Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Vaccine ; 41(12): 1951-1960, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36797101

ABSTRACT

INTRODUCTION: Borrelia burgdorferi sensu lato, the causative agents of Lyme borreliosis, are transmitted by Ixodes ticks. Tick saliva proteins are instrumental for survival of both the vector and spirochete and have been investigated as targets for vaccine targeting the vector. In Europe, the main vector for Lyme borreliosis is Ixodes ricinus, which predominantly transmits Borrelia afzelii. We here investigated the differential production of I. ricinus tick saliva proteins in response to feeding and B. afzelii infection. METHOD: Label-free Quantitative Proteomics and Progenesis QI software was used to identify, compare, and select tick salivary gland proteins differentially produced during tick feeding and in response to B. afzelii infection. Tick saliva proteins were selected for validation, recombinantly expressed and used in both mouse and guinea pig vaccination and tick-challenge studies. RESULTS: We identified 870 I. ricinus proteins from which 68 were overrepresented upon 24-hours of feeding and B. afzelii infection. Selected tick proteins were successfully validated by confirming their expression at the RNA and native protein level in independent tick pools. When used in a recombinant vaccine formulation, these tick proteins significantly reduced the post-engorgement weights of I. ricinus nymphs in two experimental animal models. Despite the reduced ability of ticks to feed on vaccinated animals, we observed efficient transmission of B. afzelii to the murine host. CONCLUSION: Using quantitative proteomics, we identified differential protein production in I. ricinus salivary glands in response to B. afzelii infection and different feeding conditions. These results provide novel insights into the process of I. ricinus feeding and B. afzelii transmission and revealed novel candidates for an anti-tick vaccine.


Subject(s)
Ixodes , Lyme Disease , Vaccines , Animals , Guinea Pigs , Mice , Proteome , Arachnid Vectors , Lyme Disease/prevention & control , Salivary Glands , Arthropod Proteins
2.
Pathogens ; 9(2)2020 Feb 24.
Article in English | MEDLINE | ID: mdl-32102367

ABSTRACT

In Europe, Ixodes ricinus ticks transmit pathogens such as Borrelia burgdorferi sensu lato and tick-borne encephalitis virus (TBEV). In addition, there is evidence for transmission to humans from I. ricinus of Anaplasma phagocytophilum, Babesia divergens, Babesia microti, Babesia venatorum, Borrelia miyamotoi, Neoehrlichia mikurensis, Rickettsia helvetica and Rickettsia monacensis. However, whether infection with these potential tick-borne pathogens results in human disease has not been fully demonstrated for all of these tick-borne microorganisms. To evaluate the available evidence for a causative relation between infection and disease, the current study analyses European case reports published from 2008 to 2018, supplemented with information derived from epidemiological and experimental studies. The evidence for human disease causality in Europe found in this review appeared to be strongest for A. phagocytophilum and B. divergens. Nonetheless, some knowledge gaps still exist. Importantly, comprehensive evidence for pathogenicity is lacking for the remaining tick-borne microorganisms. Such evidence could be gathered best through prospective studies, for example, studies enrolling patients with a fever after a tick bite, the development of specific new serological tools, isolation of these microorganisms from ticks and patients and propagation in vitro, and through experimental studies.

3.
J Immunol ; 204(2): 386-393, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31818980

ABSTRACT

The spirochete Borrelia miyamotoi has recently been shown to cause relapsing fever. Like the Lyme disease agent, Borrelia burgdorferi, B. miyamotoi is transmitted through the bite of infected ticks; however, little is known about the response of the immune system upon infection. Dendritic cells (DCs) play a central role in the early immune response against B. burgdorferi We investigated the response of DCs to two different strains of B. miyamotoi using in vitro and ex vivo models and compared this to the response elicited by B. burgdorferi. Our findings show that B. miyamotoi is phagocytosed by monocyte-derived DCs, causing upregulation of activation markers and production of proinflammatory cytokines in a similar manner to B. burgdorferi. Recognition of B. miyamotoi was demonstrated to be partially mediated by TLR2. DCs migrated out of human skin explants upon inoculation of the skin with B. miyamotoi. Finally, we showed that B. miyamotoi-stimulated DCs induced proliferation of naive CD4+ and CD8+ T cells to a larger extent than B. burgdorferi. In conclusion, we show in this study that DCs respond to and mount an immune response against B. miyamotoi that is similar to the response to B. burgdorferi and is able to induce T cell proliferation.


