate immune mechanism in Caenorhabditis elegans.
      ). However, a peroxidase (MLT-7) has been identified in C. elegans, in which loss of expression leads to significant decreases in tyrosine cross-linking of cuticle proteins and cause blistering (
      • Thein M.C.
      • Winter A.D.
      • Stepek G.
      • McCormack G.
      • Stapleton G.
      • Johnstone I.L.
      • Page A.P.
      Combined extracellular matrix cross-linking activity of the peroxidase MLT-7 and the dual oxidase BLI-3 is critical for post-embryonic viability in Caenorhabditis elegans.
      ,
      • Moribe H.
      • Konakawa R.
      • Koga D.
      • Ushiki T.
      • Nakamura K.
      • Mekada E.
      Tetraspanin is required for generation of reactive oxygen species by the dual oxidase system in Caenorhabditis elegans.
      ). Taken together, the data suggest that although Duox in higher organisms exhibit the absence or loss of tyrosine cross-linking activity, separate heme peroxidases have evolved to complement this function of Duox to mediate or enhance DTN formation, extracellular matrix modification, or host defenses from ROS.
      Studies involving mucosal surfaces in mammalian models and Drosophila gut suggest that ROS derived from Duox play an antimicrobial role (
      • Donkó A.
      • Péterfi Z.
      • Sum A.
      • Leto T.
      • Geiszt M.
      Dual oxidases.
      ). However, our data and that in mosquito (
      • Kumar S.
      • Molina-Cruz A.
      • Gupta L.
      • Rodrigues J.
      • Barillas-Mury C.
      A peroxidase/dual oxidase system modulates midgut epithelial immunity in Anopheles gambiae.
      ) suggest that Duox does not exert microbicidal effects in the arthropod gut, because knockdown inhibits bacterial burden. A major fraction of Plasmodium parasites were shown to be killed by a vigorous mosquito innate immune response, yet the existence of an analogous phenomenon in ticks, particularly pertaining to the control of B. burgdorferi levels in the gut, is unknown. We show that transcript levels of NOS or their enzymatic activity in Duox knockdown ticks are significantly enhanced, which may be linked to a potential NO-mediated borreliacidal response. Although it has been suggested to play an insignificant role, particularly in murine host immune response against B. burgdorferi, NO possesses potent borreliacidal properties against cultured spirochetes and thus could be responsible for the reduction of borrelial numbers in fed ticks. Our attempts to study the possible involvement of NOS in the generation of borreliacidal responses in ticks and assess whether RNAi or in vivo inhibitor-mediated interference of NOS reverses the deleterious effect of Duox silencing on B. burgdorferi survival remained unsuccessful but could provide clues for the biological significance of NOS up-regulation in ticks with impaired DTNs. Nevertheless, NO production by intestinal epithelial cells has been directly linked to the immune response of the vector to other enteric pathogens, including DTN-silenced mosquitoes, and thus likely represents a conserved microbicidal response in diverse arthropod species.
      In summary, here we present evidence that a molecular barrier, recently proposed in studies using malarial vectors, termed DTN, and catalyzed by a Duox/peroxidase system, allows B. burgdorferi potentially to proliferate without activating epithelial immunity, such as the NOS enzyme. Arthropod genes that are temporally induced in particular vector tissues could be functionally relevant for B. burgdorferi persistence in vivo. If any such gene products play beneficial roles in pathogen persistence and/or transmission, this information could be used to develop novel preventive measures to interfere with the spirochete infection cycle. Our data show that proteins critical for formation of the DTN, the NADPH oxidase, or Duox, as well as a heme peroxidase protein, are inducible upon B. burgdorferi infection and support their persistence in the vector. Gene products involved in innate immunity are conserved across species; however, some are unique to a given species or markedly diversified, which may reflect adaptation to different pathogens, thus presenting an opportunity to assess their roles as immunoprotective antigens. Together, this information will help clarify the biology of tick immune responses, will provide insights about adaptive strategies of a bacterium that persists in a diverse array of host and vector tissues, and may contribute to new targets for the disruption of the spirochete life cycle.

