Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
1.
Vaccine ; 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38880695

ABSTRACT

Anaplasma marginale is a tick-borne pathogen of cattle that causes bovine anaplasmosis in tropical and subtropical regions throughout the world. Killed vaccines derived from infected erythrocytes have been used for control of this disease with limited success. Recently, we described a targeted deletion mutation in the phage head-to-tail connector protein gene of A. marginale which caused bacterial attenuation in vivo and provided protection as a modified live vaccine (MLAV). Following intravenous injection of susceptible steers, the MLAV induced protective immunity against disease progression. In the current study, we demonstrated that the immunity resulting from MLAV in cattle prevents the disease progression resulting from virulent A. marginale intrastadial transmission from infected Dermacentor variabilis male ticks. The nonimmunized control steers receiving the infection from ticks developed fever, lethargy, and inappetence for several days post tick exposure with significant decreases in the packed cell volume and increases in bacteremia. In contrast, the MLAV immunized steers remained healthy after being challenged with infected ticks and this group of animals had a significant reduction in bacteremia as compared with the controls. This study demonstrated that the A. marginale MLAV provided protection against acute tick-transmitted anaplasmosis, in addition to protection documented in steers challenge-exposed with infected blood as reported previously.

2.
Pathogens ; 11(8)2022 Jul 23.
Article in English | MEDLINE | ID: mdl-35894050

ABSTRACT

Over the past two decades, RNA interference (RNAi) in ticks, in combination with omics technologies, have greatly advanced the discovery of tick gene and molecular function. While mechanisms of RNAi were initially elucidated in plants, fungi, and nematodes, the classic 2002 study by Aljamali et al. was the first to demonstrate RNAi gene silencing in ticks. Subsequently, applications of RNAi have led to the discovery of genes that impact tick function and tick-host-pathogen interactions. RNAi will continue to lead to the discovery of an array of tick genes and molecules suitable for the development of vaccines and/or pharmacologic approaches for tick control and the prevention of pathogen transmission.

3.
Expert Rev Proteomics ; 18(12): 1099-1116, 2021 12.
Article in English | MEDLINE | ID: mdl-34904495

ABSTRACT

BACKGROUND: Ticks are obligate hematophagous arthropods that synthesize the glycan Galα1-3Galß1-(3)4GlcNAc-R (α-Gal) associated with the alpha-gal syndrome (AGS) or allergy to mammalian meat consumption. RESEARCH DESIGN AND METHODS: In this study, we used a proteomics approach to characterize tick proteins in salivary glands (sialome SG), secreted saliva (sialome SA) and with α-Gal modification (alphagalactome SG and SA) in model tick species associated with the AGS in the United States (Amblyomma americanum) and Australia (Ixodes holocyclus). Selected proteins reactive to sera (IgE) from patients with AGS were identified to advance in the identification of possible proteins associated with the AGS. For comparative analysis, the α-Gal content was measured in various tick species. RESULTS: The results confirmed that ticks produce proteins with α-Gal modifications and secreted into saliva during feeding. Proteins identified in tick alphagalactome SA by sera from patients with severe AGS symptomatology may constitute candidate disease biomarkers. CONCLUSIONS: The results support the presence of tick-derived proteins with α-Gal modifications in the saliva with potential implications in AGS and other disorders and protective capacity against tick infestations and pathogen infection. Future research should focus on the characterization of the function of tick glycoproteins with α-Gal in tick biology and AGS.


Subject(s)
Saliva , Ticks , Animals , Biomarkers , Food Hypersensitivity , Humans , Salivary Glands
4.
Article in English | MEDLINE | ID: mdl-28439499

ABSTRACT

Ticks and the pathogens they transmit constitute a growing burden for human and animal health worldwide. Vector competence is a component of vectorial capacity and depends on genetic determinants affecting the ability of a vector to transmit a pathogen. These determinants affect traits such as tick-host-pathogen and susceptibility to pathogen infection. Therefore, the elucidation of the mechanisms involved in tick-pathogen interactions that affect vector competence is essential for the identification of molecular drivers for tick-borne diseases. In this review, we provide a comprehensive overview of tick-pathogen molecular interactions for bacteria, viruses, and protozoa affecting human and animal health. Additionally, the impact of tick microbiome on these interactions was considered. Results show that different pathogens evolved similar strategies such as manipulation of the immune response to infect vectors and facilitate multiplication and transmission. Furthermore, some of these strategies may be used by pathogens to infect both tick and mammalian hosts. Identification of interactions that promote tick survival, spread, and pathogen transmission provides the opportunity to disrupt these interactions and lead to a reduction in tick burden and the prevalence of tick-borne diseases. Targeting some of the similar mechanisms used by the pathogens for infection and transmission by ticks may assist in development of preventative strategies against multiple tick-borne diseases.