Subject(s)
Borrelia/physiology , Dendritic Cells/immunology , Insect Bites and Stings/immunology , Relapsing Fever/immunology , Skin/pathology , T-Lymphocytes/immunology , Ticks/immunology , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Lymphocyte Activation , Phagocytosis , Ticks/microbiology , Toll-Like Receptor 2/metabolism
4.
Parasit Vectors ; 12(1): 362, 2019 Jul 25.
Article in English | MEDLINE | ID: mdl-31345265

ABSTRACT

BACKGROUND: Rhipicephalus microplus is a hard tick species that has a high impact on cattle health and production in tropical and subtropical regions. Recently, ribosomal DNA and morphological analysis resulted in the reinstatement of R. australis as a separate species from R. microplus. Both feed on cattle and can transmit bovine pathogens such as Anaplasma and Babesia species. The current treatment with acaricides is becoming increasingly less effective due to the emergence of resistant tick strains. A promising alternative can be found in the form of anti-tick vaccines. The available commercial vaccines can be used to control tick infestation, but the lack of a knockdown effect (> 90% reduction in tick numbers as seen with effective acaricides) hampers its widespread use, hence higher efficacious vaccines are needed. Instead of searching for new protective antigens, we investigated the efficacy of vaccines that contain more than one (partially) protective antigen. For screening vaccine formulations, a previously developed in vitro feeding assay was used in which R. australis larvae are fed sera that were raised against the candidate vaccine antigens. In the present study, the efficacy of the Bm86 midgut antigen and the cytosolic Subolesin (SUB) antigen were evaluated in vitro. RESULTS: Antiserum against recombinant Bm86 (rBm86) partially inhibited larval engorgement, whereas antiserum against recombinant SUB (rSUB) did not have any effect on feeding of larvae. Importantly, when larvae were fed a combination of antiserum against rBm86 and rSUB, a synergistic effect on significantly reducing larval infestations was found. Immunohistochemical analysis revealed that the rBm86 antiserum reacted with gut epithelium of R. australis larvae, whereas the antiserum against rSUB stained salivary glands and rectal sac epithelium. CONCLUSIONS: Combining anti-Bm86 and anti-subolesin antibodies synergistically reduced R. australis larval feeding in vitro. Rhipicephalus australis is a one host tick, meaning that the larvae develop to nymphs and subsequently adults on the same host. Hence, this protective effect could be even more pronounced when larvae are used for infestation of vaccinated cattle, as the antibodies could then affect all three developmental stages. This will be tested in future in vivo experiments.


Subject(s)
Antibodies/pharmacology , Antigens/immunology , Arthropod Proteins/immunology , Immune Sera/pharmacology , Membrane Glycoproteins/immunology , Rhipicephalus/drug effects , Animals , Antigens/genetics , Arthropod Proteins/genetics , Cattle , Female , Larva/drug effects , Larva/physiology , Membrane Glycoproteins/genetics , Recombinant Proteins/immunology , Rhipicephalus/physiology , Vaccines/immunology
5.
BMC Infect Dis ; 19(1): 324, 2019 Apr 15.
Article in English | MEDLINE | ID: mdl-30987580

