Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 120(20): e2208673120, 2023 05 16.
Article in English | MEDLINE | ID: mdl-37155900

ABSTRACT

The immune deficiency (IMD) pathway directs host defense in arthropods upon bacterial infection. In Pancrustacea, peptidoglycan recognition proteins sense microbial moieties and initiate nuclear factor-κB-driven immune responses. Proteins that elicit the IMD pathway in non-insect arthropods remain elusive. Here, we show that an Ixodes scapularis homolog of croquemort (Crq), a CD36-like protein, promotes activation of the tick IMD pathway. Crq exhibits plasma membrane localization and binds the lipid agonist 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoglycerol. Crq regulates the IMD and jun N-terminal kinase signaling cascades and limits the acquisition of the Lyme disease spirochete B. burgdorferi. Additionally, nymphs silenced for crq display impaired feeding and delayed molting to adulthood due to a deficiency in ecdysteroid synthesis. Collectively, we establish a distinct mechanism for arthropod immunity outside of insects and crustaceans.


Subject(s)
Arthropods , Bacterial Infections , Borrelia burgdorferi , Ixodes , Lyme Disease , Animals , Ixodes/microbiology , Borrelia burgdorferi/genetics , NF-kappa B , Lyme Disease/microbiology
2.
J Infect Dis ; 225(1): 135-145, 2022 01 05.
Article in English | MEDLINE | ID: mdl-34139755

ABSTRACT

Human babesiosis is an emerging tick-borne malaria-like illness caused by Babesia parasites following their development in erythrocytes. Here, we show that a mutation in the Babesia microti mitochondrial cytochrome b (Cytb) that confers resistance to the antibabesial drug ELQ-502 decreases parasite fitness in the arthropod vector. Interestingly, whereas the mutant allele does not affect B. microti fitness during the mammalian blood phase of the parasite life cycle and is genetically stable as parasite burden increases, ELQ-502-resistant mutant parasites developing in the tick vector are genetically unstable with a high rate of the wild-type allele emerging during the nymphal stage. Furthermore, we show that B. microti parasites with this mutation are transmitted from the tick to the host, raising the possibility that the frequency of Cytb resistance mutations may be decreased by passage through the tick vector, but could persist in the environment if present when ticks feed.


Subject(s)
Antiprotozoal Agents/pharmacology , Babesia/genetics , Babesiosis/drug therapy , Babesiosis/transmission , Cytochromes b/genetics , Drug Resistance/genetics , Ixodes , Quinolones/pharmacology , Ticks , Animals , Babesia/drug effects , Babesia/growth & development , Babesiosis/diagnosis , Cytochromes b/drug effects , Erythrocytes/parasitology , Humans , Mutation , Parasites
3.
Parasite Immunol ; 43(5): e12808, 2021 05.
Article in English | MEDLINE | ID: mdl-33187012

ABSTRACT

Acquired tick resistance is a phenomenon wherein the host elicits an immune response against tick salivary components upon repeated tick infestations. The immune responses, potentially directed against critical salivary components, thwart tick feeding, and the animal becomes resistant to subsequent tick infestations. The development of tick resistance is frequently observed when ticks feed on non-natural hosts, but not on natural hosts. The molecular mechanisms that lead to the development of tick resistance are not fully understood, and both host and tick factors are invoked in this phenomenon. Advances in molecular tools to address the host and the tick are beginning to reveal new insights into this phenomenon and to uncover a deeper understanding of the fundamental biology of tick-host interactions. This review will focus on the expanding understanding of acquired tick resistance and highlight the impact of this understanding on anti-tick vaccine development efforts.


Subject(s)
Proteome/physiology , Tick Infestations/immunology , Ticks/physiology , Animals , Disease Models, Animal , Disease Resistance , Host-Parasite Interactions/immunology , Humans
4.
Infect Immun ; 88(12)2020 11 16.
Article in English | MEDLINE | ID: mdl-32928964

ABSTRACT

Borrelia burgdorferi causes Lyme disease, the most common tick-transmitted illness in North America. When Ixodes scapularis feed on an infected vertebrate host, spirochetes enter the tick gut along with the bloodmeal and colonize the vector. Here, we show that a secreted tick protein, I. scapularisprotein disulfide isomerase A3 (IsPDIA3), enhances B. burgdorferi colonization of the tick gut. I. scapularis ticks in which ispdiA3 has been knocked down using RNA interference have decreased spirochete colonization of the tick gut after engorging on B. burgdorferi-infected mice. Moreover, administration of IsPDIA3 antiserum to B. burgdorferi-infected mice reduced the ability of spirochetes to colonize the tick when feeding on these animals. We show that IsPDIA3 modulates inflammatory responses at the tick bite site, potentially facilitating spirochete survival at the vector-host interface as it exits the vertebrate host to enter the tick gut. These data provide functional insights into the complex interactions between B. burgdorferi and its arthropod vector and suggest additional targets to interfere with the spirochete life cycle.