Subject(s)
Arachnid Vectors/microbiology , Arachnid Vectors/virology , Disease Transmission, Infectious , Host-Pathogen Interactions , Tick-Borne Diseases/epidemiology , Ticks/physiology , Animals , Arachnid Vectors/parasitology , Humans , Ticks/microbiology , Ticks/parasitology , Ticks/virology
5.
Nat Commun ; 7: 10507, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26856261

ABSTRACT

Ticks transmit more pathogens to humans and animals than any other arthropod. We describe the 2.1 Gbp nuclear genome of the tick, Ixodes scapularis (Say), which vectors pathogens that cause Lyme disease, human granulocytic anaplasmosis, babesiosis and other diseases. The large genome reflects accumulation of repetitive DNA, new lineages of retro-transposons, and gene architecture patterns resembling ancient metazoans rather than pancrustaceans. Annotation of scaffolds representing ∼57% of the genome, reveals 20,486 protein-coding genes and expansions of gene families associated with tick-host interactions. We report insights from genome analyses into parasitic processes unique to ticks, including host 'questing', prolonged feeding, cuticle synthesis, blood meal concentration, novel methods of haemoglobin digestion, haem detoxification, vitellogenesis and prolonged off-host survival. We identify proteins associated with the agent of human granulocytic anaplasmosis, an emerging disease, and the encephalitis-causing Langat virus, and a population structure correlated to life-history traits and transmission of the Lyme disease agent.


Subject(s)
Anaplasma phagocytophilum , Arachnid Vectors/genetics , Genome/genetics , Ixodes/genetics , Ligand-Gated Ion Channels/genetics , Animals , Gene Expression Profiling , Genomics , Lyme Disease/transmission , Oocytes , Xenopus laevis
6.
Trends Microbiol ; 24(3): 173-180, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26718986

ABSTRACT

The tick-borne rickettsial pathogen Anaplasma phagocytophilum develops within membrane-bound inclusions in the host cell cytoplasm. This pathogen has evolved with its tick and vertebrate hosts through dynamic processes involving genetic traits of the pathogen and hosts that collectively mediate pathogen infection, development, persistence, and survival. Herein, we challenge the evidence of tick-host-pathogen coevolution by hypothesizing that A. phagocytophilum utilizes common molecular mechanisms for infection in both vertebrate and tick cells, including remodeling of the cytoskeleton, inhibition of cell apoptosis, and manipulation of the immune response. The discovery of these common mechanisms provides evidence that a control strategy could be developed targeted at both vertebrate and tick hosts for more complete control of A. phagocytophilum and its associated diseases.


Subject(s)
Anaplasma phagocytophilum/physiology , Anaplasmosis/microbiology , Host-Pathogen Interactions , Ticks/microbiology , Vertebrates/microbiology , Anaplasma phagocytophilum/pathogenicity , Animals , Apoptosis , Bacterial Proteins , Cytoskeleton/chemistry , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Neutrophils/microbiology , Vertebrates/immunology
7.
Parasit Vectors ; 8: 592, 2015 Nov 14.
Article in English | MEDLINE | ID: mdl-26576940

ABSTRACT

Ticks (Acari: Ixodida) are arthropod ectoparasites dependent on a bloodmeal from a vertebrate host at each developmental stage for completion of their life cycle. This tick feeding cycle impacts animal health by causing damage to hides, secondary infections, immune reactions and diseases caused by transmission of pathogens. The genus Ixodes includes several medically important species that vector diseases, including granulocytic anaplasmosis and Lyme disease. I. scapularis, commonly called the black-legged or deer tick, is a medically-important tick species in North America and therefore was the first tick genome to be sequenced, thus serving as an important resource for tick research. This Primer focuses on the normal developmental cycle and laboratory rearing of I. scapularis. Definition of normal morphology, along with a consistent source of laboratory-reared I. scapularis, are fundamental for all aspects of future research, especially the effects of genetic manipulation and the evaluation of tick vaccine efficacy. Recent research important for the advancement of tick research, namely the development of tick cell culture systems for study of ticks and tick-borne pathogens, RNA interference for genetic manipulation of ticks and discovery of candidate antigens for development of tick vaccines, are briefly presented along with areas to target for future research.