      Acknowledgment

      We sincerely thank Faith Kung for assistance with this study.

      Supplementary Material

      References

        • Stanek G.
        • Wormser G.P.
        • Gray J.
        • Strle F.
        Lyme borreliosis.
        Lancet. 2012; 379: 461-473
        • Steere A.C.
        Lyme disease.
        N. Engl. J. Med. 2001; 345: 115-125
        • Radolf J.D.
        • Caimano M.J.
        • Stevenson B.
        • Hu L.T.
        Of ticks, mice and men: understanding the dual-host lifestyle of Lyme disease spirochaetes.
        Nat. Rev. Microbiol. 2012; 10: 87-99
        • Piesman J.
        • Oliver J.R.
        • Sinsky R.J.
        Growth kinetics of the Lyme disease spirochete (Borrelia burgdorferi) in vector ticks (Ixodes dammini).
        Am. J. Trop. Med. Hyg. 1990; 42: 352-357
        • Tyson K.R.
        • Piesman J.
        Lyme disease spirochete-tick-host interactions.
        Adv. Insect Physiol. 2009; 37: 243-296
        • de Silva A.M.
        • Tyson K.R.
        • Pal U.
        Molecular characterization of the tick-Borrelia interface.
        Front. Biosci. (Landmark Ed.). 2009; 14: 3051-3063
        • Pal U.
        • Fikrig E.
        Tick interactions.
        in: Samuels D.S. Radolf J.D. Borrelia, Molecular Biology, Host Interaction and Pathogenesis. Caister Academic Press, Norfolk, UK2010: 279-298
        • Dong Y.
        • Aguilar R.
        • Xi Z.
        • Warr E.
        • Mongin E.
        • Dimopoulos G.
        Anopheles gambiae immune responses to human and rodent Plasmodium parasite species.
        PLoS Pathog. 2006; 2: e52
        • Munderloh U.G.
        • Kurtti T.J.
        Cellular and molecular interrelationships between ticks and prokaryotic tick-borne pathogens.
        Annu. Rev. Entomol. 1995; 40: 221-243
        • Yassine H.
        • Osta M.A.
        Anopheles gambiae innate immunity.
        Cell Microbiol. 2010; 12: 1-9
        • Sonenshine D.E.
        Biology of Ticks. Oxford University Press, New York1993
        • Kopácek P.
        • Hajdusek O.
        • Buresová V.
        • Daffre S.
        Tick innate immunity.
        Adv. Exp. Med. Biol. 2010; 708: 137-162
        • Sonenshine D.E.
        • Hynes W.L.
        • Ceraul S.M.
        • Mitchell R.
        • Benzine T.
        Host blood proteins and peptides in the midgut of the tick Dermacentor variabilis contribute to bacterial control.
        Exp. Appl. Acarol. 2005; 36: 207-223
        • Govind S.
        • Nehm R.H.
        Innate immunity in fruit flies: a textbook example of genomic recycling.
        PLoS Biol. 2004; 2: E276
        • Hoffmann J.A.
        • Reichhart J.M.
        Drosophila innate immunity: an evolutionary perspective.
        Nat. Immunol. 2002; 3: 121-126
        • Tanji T.
        • Hu X.
        • Weber A.N.
        • Ip Y.T.
        Toll and IMD pathways synergistically activate an innate immune response in Drosophila melanogaster.
        Mol. Cell. Biol. 2007; 27: 4578-4588
        • Tanji T.
        • Ip Y.T.
        Regulators of the Toll and Imd pathways in the Drosophila innate immune response.
        Trends Immunol. 2005; 26: 193-198
        • Valanne S.
        • Wang J.H.
        • Rämet M.
        The Drosophila Toll signaling pathway.
        J. Immunol. 2011; 186: 649-656
        • Hynes W.L.
        • Ceraul S.M.
        • Todd S.M.
        • Seguin K.C.
        • Sonenshine D.E.
        A defensin-like gene expressed in the black-legged tick, Ixodes scapularis.