Subject(s)
Borrelia burgdorferi/physiology , Ixodes/metabolism , Lyme Disease/transmission , Protein Disulfide-Isomerases/metabolism , Amino Acid Sequence , Animals , Arachnid Vectors/microbiology , Cell Line , Gene Knockdown Techniques , Humans , Immunity, Humoral , Inflammation/enzymology , Inflammation/genetics , Inflammation/metabolism , Ixodes/enzymology , Ixodes/genetics , Membrane Proteins/metabolism , Mice , Phylogeny , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/immunology , RNA Interference , Recombinant Proteins , Sequence Alignment , Spirochaetales/physiology
5.
Clin Infect Dis ; 70(8): 1768-1773, 2020 04 10.
Article in English | MEDLINE | ID: mdl-31620776

ABSTRACT

Lyme disease, caused by some Borrelia burgdorferi sensu lato, is the most common tick-borne illness in the Northern Hemisphere and the number of cases, and geographic spread, continue to grow. Previously identified B. burgdorferi proteins, lipid immunogens, and live mutants lead the design of canonical vaccines aimed at disrupting infection in the host. Discovery of the mechanism of action of the first vaccine catalyzed the development of new strategies to control Lyme disease that bypassed direct vaccination of the human host. Thus, novel prevention concepts center on proteins produced by B. burgdorferi during tick transit and on tick proteins that mediate feeding and pathogen transmission. A burgeoning area of research is tick immunity as it can unlock mechanistic pathways that could be targeted for disruption. Studies that shed light on the mammalian immune pathways engaged during tick-transmitted B. burgdorferi infection would further development of vaccination strategies against Lyme disease.


Subject(s)
Borrelia burgdorferi , Ixodes , Lyme Disease , Ticks , Vaccines , Animals , Humans , Lyme Disease/prevention & control , Vaccination
6.
Proc Natl Acad Sci U S A ; 114(5): E781-E790, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28096373

ABSTRACT

Arthropods transmit diverse infectious agents; however, the ways microbes influence their vector to enhance colonization are poorly understood. Ixodes scapularis ticks harbor numerous human pathogens, including Anaplasma phagocytophilum, the agent of human granulocytic anaplasmosis. We now demonstrate that A. phagocytophilum modifies the I. scapularis microbiota to more efficiently infect the tick. A. phagocytophilum induces ticks to express Ixodes scapularis antifreeze glycoprotein (iafgp), which encodes a protein with several properties, including the ability to alter bacterial biofilm formation. IAFGP thereby perturbs the tick gut microbiota, which influences the integrity of the peritrophic matrix and gut barrier-critical obstacles for Anaplasma colonization. Mechanistically, IAFGP binds the terminal d-alanine residue of the pentapeptide chain of bacterial peptidoglycan, resulting in altered permeability and the capacity of bacteria to form biofilms. These data elucidate the molecular mechanisms by which a human pathogen appropriates an arthropod antibacterial protein to alter the gut microbiota and more effectively colonize the vector.


Subject(s)
Anaplasma phagocytophilum/physiology , Gastrointestinal Microbiome , Host-Pathogen Interactions , Ixodes/microbiology , Animals , Antifreeze Proteins/metabolism , Arthropod Proteins/metabolism , Ehrlichiosis , Mice , Peptidoglycan/metabolism
7.
J Immunol ; 196(10): 4185-95, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27076681