Subject(s)
Entomology/methods , Ixodes/anatomy & histology , Ixodes/growth & development , Life Cycle Stages , Animals
8.
PLoS One ; 10(9): e0137237, 2015.
Article in English | MEDLINE | ID: mdl-26340562

ABSTRACT

Anaplasma phagocytophilum is an emerging zoonotic pathogen transmitted by Ixodes scapularis that causes human granulocytic anaplasmosis. Here, a high throughput quantitative proteomics approach was used to characterize A. phagocytophilum proteome during rickettsial multiplication and identify proteins involved in infection of the tick vector, I. scapularis. The first step in this research was focused on tick cells infected with A. phagocytophilum and sampled at two time points containing 10-15% and 65-71% infected cells, respectively to identify key bacterial proteins over-represented in high percentage infected cells. The second step was focused on adult female tick guts and salivary glands infected with A. phagocytophilum to compare in vitro results with those occurring during bacterial infection in vivo. The results showed differences in the proteome of A. phagocytophilum in infected ticks with higher impact on protein synthesis and processing than on bacterial replication in tick salivary glands. These results correlated well with the developmental cycle of A. phagocytophilum, in which cells convert from an intracellular reticulated, replicative form to the nondividing infectious dense-core form. The analysis of A. phagocytophilum differentially represented proteins identified stress response (GroEL, HSP70) and surface (MSP4) proteins that were over-represented in high percentage infected tick cells and salivary glands when compared to low percentage infected cells and guts, respectively. The results demonstrated that MSP4, GroEL and HSP70 interact and bind to tick cells, thus playing a role in rickettsia-tick interactions. The most important finding of these studies is the increase in the level of certain bacterial stress response and surface proteins in A. phagocytophilum-infected tick cells and salivary glands with functional implication in tick-pathogen interactions. These results gave a new dimension to the role of these stress response and surface proteins during A. phagocytophilum infection in ticks. Characterization of Anaplasma proteome contributes information on host-pathogen interactions and provides targets for development of novel control strategies for pathogen infection and transmission.


Subject(s)
Bacterial Proteins/genetics , Chaperonin 60/genetics , HSP70 Heat-Shock Proteins/genetics , Ixodes/microbiology , Membrane Proteins/genetics , Proteome/genetics , Anaplasma phagocytophilum , Animals , Bacterial Proteins/metabolism , Chaperonin 60/metabolism , Female , Gastrointestinal Tract/microbiology , Gene Expression Profiling , Gene Expression Regulation , HSP70 Heat-Shock Proteins/metabolism , Host-Pathogen Interactions , Membrane Proteins/metabolism , Molecular Sequence Annotation , Proteome/metabolism , Salivary Glands/microbiology , Signal Transduction , Stress, Physiological
9.
PLoS One ; 10(7): e0133038, 2015.
Article in English | MEDLINE | ID: mdl-26186700

ABSTRACT

Tudor staphylococcal nuclease (Tudor-SN) and Argonaute (Ago) are conserved components of the basic RNA interference (RNAi) machinery with a variety of functions including immune response and gene regulation. The RNAi machinery has been characterized in tick vectors of human and animal diseases but information is not available on the role of Tudor-SN in tick RNAi and other cellular processes. Our hypothesis is that tick Tudor-SN is part of the RNAi machinery and may be involved in innate immune response and other cellular processes. To address this hypothesis, Ixodes scapularis and I. ricinus ticks and/or cell lines were used to annotate and characterize the role of Tudor-SN in dsRNA-mediated RNAi, immune response to infection with the rickettsia Anaplasma phagocytophilum and the flaviviruses TBEV or LGTV and tick feeding. The results showed that Tudor-SN is conserved in ticks and involved in dsRNA-mediated RNAi and tick feeding but not in defense against infection with the examined viral and rickettsial pathogens. The effect of Tudor-SN gene knockdown on tick feeding could be due to down-regulation of genes that are required for protein processing and blood digestion through a mechanism that may involve selective degradation of dsRNAs enriched in G:U pairs that form as a result of adenosine-to-inosine RNA editing. These results demonstrated that Tudor-SN plays a role in tick RNAi pathway and feeding but no strong evidence for a role in innate immune responses to pathogen infection was found.