ABSTRACT

BACKGROUND: After antibiotic treatment of Lyme borreliosis, a subset of patients report persistent symptoms, also referred to as post-treatment Lyme disease syndrome. The reported prevalence of persistent symptoms varies considerably, and its pathophysiology is under debate. The LymeProspect study has been designed to investigate the prevalence, severity, and a wide range of hypotheses on the etiology of persistent symptoms among patients treated for Lyme borreliosis in the Netherlands. METHODS: LymeProspect is a prospective, observational cohort study among adults with proven or probable Lyme borreliosis, either erythema migrans or disseminated manifestations, included at the start of antibiotic treatment. During one year of follow-up, participants are subjected to questionnaires every three months and blood is collected repeatedly during the first three months. The primary outcome is the prevalence of persistent symptoms after treatment, assessed by questionnaires online focusing on fatigue (CIS, subscale fatigue severity), pain (SF-36, subscale pain) and neurocognitive dysfunction (CFQ). Potential microbiological, immunological, genetic, epidemiological and cognitive-behavioral determinants for persistent symptoms are secondary outcome measures. Control cohorts include patients with long-lasting symptoms and unconfirmed Lyme disease, population controls, and subjects having reported a tick bite not followed by Lyme borreliosis. DISCUSSION: This article describes the background and design of the LymeProspect study protocol. This study is characterized by a prospective, explorative and multifaceted design. The results of this study will provide insights into the prevalence and determinants of persistent symptoms after treatment for Lyme borreliosis, and may provide a rationale for preventive and treatment recommendations. TRIAL REGISTRATION: NTR4998 (Netherlands Trial Register). Date of registration: 13 February 2015.


Subject(s)
Lyme Disease/drug therapy , Lyme Disease/epidemiology , Adult , Aged , Animals , Anti-Bacterial Agents/therapeutic use , Bites and Stings/complications , Clinical Protocols , Cohort Studies , Erythema Chronicum Migrans/drug therapy , Erythema Chronicum Migrans/epidemiology , Erythema Chronicum Migrans/etiology , Fatigue/etiology , Humans , Lyme Disease/etiology , Middle Aged , Netherlands/epidemiology , Prevalence , Prospective Studies , Surveys and Questionnaires , Ticks
6.
Immunobiology ; 223(11): 694-698, 2018 11.
Article in English | MEDLINE | ID: mdl-30056999

ABSTRACT

Myeloid-related protein (MRP)8 and MRP14 form a complex (MRP8/14) that is released by activated neutrophils and monocytes during infection. MRP8/14 has been shown to have bacteriostatic activity in vitro against Borrelia burgdorferi, the spirochete that causes Lyme borreliosis. Furthermore, levels of MRP8/14 have been shown to be elevated in the joints of patients with Lyme arthritis. We hypothesized that MRP8/14 has a protective effect during B. burgdorferi infection. To determine the role of MRP8/14 in the immune response to B. burgdorferi, we studied the course of B. burgdorferi infection in wildtype (wt) and mrp14-/- mice. In addition, we studied the response of leukocytes from mice lacking MRP8/14 to B. burgdorferi ex vivo. We demonstrated similar levels of B. burgdorferi dissemination, cytokine and immunoglobulin production in infected wt and mrp14-/- mice after 21 days. Neutrophils and monocytes lacking MRP8/14 were undiminished in their ability to become activated or phagocytose B. burgdorferi. In conclusion, we did not find a central role of MRP8/14 in the immune response against B. burgdorferi. As the levels of MRP8/14 in the serum of infected mice were low, we speculate that MRP8/14 is not released in levels great enough to influence the course of B. burgdorferi infection.


Subject(s)
Borrelia burgdorferi/physiology , Calgranulin A/metabolism , Calgranulin B/metabolism , Lyme Disease/immunology , Monocytes/immunology , Neutrophils/immunology , Animals , Calgranulin A/genetics , Calgranulin B/genetics , Disease Models, Animal , Female , Humans , Immunity, Innate , Lyme Disease/transmission , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis
7.
Methods Mol Biol ; 1690: 291-299, 2018.
Article in English | MEDLINE | ID: mdl-29032552

ABSTRACT

Dendritic cells (DCs) are professional antigen-presenting cells that recognize and phagocytose pathogens, and help to orchestrate adaptive immune responses to combat them. DCs are abundant in the skin where Borrelia burgdorferi first enters the body during a tick bite, and are thus critical in determining the initial stages of the innate and adaptive immune responses against Borrelia. Here, we describe two methods to study the response of DCs to Borrelia; an in vitro approach using monocyte-derived DCs (moDCs) and an ex vivo approach using a human skin model.