ABSTRACT

Borrelia miyamotoi is a relapsing fever spirochete in Ixodes ticks that has been recently identified as a human pathogen causing hard tick-borne relapsing fever (HTBRF) across the Northern Hemisphere. No validated serologic test exists, and current serologic assays have low sensitivity in early HTBRF. To examine the humoral immune response against B. miyamotoi, we infected C3H/HeN mice with B. miyamotoi strain LB-2001 expressing variable small protein 1 (Vsp1) and demonstrated that spirochetemia was cleared after 3 d, coinciding with anti-Vsp1 IgM production. Clearance was also observed after passive transfer of immune sera to infected SCID mice. Next, we showed that anti-Vsp1 IgG eliminates Vsp1-expressing B. miyamotoi, selecting for spirochetes expressing a variable large protein (VlpC2) resistant to anti-Vsp1. The viability of Asian isolate B. miyamotoi HT31, expressing Vlp15/16 and Vlp18, was also unaffected by anti-Vsp1. Finally, in nine HTBRF patients, we demonstrated IgM reactivity to Vsp1 in two and against Vlp15/16 in four ∼1 wk after these patients tested positive for B. miyamotoi by PCR. Our data show that B. miyamotoi is able to express various variable major proteins (VMPs) to evade humoral immunity and that VMPs are antigenic in humans. We propose that serologic tests based on VMPs are of additional value in diagnosing HTBRF.


Subject(s)
Antibodies, Bacterial/immunology , Antibody Formation , Bacterial Outer Membrane Proteins/immunology , Lipoproteins/immunology , Relapsing Fever/immunology , Amino Acid Sequence , Animals , Antibodies, Bacterial/blood , Base Sequence , Borrelia/immunology , Female , Humans , Immunization, Passive , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin M/blood , Immunoglobulin M/immunology , Mice , Mice, Inbred C3H , Mice, SCID , Protein Structure, Tertiary
8.
PLoS Pathog ; 10(8): e1004278, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25102051

ABSTRACT

Borrelia burgdorferi transmission to the vertebrate host commences with growth of the spirochete in the tick gut and migration from the gut to the salivary glands. This complex process, involving intimate interactions of the spirochete with the gut epithelium, is pivotal to transmission. We utilized a yeast surface display library of tick gut proteins to perform a global screen for tick gut proteins that might interact with Borrelia membrane proteins. A putative fibronectin type III domain-containing tick gut protein (Ixofin3D) was most frequently identified from this screen and prioritized for further analysis. Immunization against Ixofin3D and RNA interference-mediated reduction in expression of Ixofin3D resulted in decreased spirochete burden in tick salivary glands and in the murine host. Microscopic examination showed decreased aggregation of spirochetes on the gut epithelium concomitant with reduced expression of Ixofin3D. Our observations suggest that the interaction between Borrelia and Ixofin3D facilitates spirochete congregation to the gut during transmission, and provides a "molecular exit" direction for spirochete egress from the gut.


Subject(s)
Arthropod Proteins/metabolism , Fibronectins/metabolism , Host-Parasite Interactions/physiology , Lyme Disease/transmission , Ticks/metabolism , Animals , Borrelia burgdorferi , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Mice , Microscopy, Confocal , Molecular Sequence Data , Polymerase Chain Reaction
9.
Emerg Infect Dis ; 20(7): 1183-90, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24960072

ABSTRACT

Borrelia miyamotoi sensu lato, a relapsing fever Borrelia sp., is transmitted by the same ticks that transmit B. burgdorferi (the Lyme disease pathogen) and occurs in all Lyme disease-endemic areas of the United States. To determine the seroprevalence of IgG against B. miyamotoi sensu lato in the northeastern United States and assess whether serum from B. miyamotoi sensu lato-infected persons is reactive to B. burgdorferi antigens, we tested archived serum samples from area residents during 1991-2012. Of 639 samples from healthy persons, 25 were positive for B. miyamotoi sensu lato and 60 for B. burgdorferi. Samples from ≈10% of B. miyamotoi sensu lato-seropositive persons without a recent history of Lyme disease were seropositive for B. burgdorferi. Our results suggest that human B. miyamotoi sensu lato infection may be common in southern New England and that B. burgdorferi antibody testing is not an effective surrogate for detecting B. miyamotoi sensu lato infection.


Subject(s)
Borrelia Infections/epidemiology , Borrelia/immunology , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Borrelia Infections/blood , Borrelia Infections/immunology , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Lyme Disease/blood , Lyme Disease/epidemiology , Lyme Disease/immunology , Male , Middle Aged , New England/epidemiology , Seroepidemiologic Studies
10.
J Autoimmun ; 53: 85-94, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24767831