        Med. Vet. Entomol. 2005; 19: 339-344
        • Rudenko N.
        • Golovchenko M.
        • Edwards M.J.
        • Grubhoffer L.
        Differential expression of Ixodes ricinus tick genes induced by blood feeding or Borrelia burgdorferi infection.
        J. Med. Entomol. 2005; 42: 36-41
        • Anderson J.M.
        • Sonenshine D.E.
        • Valenzuela J.G.
        Exploring the mialome of ticks: an annotated catalogue of midgut transcripts from the hard tick, Dermacentor variabilis (Acari: Ixodidae).
        BMC Genomics. 2008; 9: 552
        • Ribeiro J.M.
        • Alarcon-Chaidez F.
        • Francischetti I.M.
        • Mans B.J.
        • Mather T.N.
        • Valenzuela J.G.
        • Wikel S.K.
        An annotated catalog of salivary gland transcripts from Ixodes scapularis ticks.
        Insect Biochem. Mol. Biol. 2006; 36: 111-129
        • Kumar S.
        • Molina-Cruz A.
        • Gupta L.
        • Rodrigues J.
        • Barillas-Mury C.
        A peroxidase/dual oxidase system modulates midgut epithelial immunity in Anopheles gambiae.
        Science. 2010; 327: 1644-1648
        • Donkó A.
        • Péterfi Z.
        • Sum A.
        • Leto T.
        • Geiszt M.
        Dual oxidases.
        Philos. Trans. R. Soc. Lond. B Biol. Sci. 2005; 360: 2301-2308
        • Ha E.M.
        • Oh C.T.
        • Bae Y.S.
        • Lee W.J.
        A direct role for dual oxidase in Drosophila gut immunity.
        Science. 2005; 310: 847-850
        • Geiszt M.
        • Witta J.
        • Baffi J.
        • Lekstrom K.
        • Leto T.L.
        Dual oxidases represent novel hydrogen peroxide sources supporting mucosal surface host defense.
        FASEB J. 2003; 17: 1502-1504
        • Bae Y.S.
        • Choi M.K.
        • Lee W.J.
        Dual oxidase in mucosal immunity and host-microbe homeostasis.
        Trends Immunol. 2010; 31: 278-287
        • Elias A.F.
        • Stewart P.E.
        • Grimm D.
        • Caimano M.J.
        • Eggers C.H.
        • Tilly K.
        • Bono J.L.
        • Akins D.R.
        • Radolf J.D.
        • Schwan T.G.
        • Rosa P.
        Clonal polymorphism of Borrelia burgdorferi strain B31 MI: implications for mutagenesis in an infectious strain background.
        Infect. Immun. 2002; 70: 2139-2150
        • Kariu T.
        • Coleman A.S.
        • Anderson J.F.
        • Pal U.
        Methods for rapid transfer and localization of Lyme disease pathogens within the tick gut.
        J. Vis. Exp. 2011; 48: 2544
        • Kariu T.
        • Smith A.
        • Yang X.
        • Pal U.
        A chitin deacetylase-like protein is a predominant constituent of tick peritrophic membrane that influences the persistence of Lyme disease pathogens within the vector.
        PLoS One. 2013; 8: e78376
        • Yang X.
        • Coleman A.S.
        • Anguita J.
        • Pal U.
        A chromosomally encoded virulence factor protects the Lyme disease pathogen against host-adaptive immunity.
        PLoS Pathog. 2009; 5: e1000326
        • Pal U.
        • Li X.
        • Wang T.
        • Montgomery R.R.
        • Ramamoorthi N.
        • Desilva A.M.
        • Bao F.
        • Yang X.
        • Pypaert M.
        • Pradhan D.
        • Kantor F.S.
        • Telford S.
        • Anderson J.F.
        • Fikrig E.
        TROSPA, an Ixodes scapularis receptor for Borrelia burgdorferi.
        Cell. 2004; 119: 457-468
        • Ghigo D.
        • Riganti C.
        • Gazzano E.
        • Costamagna C.