Subject(s)
Borrelia burgdorferi/immunology , Dendritic Cells/immunology , Dendritic Cells/microbiology , Lyme Disease/immunology , Phagocytosis , Cell Culture Techniques/methods , Cells, Cultured , Humans , Immunity, Cellular , Lyme Disease/microbiology , Organ Culture Techniques/methods , Skin/immunology , Skin/microbiology
8.
Nat Rev Dis Primers ; 3: 17062, 2017 Aug 03.
Article in English | MEDLINE | ID: mdl-28770818

ABSTRACT

This corrects the article DOI: 10.1038/nrdp.2016.90.

9.
Nat Rev Dis Primers ; 2: 16090, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27976670

ABSTRACT

Lyme borreliosis is a tick-borne disease that predominantly occurs in temperate regions of the northern hemisphere and is primarily caused by the bacterium Borrelia burgdorferi in North America and Borrelia afzelii or Borrelia garinii in Europe and Asia. Infection usually begins with an expanding skin lesion, known as erythema migrans (referred to as stage 1), which, if untreated, can be followed by early disseminated infection, particularly neurological abnormalities (stage 2), and by late infection, especially arthritis in North America or acrodermatitis chronica atrophicans in Europe (stage 3). However, the disease can present with any of these manifestations. During infection, the bacteria migrate through the host tissues, adhere to certain cells and can evade immune clearance. Yet, these organisms are eventually killed by both innate and adaptive immune responses and most inflammatory manifestations of the infection resolve. Except for patients with erythema migrans, Lyme borreliosis is diagnosed based on a characteristic clinical constellation of signs and symptoms with serological confirmation of infection. All manifestations of the infection can usually be treated with appropriate antibiotic regimens, but the disease can be followed by post-infectious sequelae in some patients. Prevention of Lyme borreliosis primarily involves the avoidance of tick bites by personal protective measures.


Subject(s)
Lyme Disease/diagnosis , Lyme Disease/physiopathology , Amoxicillin/pharmacology , Amoxicillin/therapeutic use , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Borrelia burgdorferi/immunology , Borrelia burgdorferi/pathogenicity , Borrelia burgdorferi Group/immunology , Borrelia burgdorferi Group/pathogenicity , Cefuroxime/analogs & derivatives , Cefuroxime/pharmacology , Cefuroxime/therapeutic use , Doxycycline/pharmacology , Doxycycline/therapeutic use , Exanthema/etiology , Humans , Ixodes/microbiology , Lyme Disease/epidemiology , Lyme Neuroborreliosis/complications , Lyme Neuroborreliosis/etiology , Lyme Neuroborreliosis/physiopathology , Risk Factors , Zoonoses/etiology , Zoonoses/microbiology , beta-Lactams/pharmacology , beta-Lactams/therapeutic use
10.
PLoS One ; 11(10): e0164040, 2016.
Article in English | MEDLINE | ID: mdl-27695100

ABSTRACT

Borrelia burgdorferi is transmitted into the skin of the host where it encounters and interacts with two dendritic cell (DC) subsets; Langerhans cells (LCs) and dermal DCs (DDCs). These cells recognize pathogens via pattern recognition receptors, mature and migrate out of the skin into draining lymph nodes, where they orchestrate adaptive immune responses. In order to investigate the response of skin DCs during the early immunopathogenesis of Lyme borreliosis, we injected B. burgdorferi intradermally into full-thickness human skin and studied the migration of DCs out of the skin, the activation profile and phenotype of migrated cells. We found a significant increase in the migration of LCs and DDCs in response to B. burgdorferi. Notably, migration was prevented by blocking TLR2. DCs migrated from skin inoculated with higher numbers of spirochetes expressed significantly higher levels of CD83 and produced pro-inflammatory cytokines. No difference was observed in the expression of HLA-DR, CD86, CD38, or CCR7. To conclude, we have established an ex vivo human skin model to study DC-B. burgdorferi interactions. Using this model, we have demonstrated that B. burgdorferi-induced DC migration is mediated by TLR2. Our findings underscore the utility of this model as a valuable tool to study immunity to spirochetal infections.