Subject(s)
Anaplasma phagocytophilum/pathogenicity , Flavivirus/pathogenicity , Ixodes/genetics , Nuclear Proteins/genetics , RNA Interference , Amino Acid Sequence , Animals , Cell Line , Conserved Sequence , Cricetinae , Ixodes/parasitology , Ixodes/virology , Molecular Sequence Data , Nuclear Proteins/metabolism , Phylogeny , Transcriptome
10.
Ticks Tick Borne Dis ; 6(4): 522-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25943785

ABSTRACT

Anaplasma marginale is an economically important tick-borne pathogen of cattle that causes bovine anaplasmosis. A wide range of geographic strains of A. marginale have been isolated from cattle, several of which have been characterized using genomics and proteomics. While many of these strains have been propagated in tick lines, comparative analyses after propagation in tick cells have not been reported. The overall purpose of this research therefore was to compare the degree of conservation of selected genes after propagation in tick cell culture among A. marginale strains from the U.S. (the Virginia strain) and Brazil (UFMG1 and UFMG2 strains). The genes studied herein included those which encode the proteins HSP70 and SODB involved in heat shock and stress responses, respectively, and two genes that encode major surface proteins MSP4 and MSP5. Strain identities were first confirmed by sequencing the tandem repeats of the msp1a gene which encodes for the adhesin, MSP1a. The results of these studies demonstrated that the genes encoding for both stress response and heat shock proteins were highly conserved among the three A. marginale strains. Antibodies specific for MSP4, MSP5, SODB and HSP70 proteins were used to further characterize the A. marginale strains, and they reacted with all of these strains propagated in tick cell culture, providing further evidence for antigenic conservation. Although antigenic differences were not found among the three A. marginale strains, multi-locus sequence analysis (MLSA) performed with nucleotide sequences of these genes demonstrated that the A. marginale Brazilian and U.S. strains fall in different clades. These results showed that phylogenetically distant strains of A. marginale are antigenically conserved, even after several in vitro passages, supporting the use of some of the above conserved proteins as candidates for universal vaccines.


Subject(s)
Anaplasma marginale/isolation & purification , Anaplasmosis/immunology , Arachnid Vectors/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Cattle Diseases/immunology , Ticks/microbiology , Anaplasma marginale/classification , Anaplasma marginale/genetics , Anaplasma marginale/growth & development , Anaplasmosis/microbiology , Animals , Antigenic Variation , Brazil , Cattle , Cattle Diseases/microbiology , Conserved Sequence , Molecular Sequence Data , Phylogeny , United States
11.
PLoS Genet ; 11(3): e1005120, 2015 03.
Article in English | MEDLINE | ID: mdl-25815810

ABSTRACT

Anaplasma phagocytophilum is an emerging pathogen that causes human granulocytic anaplasmosis. Infection with this zoonotic pathogen affects cell function in both vertebrate host and the tick vector, Ixodes scapularis. Global tissue-specific response and apoptosis signaling pathways were characterized in I. scapularis nymphs and adult female midguts and salivary glands infected with A. phagocytophilum using a systems biology approach combining transcriptomics and proteomics. Apoptosis was selected for pathway-focused analysis due to its role in bacterial infection of tick cells. The results showed tissue-specific differences in tick response to infection and revealed differentiated regulation of apoptosis pathways. The impact of bacterial infection was more pronounced in tick nymphs and midguts than in salivary glands, probably reflecting bacterial developmental cycle. All apoptosis pathways described in other organisms were identified in I. scapularis, except for the absence of the Perforin ortholog. Functional characterization using RNA interference showed that Porin knockdown significantly increases tick colonization by A. phagocytophilum. Infection with A. phagocytophilum produced complex tissue-specific alterations in transcript and protein levels. In tick nymphs, the results suggested a possible effect of bacterial infection on the inhibition of tick immune response. In tick midguts, the results suggested that A. phagocytophilum infection inhibited cell apoptosis to facilitate and establish infection through up-regulation of the JAK/STAT pathway. Bacterial infection inhibited the intrinsic apoptosis pathway in tick salivary glands by down-regulating Porin expression that resulted in the inhibition of Cytochrome c release as the anti-apoptotic mechanism to facilitate bacterial infection. However, tick salivary glands may promote apoptosis to limit bacterial infection through induction of the extrinsic apoptosis pathway. These dynamic changes in response to A. phagocytophilum in I. scapularis tissue-specific transcriptome and proteome demonstrated the complexity of the tick response to infection and will contribute to characterize gene regulation in ticks.