ABSTRACT

The composition of the gut microbiome represents a very important environmental factor that influences the development of type 1 diabetes (T1D). We have previously shown that MyD88-deficient non-obese diabetic (MyD88-/-NOD) mice, that were protected from T1D development, had a different composition of gut microbiota compared to wild type NOD mice. The aim of our study was to investigate whether this protection could be transferred. We demonstrate that transfer of gut microbiota from diabetes-protected MyD88-deficient NOD mice, reduced insulitis and significantly delayed the onset of diabetes. Gut bacteria from MyD88-deficient mice, administered over a 3-week period, starting at 4 weeks of age, stably altered the family composition of the gut microbiome, with principally Lachnospiraceae and Clostridiaceae increased and Lactobacillaceae decreased. The transferred mice had a higher concentration of IgA and TGFß in the lumen that was accompanied by an increase in CD8(+)CD103(+) and CD8αß T cells in the lamina propria of the large intestine. These data indicate not only that gut bacterial composition can be altered after the neonatal/weaning period, but that the composition of the microbiome affects the mucosal immune system and can delay the development of autoimmune diabetes. This result has important implications for the development of probiotic treatment for T1D.


Subject(s)
Bacteria/immunology , Diabetes Mellitus, Experimental , Intestines , Microbiota/immunology , Probiotics/pharmacology , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/microbiology , Diabetes Mellitus, Experimental/therapy , Intestines/immunology , Intestines/microbiology , Mice , Mice, Inbred NOD , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology
11.
Comp Med ; 74(4): 235-245, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39289828

ABSTRACT

Due to their hematophagous life cycle, hard-bodied ticks including the genus Ixodes are a potential vector for numerous pathogenic organisms including bacteria, protozoa, viruses, and infectious prions. The natural geographic range of several hard tick species, includig Ixodes scapularis, has expanded over recent decades. Consequently, there is an ongoing need to maintain, feed, and propagate ticks for host-pathogen interaction studies to better understand and mitigate their impact on human and animal health. Artificial membrane feeding of hard ticks has advanced in recent years, has study design advantages, and should be used, when possible, to reduce animal use, but it also has several limitations that require the continued use of mammalian hosts including mice, guinea pigs, and rabbits. In this overview, we discuss the best management practices for these relevant species with respect to biosafety, health, and optimal host comfort when used in studies that depend on tick feeding. The capsule-jacket method is preferred over the ear sock-E-collar method of tick feeding on rabbit hosts because of better host health, comfort, and increased study versatility.


Subject(s)
Host-Pathogen Interactions , Ixodes , Animals , Ixodes/microbiology , Rabbits , Mice , Guinea Pigs , Humans
12.
Nat Microbiol ; 9(9): 2278-2291, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38997520

ABSTRACT

Arthropod-borne pathogens are responsible for hundreds of millions of infections in humans each year. The blacklegged tick, Ixodes scapularis, is the predominant arthropod vector in the United States and is responsible for transmitting several human pathogens, including the Lyme disease spirochete Borrelia burgdorferi and the obligate intracellular rickettsial bacterium Anaplasma phagocytophilum, which causes human granulocytic anaplasmosis. However, tick metabolic response to microbes and whether metabolite allocation occurs upon infection remain unknown. Here we investigated metabolic reprogramming in the tick ectoparasite I. scapularis and determined that the rickettsial bacterium A. phagocytophilum and the spirochete B. burgdorferi induced glycolysis in tick cells. Surprisingly, the endosymbiont Rickettsia buchneri had a minimal effect on bioenergetics. An unbiased metabolomics approach following A. phagocytophilum infection of tick cells showed alterations in carbohydrate, lipid, nucleotide and protein metabolism, including elevated levels of the pleiotropic metabolite ß-aminoisobutyric acid. We manipulated the expression of genes associated with ß-aminoisobutyric acid metabolism in I. scapularis, resulting in feeding impairment, diminished survival and reduced bacterial acquisition post haematophagy. Collectively, we discovered that metabolic reprogramming affects interspecies relationships and fitness in the clinically relevant tick I. scapularis.