        • Bosia A.
        Cycling of NADPH by glucose 6-phosphate dehydrogenase optimizes the spectrophotometric assay of nitric oxide synthase activity in cell lysates.
        Nitric Oxide. 2006; 15: 148-153
        • van Overbeek L.
        • Gassner F.
        • van der Plas C.L.
        • Kastelein P.
        • Nunes-da Rocha U.
        • Takken W.
        Diversity of Ixodes ricinus tick-associated bacterial communities from different forests.
        FEMS Microbiol. Ecol. 2008; 66: 72-84
        • Moreno C.X.
        • Moy F.
        • Daniels T.J.
        • Godfrey H.P.
        • Cabello F.C.
        Molecular analysis of microbial communities identified in different developmental stages of Ixodes scapularis ticks from Westchester and Dutchess Counties, New York.
        Environ. Microbiol. 2006; 8: 761-772
        • Ma Y.
        • Seiler K.P.
        • Tai K.F.
        • Yang L.
        • Woods M.
        • Weis J.J.
        Outer surface lipoproteins of Borrelia burgdorferi stimulate nitric oxide production by the cytokine-inducible pathway.
        Infect Immun. 1994; 62: 3663-3671
        • Modolell M.
        • Schaible U.E.
        • Rittig M.
        • Simon M.M.
        Killing of Borrelia burgdorferi by macrophages is dependent on oxygen radicals and nitric oxide and can be enhanced by antibodies to outer surface proteins of the spirochete.
        Immunol. Lett. 1994; 40: 139-146
        • Pal U.
        • Fikrig E.
        Adaptation of Borrelia burgdorferi in the vector and vertebrate host.
        Microbes Infect. 2003; 5: 659-666
        • Zhu Z.
        • Gern L.
        • Aeschlimann A.
        The peritrophic membrane of Ixodes ricinus.
        Parasitol. Res. 1991; 77: 635-641
        • Edens W.A.
        • Sharling L.
        • Cheng G.
        • Shapira R.
        • Kinkade J.M.
        • Lee T.
        • Edens H.A.
        • Tang X.
        • Sullards C.
        • Flaherty D.B.
        • Benian G.M.
        • Lambeth J.D.
        Tyrosine cross-linking of extracellular matrix is catalyzed by Duox, a multidomain oxidase/peroxidase with homology to the phagocyte oxidase subunit gp91phox.
        J. Cell Biol. 2001; 154: 879-891
        • Meitzler J.L.
        • Ortiz de Montellano P.R.
        Caenorhabditis elegans and human dual oxidase 1 (DUOX1) “peroxidase” domains: insights into heme binding and catalytic activity.
        J. Biol. Chem. 2009; 284: 18634-18643
        • Chávez V.
        • Mohri-Shiomi A.
        • Garsin D.A.
        Ce-Duox1/BLI-3 generates reactive oxygen species as a protective innate immune mechanism in Caenorhabditis elegans.
        Infect. Immun. 2009; 77: 4983-4989
        • Meitzler J.L.
        • Brandman R.
        • Ortiz de Montellano P.R.
        Perturbed heme binding is responsible for the blistering phenotype associated with mutations in the6 Caenorhabditis elegans dual oxidase 1 (DUOX1) peroxidase domain.
        J. Biol. Chem. 2010; 285: 40991-41000
        • Thein M.C.
        • Winter A.D.
        • Stepek G.
        • McCormack G.
        • Stapleton G.
        • Johnstone I.L.
        • Page A.P.
        Combined extracellular matrix cross-linking activity of the peroxidase MLT-7 and the dual oxidase BLI-3 is critical for post-embryonic viability in Caenorhabditis elegans.
        J. Biol. Chem. 2009; 284: 17549-17563
        • Moribe H.
        • Konakawa R.
        • Koga D.
        • Ushiki T.
        • Nakamura K.
        • Mekada E.
        Tetraspanin is required for generation of reactive oxygen species by the dual oxidase system in Caenorhabditis elegans.
        PLoS Genet. 2012; 8: e1002957