Subject(s)
Borrelia burgdorferi/physiology , Cell Movement/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Toll-Like Receptor 2/metabolism , Biomarkers , Cell Survival , Cytokines/metabolism , Humans , Lyme Disease/immunology , Lyme Disease/metabolism , Lyme Disease/microbiology , Skin/immunology , Skin/metabolism , Skin/microbiology
11.
Infect Genet Evol ; 42: 66-76, 2016 08.
Article in English | MEDLINE | ID: mdl-27125686

ABSTRACT

In this study we used typing based on the eight multilocus sequence typing scheme housekeeping genes (MLST) and 5S-23S rDNA intergenic spacer (IGS) to explore the population structure of Borrelia burgdorferi sensu lato isolates from patients with Lyme borreliosis (LB) and to test the association between the B. burgdorferi s.l. sequence types (ST) and the clinical manifestations they cause in humans. Isolates of B. burgdorferi from 183 LB cases across Europe, with distinct clinical manifestations, and 257 Ixodes ricinus lysates from The Netherlands, were analyzed for this study alone. For completeness, we incorporated in our analysis also 335 European B. burgdorferi s.l. MLST profiles retrieved from literature. Borrelia afzelii and Borrelia bavariensis were associated with human cases of LB while Borrelia garinii, Borrelia lusitaniae and Borrelia valaisiana were associated with questing I. ricinus ticks. B. afzelii was associated with acrodermatitis chronica atrophicans, while B. garinii and B. bavariensis were associated with neuroborreliosis. The samples in our study belonged to 251 different STs, of which 94 are newly described, adding to the overall picture of the genetic diversity of Borrelia genospecies. The fraction of STs that were isolated from human samples was significantly higher for the genospecies that are known to be maintained in enzootic cycles by mammals (B. afzelii, B. bavariensis, and Borrelia spielmanii) than for genospecies that are maintained by birds (B. garinii and B. valaisiana) or lizards (B. lusitaniae). We found six multilocus sequence types that were significantly associated to clinical manifestations in humans and five IGS haplotypes that were associated with the human LB cases. While IGS could perform just as well as the housekeeping genes in the MLST scheme for predicting the infectivity of B. burgdorferi s.l., the advantage of MLST is that it can also capture the differential invasiveness of the various STs.


Subject(s)
Arachnid Vectors/microbiology , Borrelia burgdorferi/genetics , DNA, Intergenic/genetics , Ixodes/microbiology , Lyme Disease/epidemiology , Animals , Borrelia burgdorferi/classification , Borrelia burgdorferi/isolation & purification , Genotype , Humans , Lyme Disease/diagnosis , Lyme Disease/microbiology , Male , Multilocus Sequence Typing , Netherlands/epidemiology , Phylogeny , RNA, Ribosomal, 23S/genetics , RNA, Ribosomal, 5S/genetics
12.
Vector Borne Zoonotic Dis ; 16(4): 223-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26901751