Subject(s)
Anaplasma phagocytophilum , Borrelia burgdorferi , Ixodes , Rickettsia , Animals , Ixodes/microbiology , Anaplasma phagocytophilum/metabolism , Anaplasma phagocytophilum/genetics , Rickettsia/genetics , Rickettsia/metabolism , Borrelia burgdorferi/genetics , Borrelia burgdorferi/metabolism , Mice , Lyme Disease/microbiology , Glycolysis , Metabolomics , Humans , Genetic Fitness , Symbiosis
13.
PLoS Pathog ; 7(6): e1002079, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21695244

ABSTRACT

Borrelia burgdorferi, the causative agent of Lyme disease, is transmitted to humans by bite of Ixodes scapularis ticks. The mechanisms by which the bacterium is transmitted from vector to host are poorly understood. In this study, we show that the F(ab)(2) fragments of BBE31, a B.burgdorferi outer-surface lipoprotein, interfere with the migration of the spirochete from tick gut into the hemolymph during tick feeding. The decreased hemolymph infection results in lower salivary glands infection, and consequently attenuates mouse infection by tick-transmitted B. burgdorferi. Using a yeast surface display approach, a tick gut protein named TRE31 was identified to interact with BBE31. Silencing tre31 also decreased the B. burgdorferi burden in the tick hemolymph. Delineating the specific spirochete and arthropod ligands required for B. burgdorferi movement in the tick may lead to new strategies to interrupt the life cycle of the Lyme disease agent.


Subject(s)
Borrelia burgdorferi/pathogenicity , Gastrointestinal Tract/microbiology , Hemolymph/microbiology , Lyme Disease/microbiology , Ticks/microbiology , Animals , Bacterial Outer Membrane Proteins , Lipoproteins , Molecular Sequence Data , Movement
14.
EMBO Rep ; 12(11): 1196-203, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-21921936

ABSTRACT

Ixodes ticks harbour several human pathogens belonging to the order Rickettsiales, including Anaplasma phagocytophilum, the agent of human anaplasmosis. When ticks feed on A. phagocytophilum-infected mice, the pathogen enters the ticks' gut. The bacteria then migrate from the gut to infect the salivary glands of the ticks and are transmitted to the next host via the saliva. The molecular mechanisms that enable the migration of A. phagocytophilum from the gut to the salivary glands are poorly understood. Here we show that a secreted tick protein, P11, is important in this process. We show that P11 enables A. phagocytophilum to infect tick haemocytes, which are required for the migration of A. phagocytophilum from the gut to the salivary glands. Silencing of p11 impaired the A. phagocytophilum infection of tick haemocytes in vivo and consequently decreased pathogen infection of the salivary glands. In vitro experiments showed that P11 could bind to A. phagocytophilum and thus facilitate its infection of tick cells. This report provides new insights into A. phagocytophilum infection of ticks and reveals new avenues to interrupt the life cycle of Anaplasma and related Rickettsial pathogens.


Subject(s)
Anaplasma phagocytophilum/physiology , Gastrointestinal Tract/microbiology , Ixodes/immunology , Protozoan Proteins/metabolism , Salivary Glands/microbiology , Salivary Proteins and Peptides/metabolism , Animals , Antibodies, Protozoan/immunology , Ehrlichiosis/microbiology , Feeding Behavior/physiology , Gene Expression Regulation , Gene Silencing , Hemocytes/microbiology , Hemolymph/microbiology , Humans , Mice , Movement , Phagocytosis , Protein Binding , Protozoan Proteins/immunology , Salivary Proteins and Peptides/genetics
15.
J Infect Dis ; 206(8): 1233-41, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22859824

ABSTRACT

Ixodes scapularis transmits the agent of human granulocytic anaplasmosis, among other pathogens. The mechanisms used by the tick to control Anaplasma phagocytophilum are not known. We demonstrate that the I. scapularis Janus kinase (JAK)-signaling transducer activator of transcription (STAT) pathway plays a critical role in A. phagocytophilum infection of ticks. The A. phagocytophilum burden increases in salivary glands and hemolymph when the JAK-STAT pathway is suppressed by RNA interference. The JAK-STAT pathway exerts its anti-Anaplasma activity presumably through STAT-regulated effectors. A salivary gland gene family encoding 5.3-kDa antimicrobial peptides is highly induced upon A. phagocytophilum infection of tick salivary glands. Gene expression and electrophoretic mobility shift assays showed that the 5.3-kDa antimicrobial peptide-encoding genes are regulated by tick STAT. Silencing of these genes increased A. phagocytophilum infection of tick salivary glands and transmission to mammalian host. These data suggest that the JAK-STAT signaling pathway plays a key role in controlling A. phagocytophilum infection in ticks by regulating the expression of antimicrobial peptides.