Subject(s)
Anaplasma phagocytophilum/genetics , Anaplasmosis/genetics , Apoptosis/genetics , Systems Biology , Anaplasma phagocytophilum/pathogenicity , Anaplasmosis/microbiology , Anaplasmosis/transmission , Animals , Cell Differentiation/genetics , Female , Gene Expression Regulation , Humans , Insect Vectors/genetics , Insect Vectors/microbiology , Ixodes/microbiology , Organ Specificity , RNA Interference , Salivary Glands/metabolism , Salivary Glands/microbiology , Signal Transduction/genetics , Transcriptome/genetics
12.
Ticks Tick Borne Dis ; 5(6): 744-52, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25127160

ABSTRACT

Anaplasma phagocytophilum, transmitted by ticks of the genus Ixodes, was first described in Scotland as the agent of tick-borne fever in sheep and more recently as the cause of human granulocytic anaplasmosis in the U.S. and Europe. We previously reported sheep as an experimental host for the human NY-18 isolate of A. phagocytophilum. While clinical signs were not observed and infected granulocytes were not seen in stained blood smears, these sheep served as a good host for infection of ticks. In this research we characterized tick feeding sites to better understand tick/host/pathogen interactions. Ixodes scapularis adults were allowed to feed for 2 and 4 days on experimentally infected sheep, after which biopsies were taken beneath tick feeding sites for histopathology, PCR and immunohistochemistry (IHC) studies. In addition, the expression of selected immune response genes was studied in blood and feeding site biopsies. While necrosis was too advanced in 4-day biopsies for accurate cell counts, higher numbers of eosinophils and neutrophils were found in 2-day biopsies from infected sheep as compared with the uninfected controls. An unexpected result was the documentation of higher dermal inflammation in infected sheep at sites without ticks. A. phagocytophilum infected granulocytes were localized by immunohistochemistry (IHC) in skin biopsies using rabbit antibodies against the recombinant A. phagocytophilum major surface protein 4 as the primary antibody for indirect peroxidase-anti-peroxidase and fluorescent antibody IHC. These infected cells are likely to be the source of infection for ticks. Sheep therefore served as good hosts for studying host/pathogen/tick interactions of this human strain of A. phagocytophilum, and provided a means of producing infected ticks for future studies on tick/pathogen developmental and transmission cycles.


Subject(s)
Anaplasma phagocytophilum/physiology , Anaplasmosis/transmission , Ehrlichiosis/transmission , Host-Pathogen Interactions , Ixodes/microbiology , Sheep Diseases/transmission , Anaplasmosis/microbiology , Animals , Ehrlichiosis/microbiology , Female , Humans , Male , Models, Animal , Sheep , Sheep Diseases/microbiology , Zoonoses
13.
Ticks Tick Borne Dis ; 5(3): 225-33, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24548526

ABSTRACT

Control of ticks on dogs is often done by application of repellents that contain permethrin as the active ingredient. In this research, we studied the role of a glutathione S-transferase (GST) gene in detoxification of permethrin by ticks using a gene silencing method RNA interference (RNAi). The brown dog tick, Rhipicephalus sanguineus, used in these studies, has a notable host preference for dogs, but also infests other mammals. In this research, R. sanguineus females were injected with gst double-stranded RNA (dsRNA) to effect gene silencing by RNAi and then exposed to sublethal doses of permethrin. Sixty hours after injection, the females were allowed to feed on sheep. The female ticks subjected to RNAi proved to be more susceptible to permethrin than the untreated controls. The effect of gene silencing was most notable in the highest dose group (50.3 ppm) in which all ticks died, while in the corresponding controls that were not subjected to RNAi this dose was not lethal. The acaricide treatment of the ticks resulted in a change in tick attachment behavior. Acaricide-treated ticks attached in a scattered pattern in contrast to the control ticks that attached and fed tightly clustered together. The time required for repletion for both the injected and non-injected females exposed to the higher permethrin level was shorter than that observed in the lower-dose groups and unexposed controls, and this more rapid attachment and feeding would likely favor more rapid transmission of pathogens. However, engorgement and egg mass weights were not significantly different among the experimental groups. This research demonstrated that the silencing of the gst gene increased the tick's susceptibility to permethrin. Overall, these results have contributed to our understanding of the detoxification mechanism of ticks and provide new considerations for the formulation of treatment strategies.