ABSTRACT

INTRODUCTION: We previously identified tick salivary lectin pathway inhibitor (TSLPI) in Ixodes scapularis, a vector for Borrelia burgdorferi sensu stricto (s.s.) in North America. TSLPI is a salivary protein facilitating B. burgdorferi s.s. transmission and acquisition by inhibiting the host lectin complement pathway through interference with mannose binding lectin (MBL) activity. Since Ixodes ricinus is the predominant vector for Lyme borreliosis in Europe and transmits several complement sensitive B. burgdorferi sensu lato (s.l.) strains, we aimed to identify, describe, and characterize the I. ricinus ortholog of TSLPI. METHODS: We performed (q)PCRs on I. ricinus salivary gland cDNA to identify a TSLPI ortholog. Next, we generated recombinant (r)TSLPI in a Drosophila expression system and examined inhibition of the MBL complement pathway and complement-mediated killing of B. burgdorferi s.l. in vitro. RESULTS: We identified a TSLPI ortholog in I. ricinus salivary glands with 93% homology at the RNA and 89% at the protein level compared to I. scapularis TSLPI, which was upregulated during tick feeding. In silico analysis revealed that TSLPI appears to be part of a larger family of Ixodes salivary proteins among which I. persulcatus basic tail salivary proteins and I. scapularis TSLPI and Salp14. I. ricinus rTSLPI inhibited the MBL complement pathway and protected B. burgdorferi s.s. and Borrelia garinii from complement-mediated killing. CONCLUSION: We have identified a TSLPI ortholog, which protects B. burgdorferi s.l. from complement-mediated killing in I. ricinus, the major vector for tick-borne diseases in Europe.


Subject(s)
Arthropod Proteins/biosynthesis , Borrelia burgdorferi Group/physiology , Ixodes/metabolism , Salivary Glands/metabolism , Salivary Proteins and Peptides/biosynthesis , Animals , Arachnid Vectors/metabolism , Arachnid Vectors/microbiology , Complement Pathway, Mannose-Binding Lectin , Humans , Ixodes/genetics , Ixodes/microbiology , Lyme Disease/immunology , Lyme Disease/metabolism , Lyme Disease/transmission , Salivary Glands/microbiology
13.
Ticks Tick Borne Dis ; 7(2): 371-7, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26739030

ABSTRACT

Only a few reported cases indicate that Rickettsia helvetica and Rickettsia monacensis can cause disease in humans. Exposure to these two spotted fever group (SFG) rickettsiae occurs through bites of Ixodes ricinus, also the primary vector of Lyme borreliosis in Europe. To date, it is unclear how often exposure to these two microorganisms results in infection or disease. We show that of all the Borrelia burgdorferi s.l.-positive ticks, 25% were co-infected with rickettsiae. Predominantly R. helvetica was detected while R. monacensis was only found in approximately 2% of the ticks. In addition, exposure to tick-borne pathogens was compared by serology in healthy blood donors, erythema migrans (EM)-patients, and patients suspected of Lyme neuroborreliosis (LNB). As could be expected, seroreactivity against B. burgdorferi sensu lato was lower in blood donors (6%) compared to EM patients (34%) and suspected LNB cases (64%). Interestingly, seroreactivity against SFG Rickettsia antigens was not detected in serum samples from blood donors (0%), but 6% of the EM patients and 21% of the LNB suspects showed anti-rickettsial antibodies. Finally, the presence of B. burgdorferi s.l. and Rickettsia spp. in cerebrospinal fluid samples of a large cohort of patients suspected of LNB (n=208) was investigated by PCR. DNA of B. burgdorferi s.l., R. helvetica and R. monacensis was detected in seventeen, four and one patient, respectively. In conclusion, our data show that B. burgdorferi s.l. and SFG rickettsiae co-infection occurs in Dutch I. ricinus and that Lyme borreliosis patients, or patients suspected of Lyme borreliosis, are indeed exposed to both tick-borne pathogens. Whether SFG rickettsiae actually cause disease, and whether co-infections alter the clinical course of Lyme borreliosis, is not clear from our data, and warrants further investigation.


Subject(s)
Borrelia burgdorferi Group/isolation & purification , Lyme Disease/microbiology , Rickettsia Infections/microbiology , Rickettsia/isolation & purification , Tick-Borne Diseases/microbiology , Adult , Aged , Animals , Arachnid Vectors/microbiology , Base Sequence , Borrelia burgdorferi Group/genetics , Borrelia burgdorferi Group/immunology , Coinfection , Female , Humans , Ixodes/microbiology , Lyme Disease/epidemiology , Male , Middle Aged , Netherlands/epidemiology , Rickettsia/genetics , Rickettsia/immunology , Rickettsia Infections/epidemiology , Sequence Alignment , Tick-Borne Diseases/epidemiology
14.
Vaccine ; 34(8): 1025-33, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26801064

ABSTRACT

Outer Membrane Vesicles (OMVs) are gaining attention as vaccine candidates. The successful expression of heterologous antigens in OMVs, with the OMV functioning both as adjuvant and delivery vehicle, has greatly enhanced their vaccine potential. Since there are indications that surface exposed antigens might induce a superior immune response, targeting of heterologous antigens to the OMV surface is of special interest. Several systems for surface display of heterologous antigens on OMVs have been developed. However, these systems have not been used to display lipidated membrane-associated proteins known as lipoproteins, which are emerging as key targets for protective immunity. We were therefore interested to see whether we could express a foreign lipoprotein on the outer surface of OMVs. When outer surface protein A (OspA), a borrelial surface-exposed lipoprotein, was expressed in meningococci, it was found that although OspA was present in OMVs, it was no longer surface-exposed. Therefore, a set of fusions of OspA to different regions of factor H binding protein (fHbp), a meningococcal surface-exposed lipoprotein, were designed and tested for their surface-exposure. An N-terminal part of fHbp was found to be necessary for the successful surface display of OspA on meningococcal OMVs. When mice were immunized with this set of OMVs, an OspA-specific antibody response was only elicited by OMVs with clearly surface-exposed OspA, strengthening the idea that the exact positioning of an antigen in the OMV affects the immune response. This method for the surface display of heterologous lipoproteins on OMVs is a step forward in the development of OMVs as a vaccine platform.


Subject(s)
Antigens, Surface/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Lipoproteins/immunology , Neisseria meningitidis/immunology , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Borrelia burgdorferi , Female , Mice , Mice, Inbred BALB C
16.
Immunobiology ; 220(10): 1141-50, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26093919

ABSTRACT

Borrelia burgdorferi can be categorized based on restriction fragment length polymorphism analysis into ribosomal spacer type (RST) 1, 2 and 3. A correlation between RST type and invasiveness of Borrelia isolates has been demonstrated in clinical studies and experimental models, and RST 1 isolates are more likely to cause disseminated disease than RST 3 isolates. We hypothesized that this could partially be due to increased susceptibility of RST 3 isolates to killing by the innate immune system early in infection. Thus, we investigated the interaction of five RST 1 and five RST 3 isolates with various components of the human innate immune system in vitro. RST 3 isolates induced significantly greater upregulation of activation markers in monocyte-derived dendritic cells compared to RST 1 isolates at a low multiplicity of infection. However, RST 1 isolates stimulated greater interleukin-6 production. At a high multiplicity of infection no differences in dendritic cell activation or cytokine production were observed. In addition, we observed no differences in the ability of RST 1 and RST 3 isolates to activate monocytes or neutrophils and all strains were phagocytosed at a comparable rate. Finally, all isolates tested were equally resistant to complement-mediated killing, as determined by dark-field microscopy and a growth inhibition assay. In conclusion, we demonstrate that the RST 1 and 3 isolates showed no distinction in their susceptibility to the various components of the human immune system studied here, suggesting that other factors are responsible for their differential invasiveness.


Subject(s)
Borrelia burgdorferi/immunology , Genotype , Immunity, Innate , Interleukin-6/immunology , Lyme Disease/immunology , Borrelia burgdorferi/genetics , Borrelia burgdorferi/isolation & purification , Complement System Proteins/immunology , Female , Humans , Lyme Disease/genetics , Lyme Disease/pathology , Male
17.
Trends Parasitol ; 31(6): 260-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25892254

ABSTRACT

Borrelia miyamotoi is a relapsing fever spirochete that has only recently been identified as a human pathogen. Borrelia miyamotoi is genetically and ecologically distinct from Borrelia burgdorferi sensu lato, while both are present in Ixodes ticks. Over 50 patients with an acute febrile illness have been described with a B. miyamotoi infection, and two infected immunocompromised patients developed a meningoencephalitis. Seroprevalence studies indicate exposure in the general population and in specific risk groups, such as patients initially suspected of having human granulocytic anaplasmosis. Here, we review the available literature on B. miyamotoi, describing its presence in ticks, reservoir hosts, and humans, and discussing its potential impact on public health.


Subject(s)
Borrelia Infections/transmission , Borrelia/physiology , Ixodes/parasitology , Animals , Arachnid Vectors/parasitology , Borrelia Infections/diagnosis , Borrelia Infections/epidemiology , Borrelia Infections/pathology , Borrelia Infections/prevention & control , Disease Reservoirs , Humans , Immunocompromised Host , Relapsing Fever/diagnosis , Relapsing Fever/epidemiology , Relapsing Fever/prevention & control , Relapsing Fever/transmission
18.
Ned Tijdschr Geneeskd ; 158: A7986, 2014.
Article in Dutch | MEDLINE | ID: mdl-25248736

ABSTRACT

The prevalence of ticks seems to have increased with time, and the number of patients with Lyme disease in the Netherlands is also increasing. Lyme disease and other tick-transmitted diseases now attract a lot of attention. Several national initiatives at different levels are now in progress, with the aim of suppressing Lyme disease and providing better care for patients with indications of having these diseases. A more uniform approach between different treatment centres, joint research and further expansion of education and continuing education for physicians and the public could lead to further improvement.


Subject(s)
Arachnid Vectors/microbiology , Lyme Disease/epidemiology , Lyme Disease/prevention & control , Ticks/microbiology , Animals , Humans , Lyme Disease/transmission , Netherlands/epidemiology , Prevalence , Risk Factors
19.
Parasit Vectors ; 7: 77, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24559082

ABSTRACT

Ixodes ricinus transmits bacterial, protozoal and viral pathogens, causing disease and forming an increasing health concern in Europe. ANTIDotE is an European Commission funded consortium of seven institutes, which aims to identify and characterize tick proteins involved in feeding and pathogen transmission. The knowledge gained will be used to develop and evaluate anti-tick vaccines that may prevent multiple human tick-borne diseases. Strategies encompassing anti-tick vaccines to prevent transmission of pathogens to humans, animals or wildlife will be developed with relevant stakeholders with the ultimate aim of reducing the incidence of tick-borne diseases in humans.


Subject(s)
Arthropod Proteins/immunology , Tick Infestations/prevention & control , Tick-Borne Diseases/prevention & control , Ticks/immunology , Vaccines , Animals , Antidotes , Arthropod Proteins/isolation & purification , Europe/epidemiology , Humans , Tick Infestations/epidemiology , Tick-Borne Diseases/epidemiology
20.
Trends Parasitol ; 30(2): 95-103, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24388562

ABSTRACT

Borrelia burgdorferi sensu lato, the causative agent of Lyme borreliosis, is inoculated into the skin during an Ixodes tick bite where it is recognised and captured by dendritic cells (DCs). However, considering the propensity of Borrelia to disseminate, it would appear that DCs fall short in mounting a robust immune response against it. Many aspects of the DC-driven immune response to Borrelia have been examined. Recently, components of tick saliva have been identified that sabotage DC responses and aid Borrelia infection. In this review, we summarise what is currently known about the immune response of DCs to Borrelia and explore the mechanisms by which Borrelia manages to circumvent this immune response, with or without the help of tick salivary proteins.


Subject(s)
Borrelia burgdorferi/immunology , Dendritic Cells/immunology , Dendritic Cells/microbiology , Ixodes/immunology , Lyme Disease/immunology , Saliva/immunology , Animals , Humans , Ixodes/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...