Subject(s)
Anaplasma phagocytophilum/immunology , Antimicrobial Cationic Peptides/immunology , Ixodes/microbiology , Janus Kinase 1/immunology , STAT Transcription Factors/immunology , Signal Transduction , Animals , Antimicrobial Cationic Peptides/biosynthesis , Electrophoretic Mobility Shift Assay , Gene Expression Profiling , Gene Expression Regulation , Ixodes/immunology , Salivary Glands/immunology , Salivary Glands/microbiology
16.
Trends Parasitol ; 39(12): 1100-1113, 2023 12.
Article in English | MEDLINE | ID: mdl-37838514

ABSTRACT

Ticks are hematophagous arthropods that transmit disease-causing pathogens worldwide. Tick saliva deposited into the tick-bite site is composed of an array of immunomodulatory proteins that ensure successful feeding and pathogen transmission. These salivary proteins are often glycosylated, and glycosylation is potentially critical for the function of these proteins. Some salivary glycans are linked to the phenomenon of red meat allergy - an allergic response to red meat consumption in humans exposed to certain tick species. Tick salivary glycans are also invoked in the phenomenon of acquired tick resistance wherein non-natural host species exposed to tick bites develop an immune response that thwarts subsequent tick feeding. This review dwells on our current knowledge of these two phenomena, thematically linked by salivary glycans.


Subject(s)
Food Hypersensitivity , Tick Bites , Ticks , Humans , Animals , Tick Bites/complications , Sugars , Food Hypersensitivity/etiology , Polysaccharides
17.
Sci Transl Med ; 15(718): eadi7829, 2023 10 18.
Article in English | MEDLINE | ID: mdl-37851823

ABSTRACT

The deer tick transmits nearly half of the known tick-borne pathogens in the United States, and its expanding geographic range increases the risk of human infection. To decrease the abundance of and infection risk from deer ticks, approaches that include vaccines for human use and for animal hosts are desired.


Subject(s)
Ixodes , Tick Infestations , Animals , Humans
18.
Pathogens ; 12(1)2023 Jan 13.
Article in English | MEDLINE | ID: mdl-36678479

ABSTRACT

The blacklegged tick, Ixodes scapularis, is the predominant vector of Borrelia burgdorferi, the agent of Lyme disease in the USA. Natural hosts of I. scapularis such as Peromyscus leucopus are repeatedly infested by these ticks without acquiring tick resistance. However, upon repeated tick infestations, non-natural hosts such as guinea pigs, mount a robust immune response against critical tick salivary antigens and acquire tick resistance able to thwart tick feeding and Borrelia burgdorferi transmission. The salivary targets of acquired tick resistance could serve as vaccine targets to prevent tick feeding and the tick transmission of human pathogens. Currently, there is no animal model able to demonstrate both tick resistance and diverse clinical manifestations of Lyme disease. Non-human primates serve as robust models of human Lyme disease. By evaluating the responses to repeated tick infestation, this animal model could accelerate our ability to define the tick salivary targets of acquired resistance that may serve as vaccines to prevent the tick transmission of human pathogens. Towards this goal, we assessed the development of acquired tick resistance in non-human primates upon repeated tick infestations. We report that following repeated tick infestations, non-human primates do not develop the hallmarks of acquired tick resistance observed in guinea pigs. However, repeated tick infestations elicit immune responses able to impair the tick transmission of B. burgdorferi. A mechanistic understanding of the protective immune responses will provide insights into B. burgdorferi-tick-host interactions and additionally contribute to anti-tick vaccine discovery.

19.
Vaccine ; 41(34): 4996-5002, 2023 07 31.
Article in English | MEDLINE | ID: mdl-37407406

ABSTRACT

Acquired resistance to ticks can develop when animals are repeatedly exposed to ticks. Recently, acquired resistance to Ixodes scapularis was induced in guinea pigs immunized with an mRNA-lipid nanoparticle vaccine (19ISP) encoding 19 I. scapularis proteins. Here, we evaluated specific mRNAs present in 19ISP to identify critical components associated with resistance to ticks. A lipid nanoparticle containing 12 mRNAs which included all the targets within 19ISP that elicited strong humoral responses in guinea pigs, was sufficient to induce robust resistance to ticks. Lipid nanoparticles containing fewer mRNAs or a single mRNA were not able to generate strong resistance to ticks. All lipid nanoparticles containing salp14 mRNA, however, were associated with increased redness at the tick bite site - which is the first manifestation of acquired resistance to ticks. This study demonstrates that more than one I. scapularis target within 19ISP is required for resistance to ticks, and that additional targets may also play a role in this process.


Subject(s)
Ixodes , Lyme Disease , Animals , Guinea Pigs , RNA, Messenger , Ixodes/genetics
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL