Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
1.
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
3.
Dev Comp Immunol ; 119: 104012, 2021 06.
Article in English | MEDLINE | ID: mdl-33484780

ABSTRACT

Ticks, being obligate hematophagous arthropods, are exposed to various blood-borne pathogens, including arboviruses. Consequently, their feeding behavior can readily transmit economically important viral pathogens to humans and animals. With this tightly knit vector and pathogen interaction, the replication and transmission of tick-borne viruses (TBVs) must be highly regulated by their respective tick vectors to avoid any adverse effect on the ticks' biological development and viability. Knowledge about the tick-virus interface, although gaining relevant advances in recent years, is advancing at a slower pace than the scientific developments related to mosquito-virus interactions. The unique and complicated feeding behavior of ticks, compared to that of other blood-feeding arthropods, also limits the studies that would further elaborate the antiviral immunity of ticks against TBVs. Hence, knowledge of molecular and cellular immune mechanisms at the tick-virus interface, will further elucidate the successful viral replication of TBVs in ticks and their effective transmission to human and animal hosts.


Subject(s)
Arachnid Vectors/immunology , Immunity, Innate/immunology , Tick Infestations/immunology , Ticks/immunology , Viruses/immunology , Animals , Arachnid Vectors/genetics , Arachnid Vectors/virology , Hemolymph/immunology , Hemolymph/metabolism , Hemolymph/virology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate/genetics , Models, Immunological , Salivary Glands/immunology , Salivary Glands/metabolism , Salivary Glands/virology , Tick Infestations/genetics , Tick Infestations/virology , Ticks/genetics , Ticks/virology , Virus Replication/genetics , Virus Replication/immunology , Viruses/genetics , Viruses/growth & development
4.
Parasite Immunol ; 43(5): e12816, 2021 05.
Article in English | MEDLINE | ID: mdl-33368329

ABSTRACT

The bacterial pathogen Borrelia burgdorferi is the causative agent of Lyme disease and is transmitted to humans through an Ixodes tick vector. B. burgdorferi is able to survive in both mammalian and tick hosts through careful modulation of its gene expression. This allows B. burgdorferi to adapt to the environmental and nutritional changes that occur when it is transmitted between the two hosts. Distinct interactions between the spirochete and its host occur at every step of the enzootic cycle and dictate the ability of the spirochete to survive until the next stage of the cycle. Studying the interface between B. burgdorferi, the Ixodes tick vector and the natural mammalian reservoirs has been made significantly more feasible through the complete genome sequences of the organisms and the advent of high throughput screening technologies. Ultimately, a thorough investigation of the interplay between the two domains (and two phyla within one domain) is necessary in order to completely understand how the pathogen is transmitted.


Subject(s)
Arachnid Vectors/microbiology , Borrelia burgdorferi/physiology , Host Microbial Interactions/physiology , Ixodes/microbiology , Lyme Disease/microbiology , Mammals/microbiology , Animals , Arachnid Vectors/immunology , Borrelia burgdorferi/genetics , Gene Expression , Humans , Ixodes/immunology , Lyme Disease/epidemiology , Lyme Disease/transmission , Mammals/blood , Mammals/parasitology , Microbiota , Nymph/microbiology , Salivary Glands/microbiology
5.
Am J Trop Med Hyg ; 104(1): 175-183, 2021 01.
Article in English | MEDLINE | ID: mdl-33258439

ABSTRACT

In many regions where ticks negatively impact public health or economic production, multiple medically important tick species may have overlapping geographic distribution, and in North America, this includes members of Ixodes, Dermacentor, and Amblyomma genera. Acquired tick resistance is the process by which some animals develop an immune response against feeding ticks after one or more exposures. This form of immunity can restrict the ability of ticks to feed and may inhibit transmission of pathogens. Likewise, many proteins present in tick saliva are conserved among tick species, and prior studies have reported cross-protective host immunity against certain combinations of ticks. In this study, we used a guinea pig model to assess whether host resistance against Ixodes scapularis could confer protection against two other medically important tick vectors, Dermacentor variabilis and Amblyomma americanum. Tick challenges using nymphs were used to induce host resistance against a primary species, followed by additional challenge using a secondary tick species. Tick attachment to hosts and engorgement weights were reduced significantly for D. variabilis and A. americanum feeding on I. scapularis-sensitized hosts. Reciprocally, I. scapularis engorgement weights were reduced to a lesser extent, and attachment was unaffected when feeding on hosts sensitized with either D. variabilis or A. americanum. These results indicate that immunity against I. scapularis could potentially be exploited for use in an anti-tick vaccine targeting multiple tick species and their associated pathogens.


Subject(s)
Arachnid Vectors/immunology , Disease Susceptibility/immunology , Guinea Pigs , Ixodes/immunology , Rodent Diseases/parasitology , Tick Infestations/veterinary , Animals , Laboratory Animal Science , Rodent Diseases/immunology , Tick Infestations/immunology
6.
Curr Protein Pept Sci ; 21(5): 517-526, 2020.
Article in English | MEDLINE | ID: mdl-31613726

ABSTRACT

Lyme disease (LD) is an infectious disease caused by the spirochetes of genus borrelia, which are transmitted by the ticks of the genus ixodes. LD is transmitted by the spirochete B. burgdorferi sensu lato. Once in contact with the host through a tick bite, the pathogen comes into contact with the host defense, and must escape this machinery to establish LD, thus using a large number of mechanisms involving the vector of the pathogen, the pathogen itself and also the host. The initial diagnosis of the disease can be made based on the clinical symptoms of LD and the disease can be treated and cured with antibiotics if the diagnosis is made early in the beginning of the disease. Contrariwise, if LD is left untreated, the pathogen disseminates throughout the tissues and organs of the body, where it establishes different types of disease manifestations. In the nervous system, the inflammation caused by B. burgdorferi is known as Lyme neuroborreliosis (LNB). LNB is one of the principal manifestations of LD. In this review, we systematically describe the different molecular interactions among B. burgdorferi, the vector (tick) and the mammalian host.


Subject(s)
Arachnid Vectors/microbiology , Bacterial Proteins/genetics , Borrelia burgdorferi/pathogenicity , Host-Pathogen Interactions/genetics , Ixodes/microbiology , Lyme Disease/genetics , Membrane Proteins/genetics , Receptors, Cell Surface/genetics , Animals , Arachnid Vectors/immunology , Bacterial Proteins/immunology , Borrelia burgdorferi/immunology , Cytokines/genetics , Cytokines/immunology , Gene Expression Regulation , Host-Pathogen Interactions/immunology , Humans , Ixodes/immunology , Lyme Disease/immunology , Lyme Disease/microbiology , Lyme Disease/pathology , Membrane Proteins/immunology , Nervous System/immunology , Nervous System/microbiology , Nervous System/pathology , Protein Isoforms/genetics , Protein Isoforms/immunology , Receptors, Cell Surface/immunology , Saliva/microbiology , Signal Transduction
7.
Dev Comp Immunol ; 100: 103409, 2019 11.
Article in English | MEDLINE | ID: mdl-31200008

ABSTRACT

The blood-feeding behavior of ticks has resulted in them becoming one of the most important vectors of disease-causing pathogens. Ticks possess a well-developed innate immune system to counter invading pathogens. However, the coevolution of ticks with tick-borne pathogens has adapted these pathogens to the tick's physiology and immune response through several mechanisms including transcriptional regulation. The recent development in tick and tick-borne disease research greatly involved the "omics" approach. The omics approach takes a look en masse at the different genes, proteins, metabolomes, and the microbiome of the ticks that could be differentiated during pathogen infection. Data from this approach revealed that oxidative stress-related molecules in ticks are differentiated and possibly being exploited by the pathogens to evade the tick's immune response. In this study, we review and discuss transcriptomic and proteomic data for some oxidative stress molecules differentially expressed during pathogen infection. We also discuss metabolomics and microbiome data as well as functional genomics in order to provide insight into the tick-pathogen interaction.


Subject(s)
Arachnid Vectors/immunology , Host-Pathogen Interactions/immunology , Oxidative Stress/immunology , Tick-Borne Diseases/prevention & control , Ticks/microbiology , Animals , Arthropod Proteins/genetics , Arthropod Proteins/immunology , Arthropod Proteins/metabolism , Gene Expression Profiling , Genomics , Host-Pathogen Interactions/drug effects , Humans , Metabolomics , Microbiota/drug effects , Microbiota/immunology , Oxidative Stress/drug effects , Proteomics , Tick-Borne Diseases/microbiology , Tick-Borne Diseases/transmission , Ticks/drug effects , Ticks/immunology
8.
Transbound Emerg Dis ; 66(1): 422-434, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30300470

ABSTRACT

Crimean-Congo haemorrhagic fever (CCHF) is a tick borne viral disease reported from different parts of the world. The distribution of the CCHF cases are linked with the distribution of the principal vector, Hyalomma anatolicum in the ecosystem. Presently, vector control is mainly dependent on repeated application of acaricides, results in partial efficacy and generated acaricide resistant tick strains. Amongst the different components of integrated management programme, immunization of hosts is considered as one of the sustainable component. To restrict CCHF virus spreading, use of anti-Hyalomma vaccines appears as a viable solution. Accordingly, present study was under taken to characterize and evaluate vaccine potential of two conserved molecules, ferritin2 (FER2) and tropomyosin (TPM). Silencing of the genes conferred a cumulative reduction (rejection + unable to engorge) of 61.3% in FER2 and 70.2% in TPM respectively. Furthermore, 44.2% and 72.7% reduction in engorgement weight, 63.6% and 94.9% reduction in egg masses in FER2 and TPM silenced ticks in comparison to LUC-control group was recorded. The recombinant protein, rHaFER2 was characterized as 35 kDa protein with pI of 5.84 and possesses iron binding domains. While rHaTPM is a 51kDa protein with pI of 4.94 having calcium binding domains. Immunization of cross-bred calves by rHaFER2 conferred 51.7% and 51.2% protection against larvae and adults of H. anatolicum challenge infestations. While rHaTPM conferred 63.7% and 66.4% protection against larvae and adults infestations, respectively. The results were comparable with the data generated by RNAi and it clearly showed the possibility for the development of anti-hyalomma vaccine to manage CCHF virus and Theileria annulata infection in human and animals.


Subject(s)
Arachnid Vectors/immunology , Cattle Diseases/prevention & control , Ferritins/immunology , Hemorrhagic Fever Virus, Crimean-Congo/immunology , Hemorrhagic Fever, Crimean/veterinary , Protozoan Vaccines/administration & dosage , Ticks/virology , Tropomyosin/immunology , Animals , Arachnid Vectors/virology , Cattle , Cattle Diseases/virology , Disease Vectors , Hemorrhagic Fever, Crimean/prevention & control , Hemorrhagic Fever, Crimean/virology , Humans , Immunization/veterinary , Male , Parasite Egg Count , Protozoan Vaccines/immunology , Recombinant Proteins/immunology
9.
Am J Pathol ; 189(1): 115-123, 2019 01.
Article in English | MEDLINE | ID: mdl-30315767

ABSTRACT

Many aspects of rickettsial infections have been characterized, including pathogenic and immune pathways and mechanisms of rickettsial survival within the vertebrate host and tick vector. However, very few studies are focused on the complex pathogen-vector-host interactions during tick feeding. Therefore, our objective was to develop a tick transmission model of the spotted fever group of rickettsial infections to study the initial events in disease development. The most appropriate strain of mouse was identified for evaluation as a transmission model, and the course of infection, bacterial levels, histopathologic changes, and antibody response during tick transmission in mice infested with Amblyomma maculatum ticks carrying the emerging pathogen, Rickettia parkeri, were studied. Results showed distinct clinical signs in C3H/HeN mice infected intravenously, leading to selection of this mouse strain for tick transmission studies. Active infection of animals was observed after tick vector transmission. The bacteria disseminated systemically and spread to several organs at 24 hours after tick attachment, with peak bacterial load at day 6 after tick attachment. Skin, lung, and liver showed the greatest pathologic changes, with inflammatory cellular infiltration and necrosis. These findings indicate the feasibility of using murine infection with R. parkeri by A. maculatum tick transmission as a model to study different aspects of the spotted fever group of rickettsial disease establishment.


Subject(s)
Arachnid Vectors/microbiology , Ixodidae/microbiology , Rickettsia/pathogenicity , Spotted Fever Group Rickettsiosis , Animals , Antibodies, Bacterial/immunology , Antibody Formation , Arachnid Vectors/immunology , Disease Models, Animal , Humans , Inflammation/immunology , Inflammation/pathology , Ixodidae/immunology , Mice , Mice, Inbred BALB C , Necrosis , Organ Specificity , Species Specificity , Spotted Fever Group Rickettsiosis/immunology , Spotted Fever Group Rickettsiosis/pathology , Spotted Fever Group Rickettsiosis/transmission
10.
J Math Biol ; 78(5): 1331-1364, 2019 04.
Article in English | MEDLINE | ID: mdl-30478760

ABSTRACT

Ixodid ticks are acknowledged as one of the most important hematophagous arthropods because of their ability in transmitting a variety of tick-borne diseases. Mathematical models have been developed, based on emerging knowledge about tick ecology, pathogen epidemiology and their interface, to understand tick population dynamics and tick-borne diseases spread patterns. However, no serious effort has been made to model and assess the impact of host immunity triggered by tick feeding on the distribution of the tick population according to tick stages and on tick population extinction and persistence. Here, we construct a novel mathematical model taking into account the effect of host immunity status on tick population dynamics, and analyze the long-term behaviours of the model solutions. Two threshold values, [Formula: see text] and [Formula: see text], are introduced to measure the reproduction ratios for the tick-host interaction in the absence and presence of host immunity. We then show that these two thresholds (sometimes under additional conditions) can be used to predict whether the tick population goes extinct ([Formula: see text]) and the tick population grows without bound ([Formula: see text]). We also prove tick permanence (persistence and boundedness of the tick population) and the existence of a tick persistence equilibrium if [Formula: see text]. As the host species adjust their immunity to tick infestation levels, they form for the tick population an environment with a carrying capacity very much like that in logistic growth. Numerical results show that the host immune reactions decrease the size of the tick population at equilibrium and apparently reduce the tick-borne infection risk.


Subject(s)
Arachnid Vectors/immunology , Host-Pathogen Interactions/immunology , Models, Immunological , Tick-Borne Diseases/immunology , Tick-Borne Diseases/transmission , Ticks/immunology , Animals , Arachnid Vectors/pathogenicity , Humans , Lyme Disease/immunology , Lyme Disease/parasitology , Lyme Disease/transmission , Mathematical Concepts , Population Dynamics , Tick Infestations/immunology , Tick Infestations/parasitology , Tick-Borne Diseases/parasitology , Ticks/growth & development , Ticks/pathogenicity
11.
Article in English | MEDLINE | ID: mdl-29896452

ABSTRACT

Although Ixodes scapularis and other related tick species are considered prolific vectors for a number of important human diseases, many aspects of their biology, microbial interactions, and immunity are largely unknown; in particular, how these ancient vectors recognize invading pathogens like Borrelia burgdorferi and influence their persistence. The analysis of the Ixodes genome and a limited set of transcriptomic data have established that ticks encode many components of classical immune pathways; yet at the same time, they lack many key orthologs of these recognition networks. Therefore, whether a given immune pathway is active in Ixodes ticks and how precisely they exert its microbicidal functions are only incompletely delineated. A few recent studies have suggested that classical pathways like the Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) as well as immunodeficiency (IMD) pathways are fully functional in I. scapularis, and upon challenge with microbes, generate potent microbicidal responses against diverse tick-borne pathogens including B. burgdorferi. These studies also highlight novel concepts of vector immunity that include both a direct and an indirect mode of recognition of pathogens, as well as the influence of the gut microbiome, which ultimately dictates the outcome of a robust microbicidal response. Further understanding of how Ixodes ticks recognize and suppress invading microbes like B. burgdorferi will enrich our fundamental knowledge of vector immunobiology, thereby contributing to the development of future interventions to better control the tick-borne pathogen.


Subject(s)
Borrelia burgdorferi/immunology , Immunity , Ixodes/immunology , Lyme Disease/immunology , Animals , Anti-Infective Agents , Arachnid Vectors/genetics , Arachnid Vectors/immunology , Arachnid Vectors/microbiology , Borrelia burgdorferi/pathogenicity , Gastrointestinal Microbiome/immunology , Genome, Insect , Host-Pathogen Interactions/immunology , Ixodes/genetics , Ixodes/microbiology , Lyme Disease/microbiology , Signal Transduction , Transcriptome , Tyrosine/analogs & derivatives , Tyrosine/genetics , Tyrosine/immunology , Tyrosine/metabolism
12.
Biol Aujourdhui ; 212(3-4): 107-117, 2018.
Article in French | MEDLINE | ID: mdl-30973140

ABSTRACT

Ticks are the most important vectors of pathogens in human and veterinary medicine. These strictly haematophagous acarines produce a saliva containing a variety of bioactive molecules affecting host pharmacology and immunity. This process is vital for hard ticks to prevent rejection by the host during the blood meal that lasts several days. All actors involved in the immunity interplay are impacted by this saliva, the innate immunity being represented by resident and migrating immune cells, as well as the T and B lymphocytes of the adaptive immune system. The skin plays a key role in vector-borne diseases. During the long co-evolution with the tick, the infectious agents benefit from this favorable environment to be transmitted efficiently into the skin and to multiply in the vertebrate host. Therefore, the saliva is an important virulence booster, which enhances substantially their pathogenicity.


Subject(s)
Arachnid Vectors , Disease Transmission, Infectious , Host-Pathogen Interactions/immunology , Immunomodulation/physiology , Saliva/physiology , Ticks , Animals , Arachnid Vectors/immunology , Arachnid Vectors/metabolism , Humans , Immunity, Innate/physiology , Immunomodulation/genetics , Saliva/immunology , Saliva/metabolism , Ticks/immunology , Ticks/metabolism , Ticks/microbiology , Ticks/virology , Virulence/immunology
13.
Parasit Vectors ; 10(1): 313, 2017 Jun 27.
Article in English | MEDLINE | ID: mdl-28655322

ABSTRACT

BACKGROUND: Borrelia burgdorferi (sensu lato), the causative agent of Lyme borreliosis is a bacterium transmitted by hard ticks, Ixodes spp. Bacteria are injected into the host skin during the tick blood meal with tick saliva. There, Borrelia and saliva interact together with skin cells such as keratinocytes, fibroblasts, mast cells and other specific immune cells before disseminating to target organs. METHODS: To study the role of mast cells in the transmission of Lyme borreliosis, we isolated mouse primary mast cells from bone marrow and incubated them in the presence of Borrelia burgdorferi (sensu stricto) and tick salivary gland extract. We further analyzed their potential role in vivo, in a mouse model of deficient in mast cells (Kit wsh-/- mice). RESULTS: To our knowledge, we report here for the first time the bacteria ability to induce the inflammatory response of mouse primary mast cells. We show that OspC, a major surface lipoprotein involved in the early transmission of Borrelia, induces the degranulation of primary mast cells but has a limited effect on the overall inflammatory response of these cells. In contrast, whole bacteria have an opposite effect. We also show that mast cell activation is significantly inhibited by tick salivary gland extract. Finally, we demonstrate that mast cells are likely not the only host cells involved in the early transmission and dissemination of Borrelia since the use of mast cell deficient Kit wsh-/- mice shows a limited impact on these two processes in the context of this mouse genetic background. CONCLUSIONS: The absence of mast cells did not change the replication rate of Borrelia in the skin. However, in the absence of mast cells, Borrelia dissemination to the joints was faster. Mast cells do not control skin bacterial proliferation during primary infection and the establishment of the primary infection, as shown in the C57BL/6 mouse model studied. Nevertheless, the Borrelia induced cytotokine modulation on mast cells might be involved in long term and/or repeated infections and protect from Lyme borreliosis due to the development of a hypersensitivity to tick saliva.


Subject(s)
Antigens, Bacterial/metabolism , Arachnid Vectors/immunology , Bacterial Outer Membrane Proteins/metabolism , Borrelia burgdorferi/immunology , Ixodes/immunology , Lyme Disease/immunology , Mast Cells/immunology , Animals , Antigens, Bacterial/genetics , Arachnid Vectors/microbiology , Bacterial Outer Membrane Proteins/genetics , Borrelia burgdorferi/physiology , Cytokines/immunology , Disease Models, Animal , Female , Gene Expression Regulation , Host-Pathogen Interactions , Ixodes/microbiology , Lyme Disease/microbiology , Lyme Disease/transmission , Mast Cells/microbiology , Mice , Saliva/immunology , Saliva/microbiology , Salivary Glands/immunology , Salivary Glands/microbiology
14.
Dev Comp Immunol ; 74: 110-124, 2017 09.
Article in English | MEDLINE | ID: mdl-28431895

ABSTRACT

Anopheles gambiae is a major vector of human malaria and its immune system in part determines the fate of ingested parasites. Proteins, hemocytes and fat body in hemolymph are critical components of this system, mediating both humoral and cellular defenses. Here we assessed differences in the hemolymph proteomes of water- and E. coli-pricked mosquito larvae by a gel-LC-MS approach. Among the 1756 proteins identified, 603 contained a signal peptide but accounted for two-third of the total protein amount on the quantitative basis. The sequence homology search indicated that 233 of the 1756 may be related to defense. In general, we did not detect substantial differences between the control and induced plasma samples in terms of protein numbers or levels. Protein distributions in the gel slices suggested post-translational modifications (e.g. proteolysis) and formation of serpin-protease complexes and high Mr immune complexes. Based on the twenty-five most abundant proteins, we further suggest that major functions of the larval hemolymph are storage, transport, and immunity. In summary, this study provided first data on constitution, levels, and possible functions of hemolymph proteins in the mosquito larvae, reflecting complex changes occurring in the fight against E. coli infection.


Subject(s)
Anopheles/immunology , Arachnid Vectors/immunology , Escherichia coli Infections/immunology , Escherichia coli/immunology , Hemocytes/metabolism , Hemolymph/metabolism , Malaria/immunology , Animals , Anopheles/microbiology , Antigen-Antibody Complex/metabolism , Hemocytes/immunology , Hemocytes/microbiology , Hemolymph/immunology , Hemolymph/microbiology , Humans , Immunity , Larva , Plasmodium/physiology , Proteolysis , Proteome , Serpins/metabolism
15.
Article in English | MEDLINE | ID: mdl-28361038

ABSTRACT

The present concept of the transmission of Lyme disease from Borrelia-infected Ixodes sp. ticks to the naïve host assumes that a low number of spirochetes that manage to penetrate the midgut epithelium migrate through the hemocoel to the salivary glands and subsequently infect the host with the aid of immunomodulatory compounds present in tick saliva. Therefore, humoral and/or cellular immune reactions within the tick hemocoel may play an important role in tick competence to act as a vector for borreliosis. To test this hypothesis we have examined complement-like reactions in the hemolymph of the hard tick Ixodes ricinus against Borrelia afzelii (the most common vector and causative agent of Lyme disease in Europe). We demonstrate that I. ricinus hemolymph does not exhibit borreliacidal effects comparable to complement-mediated lysis of bovine sera. However, after injection of B. afzelii into the tick hemocoel, the spirochetes were efficiently phagocytosed by tick hemocytes and this cellular defense was completely eliminated by pre-injection of latex beads. As tick thioester-containing proteins (T-TEPs) are components of the tick complement system, we performed RNAi-mediated silencing of all nine genes encoding individual T-TEPs followed by in vitro phagocytosis assays. Silencing of two molecules related to the C3 complement component (IrC3-2 and IrC3-3) significantly suppressed phagocytosis of B. afzelii, while knockdown of IrTep (insect type TEP) led to its stimulation. However, RNAi-mediated silencing of T-TEPs or elimination of phagocytosis by injection of latex beads in B. afzelii-infected I. ricinus nymphs had no obvious impact on the transmission of spirochetes to naïve mice, as determined by B. afzelii infection of murine tissues following tick infestation. This result supports the concept that Borrelia spirochetes are capable of avoiding complement-related reactions within the hemocoel of ticks competent to transmit Lyme disease.


Subject(s)
Arachnid Vectors/microbiology , Borrelia burgdorferi Group/immunology , Complement System Proteins/metabolism , Hemocytes/immunology , Ixodes/microbiology , Lyme Disease/transmission , Phagocytosis , Animals , Arachnid Vectors/immunology , Arthropod Proteins/metabolism , Disease Models, Animal , Disease Transmission, Infectious , Ixodes/immunology , Mice
16.
Exp Parasitol ; 170: 227-235, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27729251

ABSTRACT

Rhipicephalus (Boophilus) microplus ticks are obligatory hematophagous ectoparasites of cattle and act as vectors for disease-causing microorganisms. Conventional tick control is based on the use of chemical acaricides; however, their uncontrolled use has increased tSresistant tick populations, as well as food and environmental contamination. Alternative immunological tick control has shown to be partially effective. The only anti-tick vaccine commercially available at present in the world is based on intestinal Bm86 protein, and shows a variable effectiveness depending on tick strains or geographic isolates. Therefore, there is a need to characterize new antigens in order to improve immunological protection. The aim of this work was to identify immunogenic proteins from ovarian tissue extracts of R. microplus, after cattle immunization. Results showed that ovarian proteins complexed with the adjuvant Montanide ISA 50 V generated a strong humoral response on vaccinated cattle. IgG levels peaked at fourth post-immunization week and remained high until the end of the experiment. 1D and 2D SDS-PAGE-Western blot assays with sera from immunized cattle recognized several ovarian proteins. Reactive bands were cut and analyzed by LC-MS/MS. They were identified as Vitellogenin, Vitellogenin-2 precursor and Yolk Cathepsin. Our findings along with bioinformatic analysis indicate that R. microplus has several Vitellogenin members, which are proteolytically processed to generate multiple polypeptide fragments. This apparent complexity of vitellogenic tick molecular targets gives the opportunity to explore their potential usefulness as vaccine candidates but, at the same time, imposes a challenge on the selection of the appropriate set of antigens.


Subject(s)
Arachnid Vectors/immunology , Arthropod Proteins/immunology , Rhipicephalus/immunology , Tick Control/methods , Animals , Blotting, Western , Cattle , Cattle Diseases/prevention & control , DNA, Complementary/biosynthesis , Electrophoresis/methods , Embryonic Development/immunology , Enzyme-Linked Immunosorbent Assay , Female , Immunoglobulin G/blood , Larva/immunology , Oogenesis/immunology , Ovary/immunology , Polymerase Chain Reaction , Proteomics/methods , RNA/genetics , RNA/isolation & purification , Reproduction/immunology , Tandem Mass Spectrometry , Tick Infestations/prevention & control , Tick Infestations/veterinary , Vaccines , Vitellogenins/biosynthesis , Vitellogenins/immunology
17.
Parasit Vectors ; 9(1): 484, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27589998

ABSTRACT

BACKGROUND: Rhipicephalus appendiculatus is the primary vector of Theileria parva, the etiological agent of East Coast fever (ECF), a devastating disease of cattle in sub-Saharan Africa. We hypothesized that a vaccine targeting tick proteins that are involved in attachment and feeding might affect feeding success and possibly reduce tick-borne transmission of T. parva. Here we report the evaluation of a multivalent vaccine cocktail of tick antigens for their ability to reduce R. appendiculatus feeding success and possibly reduce tick-transmission of T. parva in a natural host-tick-parasite challenge model. METHODS: Cattle were inoculated with a multivalent antigen cocktail containing recombinant tick protective antigen subolesin as well as two additional R. appendiculatus saliva antigens: the cement protein TRP64, and three different histamine binding proteins. The cocktail also contained the T. parva sporozoite antigen p67C. The effect of vaccination on the feeding success of nymphal and adult R. appendiculatus ticks was evaluated together with the effect on transmission of T. parva using a tick challenge model. RESULTS: To our knowledge, this is the first evaluation of the anti-tick effects of these antigens in the natural host-tick-parasite combination. In spite of evidence of strong immune responses to all of the antigens in the cocktail, vaccination with this combination of tick and parasite antigens did not appear to effect tick feeding success or reduce transmission of T. parva. CONCLUSION: The results of this study highlight the importance of early evaluation of anti-tick vaccine candidates in biologically relevant challenge systems using the natural tick-host-parasite combination.


Subject(s)
Antigens/immunology , Arachnid Vectors/parasitology , Arthropod Proteins/immunology , Rhipicephalus/parasitology , Theileria parva/physiology , Theileriasis/transmission , Animals , Antigens/genetics , Arachnid Vectors/immunology , Arachnid Vectors/physiology , Arthropod Proteins/genetics , Feeding Behavior , Humans , Immunity, Humoral , Mice , Rhipicephalus/immunology , Rhipicephalus/physiology , Theileriasis/parasitology
18.
Infect Immun ; 84(6): 1796-1805, 2016 06.
Article in English | MEDLINE | ID: mdl-27045038

ABSTRACT

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


Subject(s)
Anaplasma phagocytophilum/immunology , Annexin A2/immunology , Apoptosis Regulatory Proteins/immunology , Calcium-Binding Proteins/immunology , Cystatins/immunology , Ehrlichiosis/microbiology , Immune Evasion , Amino Acid Sequence , Anaplasma phagocytophilum/genetics , Animals , Annexin A2/chemistry , Annexin A2/genetics , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Arachnid Vectors/chemistry , Arachnid Vectors/genetics , Arachnid Vectors/immunology , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/genetics , Caspase 1/genetics , Caspase 1/immunology , Caspases/genetics , Caspases/immunology , Caspases, Initiator , Cystatins/chemistry , Cystatins/genetics , Ehrlichiosis/immunology , Ehrlichiosis/pathology , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation , Humans , Inflammasomes/genetics , Inflammasomes/immunology , Interleukin-18/genetics , Interleukin-18/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Ixodes/chemistry , Ixodes/genetics , Ixodes/immunology , Macrophages/immunology , Macrophages/microbiology , Mice , Models, Molecular , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Signal Transduction
19.
Trends Parasitol ; 32(5): 368-377, 2016 05.
Article in English | MEDLINE | ID: mdl-26830726

ABSTRACT

The saliva of ixodid ticks contains a mixture of bioactive molecules that target a wide spectrum of host defense mechanisms to allow ticks to feed on the vertebrate host for several days. Tick salivary proteins cluster in multigenic protein families, and individual family members display redundancy and pluripotency in their action to ameliorate or evade host immune responses. It is now clear that members of different protein families can target the same cellular or molecular pathway of the host physiological response to tick feeding. We present and discuss our hypothesis that redundancy and pluripotency evolved in tick salivary immunomodulators to evade immune recognition by the host while retaining the immunomodulatory potential of their saliva.


Subject(s)
Arachnid Vectors/immunology , Arthropod Proteins/immunology , Host-Parasite Interactions/immunology , Immune Evasion/immunology , Ixodidae/immunology , Salivary Proteins and Peptides/immunology , Animals , Arachnid Vectors/parasitology , Humans , Ixodidae/parasitology , Parasitic Diseases/immunology , Parasitic Diseases/transmission
20.
PLoS One ; 10(9): e0136180, 2015.
Article in English | MEDLINE | ID: mdl-26352137

ABSTRACT

Ticks are vectors for various, including pathogenic, microbes. Tick saliva contains multiple anti-host defense factors that enable ticks their bloodmeals yet also facilitate microbe transmission. Lyme disease-causing borreliae profit specifically from the broadly conserved tick histamine release factor (tHRF), and from cysteine-rich glycoproteins represented by Salp15 from Ixodes scapularis and Iric-1 from Ixodes ricinus ticks which they recruit to their outer surface protein C (OspC). Hence these tick proteins are attractive targets for anti-tick vaccines that simultaneously impair borrelia transmission. Main obstacles are the tick proteins´ immunosuppressive activities, and for Salp15 orthologs, the lack of efficient recombinant expression systems. Here, we exploited the immune-enhancing properties of hepatitis B virus core protein (HBc) derived capsid-like particles (CLPs) to generate, in E. coli, nanoparticulate vaccines presenting tHRF and, as surrogates for the barely soluble wild-type proteins, cysteine-free Salp15 and Iric-1 variants. The latter CLPs were exclusively accessible in the less sterically constrained SplitCore system. Mice immunized with tHRF CLPs mounted a strong anti-tHRF antibody response. CLPs presenting cysteine-free Salp15 and Iric-1 induced antibodies to wild-type, including glycosylated, Salp15 and Iric-1. The broadly distributed epitopes included the OspC interaction sites. In vitro, the anti-Salp15 antibodies interfered with OspC binding and enhanced human complement-mediated killing of Salp15 decorated borreliae. A mixture of all three CLPs induced high titered antibodies against all three targets, suggesting the feasibility of combination vaccines. These data warrant in vivo validation of the new candidate vaccines´ protective potential against tick infestation and Borrelia transmission.


Subject(s)
Antibodies, Neutralizing/biosynthesis , Antigen Presentation , Arachnid Vectors/immunology , Borrelia burgdorferi/immunology , Capsid/immunology , Hepatitis B Core Antigens/immunology , Hepatitis B virus/immunology , Ixodes/immunology , Salivary Proteins and Peptides/immunology , Vaccines, Synthetic , Amino Acid Sequence , Animals , Antibodies, Neutralizing/immunology , Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/metabolism , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Epitopes/metabolism , Hepatitis B Core Antigens/chemistry , Hepatitis B Core Antigens/genetics , Humans , Lyme Disease/prevention & control , Lyme Disease/transmission , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/immunology , Recombinant Fusion Proteins/immunology , Salivary Proteins and Peptides/genetics , Salivary Proteins and Peptides/metabolism , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL
...