Subject(s)
Acaricides/administration & dosage , Glutathione Transferase/genetics , Permethrin/administration & dosage , Rhipicephalus sanguineus/enzymology , Sheep Diseases/parasitology , Tick Infestations/veterinary , Animals , Base Sequence , Feeding Behavior , Female , Inactivation, Metabolic , Models, Biological , Molecular Sequence Data , Oviposition , RNA Interference , Rhipicephalus sanguineus/drug effects , Rhipicephalus sanguineus/physiology , Sequence Analysis, DNA/veterinary , Sheep , Tick Infestations/parasitology
14.
PLoS One ; 8(6): e65243, 2013.
Article in English | MEDLINE | ID: mdl-23776456

ABSTRACT

Bovine anaplasmosis is caused by cattle infection with the tick-borne bacterium, Anaplasma marginale. The major surface protein 1a (MSP1a) has been used as a genetic marker for identifying A. marginale strains based on N-terminal tandem repeats and a 5'-UTR microsatellite located in the msp1a gene. The MSP1a tandem repeats contain immune relevant elements and functional domains that bind to bovine erythrocytes and tick cells, thus providing information about the evolution of host-pathogen and vector-pathogen interactions. Here we propose one nomenclature for A. marginale strain classification based on MSP1a. All tandem repeats among A. marginale strains were classified and the amino acid variability/frequency in each position was determined. The sequence variation at immunodominant B cell epitopes was determined and the secondary (2D) structure of the tandem repeats was modeled. A total of 224 different strains of A. marginale were classified, showing 11 genotypes based on the 5'-UTR microsatellite and 193 different tandem repeats with high amino acid variability per position. Our results showed phylogenetic correlation between MSP1a sequence, secondary structure, B-cell epitope composition and tick transmissibility of A. marginale strains. The analysis of MSP1a sequences provides relevant information about the biology of A. marginale to design vaccines with a cross-protective capacity based on MSP1a B-cell epitopes.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/immunology , 5' Untranslated Regions/genetics , Animals , Bacterial Outer Membrane Proteins/genetics , Cattle , Computational Biology , Epitopes, B-Lymphocyte/genetics , Genotype , Microsatellite Repeats/genetics , Protein Structure, Secondary , Tandem Repeat Sequences/genetics
15.
PLoS One ; 8(6): e65915, 2013.
Article in English | MEDLINE | ID: mdl-23776567

ABSTRACT

BACKGROUND: Tick Subolesin and its ortholog in insects and vertebrates, Akirin, have been suggested to play a role in the immune response through regulation of nuclear factor-kappa B (NF-kB)-dependent and independent gene expression via interaction with intermediate proteins that interact with NF-kB and other regulatory proteins, bind DNA or remodel chromatin to regulate gene expression. The objective of this study was to characterize the structure and regulation of subolesin in Ixodes scapularis. I. scapularis is a vector of emerging pathogens such as Borrelia burgdorferi, Anaplasma phagocytophilum and Babesia microti that cause in humans Lyme disease, anaplasmosis and babesiosis, respectively. The genome of I. scapularis was recently sequenced, and this tick serves as a model organism for the study of vector-host-pathogen interactions. However, basic biological questions such as gene organization and regulation are largely unknown in ticks and other arthropod vectors. PRINCIPAL FINDINGS: The results presented here provide evidence that subolesin/akirin are evolutionarily conserved at several levels (primary sequence, gene organization and function), thus supporting their crucial biological function in metazoans. These results showed that NF-kB (Relish) is involved in the regulation of subolesin expression in ticks, suggesting that as in other organisms, different NF-kB integral subunits and/or unknown interacting proteins regulate the specificity of the NF-kB-mediated gene expression. These results suggested a regulatory network involving cross-regulation between NF-kB (Relish) and Subolesin and Subolesin auto-regulation with possible implications in tick immune response to bacterial infection. SIGNIFICANCE: These results advance our understanding of gene organization and regulation in I. scapularis and have important implications for arthropod vectors genetics and immunology highlighting the possible role of NF-kB and Subolesin/Akirin in vector-pathogen interactions and for designing new strategies for the control of vector infestations and pathogen transmission.


Subject(s)
Antigens/genetics , Arthropod Proteins/genetics , Arthropod Vectors/metabolism , Gene Expression Regulation/immunology , Gene Regulatory Networks/immunology , Ixodes/metabolism , NF-kappa B/metabolism , Animals , Antigens/metabolism , Arthropod Proteins/metabolism , Base Sequence , Conserved Sequence/genetics , DNA Primers/genetics , Electrophoresis, Capillary , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Gene Components , Ixodes/immunology , Models, Biological , Molecular Sequence Data , RNA Interference , Sequence Analysis, DNA
16.
Infect Immun ; 81(7): 2415-25, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23630955

ABSTRACT

Anaplasma phagocytophilum causes human granulocytic anaplasmosis. Infection with this zoonotic pathogen affects gene expression in both the vertebrate host and the tick vector, Ixodes scapularis. Here, we identified new genes, including spectrin alpha chain or alpha-fodrin (CG8) and voltage-dependent anion-selective channel or mitochondrial porin (T2), that are involved in A. phagocytophilum infection/multiplication and the tick cell response to infection. The pathogen downregulated the expression of CG8 in tick salivary glands and T2 in both the gut and salivary glands to inhibit apoptosis as a mechanism to subvert host cell defenses and increase infection. In the gut, the tick response to infection through CG8 upregulation was used by the pathogen to increase infection due to the cytoskeleton rearrangement that is required for pathogen infection. These results increase our understanding of the role of tick genes during A. phagocytophilum infection and multiplication and demonstrate that the pathogen uses similar strategies to establish infection in both vertebrate and invertebrate hosts.


Subject(s)
Anaplasma phagocytophilum/pathogenicity , Apoptosis , Carrier Proteins/metabolism , Cytoskeleton/metabolism , Ixodes/microbiology , Microfilament Proteins/metabolism , Anaplasma phagocytophilum/genetics , Animals , Carrier Proteins/genetics , Caspase 9/genetics , Caspase 9/metabolism , Cell Line , Feeding Behavior , Female , Gastrointestinal Tract/microbiology , Gene Expression Regulation , Gene Knockdown Techniques , Host-Pathogen Interactions , Ixodes/genetics , Ixodes/metabolism , Male , Microfilament Proteins/genetics , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Phylogeny , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Salivary Glands/microbiology , Spectrin/genetics , Spectrin/metabolism , Voltage-Dependent Anion Channels/genetics , Voltage-Dependent Anion Channels/metabolism
17.
Vet Microbiol ; 161(3-4): 339-43, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23041076

ABSTRACT

The ovine brucellosis caused by Brucella ovis has tropism for reproductive tissues but until now the mechanism of bacterial persistence is not understood. Cytokine expression profiles were studied for 8 months in rams after being experimentally infected with the rough virulent strain of B. ovis (R-B. ovis) to study the pathogenesis of B. ovis and immune mechanism possibly associated to bacteria tropism and persistence. The messenger RNA (mRNA) expression levels of interleukin-1α (IL-1α), IL-1ß, IL-6, IL-10, IL-12, interferon-γ (INF-γ) and tumour necrosis factor-α (TNF-α) cytokines were quantified by real-time quantitative RT-PCR (qRT-PCR) in reproductive tissues (epididymus, testicles, ampolae, vesicular glands and bulbourethral glands), and non-reproductive (liver, spleen and kidneys) tissues at 30, 60, 120 and 240 days post infection (dpi). During the acute phase of infection at 30 dpi, the host immune response was most notable demonstrating an up-regulation of several cytokines in reproductive tissues, including the epididymus (IL-6, IL-1ß and IL-1α), testicles (INF-γ and IL-12), bulbourethral glands (IL-6 and TNF-α) and ampolae (INF-γ, IL-10, IL-1ß and IL-1α). During the development of infection, cytokine gene expression levels decreased, providing evidence of immunosuppression and evidence of immune evasion that favoured persistence of chronic R-B. ovis infection. During the chronic phase of R-B. ovis infection (120 and 240 dpi), cytokine production was down-regulated in the epididymus (IL-1ß and IL-1α), testicles (INF-γ and IL-12), and ampolae (INF-γ, IL-10, IL-1ß and IL-1α), with the exception of the bulbourethral glands (IL-6 and TNF-α) and epididymus (IL-6); in these tissues, R-B. ovis infection resulted in up-regulation of the pro-inflammatory cytokine IL-6. Herein, we report cytokine expression profiles in tissues of rams experimentally infected with the rough strain of B. ovis, which are associated with bacterial persistence and macrophage activation.


Subject(s)
Brucella ovis/pathogenicity , Brucellosis/veterinary , Cytokines/biosynthesis , Genitalia, Male/immunology , Genitalia, Male/microbiology , Sheep Diseases/microbiology , Animals , Brucella ovis/genetics , Brucella ovis/immunology , Brucellosis/genetics , Brucellosis/immunology , Cytokines/genetics , Cytokines/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/microbiology , Macrophage Activation , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sheep , Sheep Diseases/genetics , Sheep Diseases/immunology , Up-Regulation
19.
Parasit Vectors ; 5: 181, 2012 Aug 30.
Article in English | MEDLINE | ID: mdl-22935149

ABSTRACT

BACKGROUND: Anaplasma phagocytophilum infects a wide variety of hosts and causes granulocytic anaplasmosis in humans, horses and dogs and tick-borne fever in ruminants. Infection with A. phagocytophilum results in the modification of host gene expression and immune response. The objective of this research was to characterize gene expression in pigs (Sus scrofa) naturally and experimentally infected with A. phagocytophilum trying to identify mechanisms that help to explain low infection prevalence in this species. RESULTS: For gene expression analysis in naturally infected pigs, microarray hybridization was used. The expression of differentially expressed immune response genes was analyzed by real-time RT-PCR in naturally and experimentally infected pigs. Results suggested that A. phagocytophilum infection affected cytoskeleton rearrangement and increased both innate and adaptive immune responses by up regulation of interleukin 1 receptor accessory protein-like 1 (IL1RAPL1), T-cell receptor alpha chain (TCR-alpha), thrombospondin 4 (TSP-4) and Gap junction protein alpha 1 (GJA1) genes. Higher serum levels of IL-1 beta, IL-8 and TNF-alpha in infected pigs when compared to controls supported data obtained at the mRNA level. CONCLUSIONS: These results suggested that pigs are susceptible to A. phagocytophilum but control infection, particularly through activation of innate immune responses, phagocytosis and autophagy. This fact may account for the low infection prevalence detected in pigs in some regions and thus their low or no impact as a reservoir host for this pathogen. These results advanced our understanding of the molecular mechanisms at the host-pathogen interface and suggested a role for newly reported genes in the protection of pigs against A. phagocytophilum.


Subject(s)
Anaplasma phagocytophilum/immunology , Anaplasma phagocytophilum/pathogenicity , Host-Pathogen Interactions , Sus scrofa/immunology , Sus scrofa/microbiology , Transcriptome , Animals , Autophagy , Cytokines/biosynthesis , Cytokines/metabolism , Immunity, Innate , Male , Microarray Analysis , Phagocytosis , Real-Time Polymerase Chain Reaction , Swine
20.
PLoS One ; 7(4): e35441, 2012.
Article in English | MEDLINE | ID: mdl-22530023

ABSTRACT

BACKGROUND: The γ-proteobacterium Francisella tularensis is the etiologic agent of seasonal tick-transmitted tularemia epizootics in rodents and rabbits and of incidental infections in humans. The biology of F. tularensis in its tick vectors has not been fully described, particularly with respect to its quanta and duration of colonization, tissue dissemination, and transovarial transmission. A systematic study of the colonization of Dermacentor variabilis by the F. tularensis subsp. holarctica live vaccine strain (LVS) was undertaken to better understand whether D. variabilis may serve as an inter-epizootic reservoir for F. tularensis. METHODOLOGY/PRINCIPAL FINDINGS: Colony-reared larva, nymph, and adult D. variabilis were artificially fed LVS via glass capillary tubes fitted over the tick mouthparts, and the level of colonization determined by microbial culture. Larvae and nymphs were initially colonized with 8.8 ± 0.8 × 10(1) and 1.1 ± 0.03 × 10(3) CFU/tick, respectively. Post-molting, a significant increase in colonization of both molted nymphs and adults occurred, and LVS persisted in 42% of molted adult ticks at 126 days post-capillary tube feeding. In adult ticks, LVS initially colonized the gut, disseminated to hemolymph and salivary glands by 21 days, and persisted up to 165 days. LVS was detected in the salivary secretions of adult ticks after four days post intra-hemocoelic inoculation, and LVS recovered from salivary gland was infectious to mice with an infectious dose 50% of 3 CFU. LVS in gravid female ticks colonized via the intra-hemocoelic route disseminated to the ovaries and then to the oocytes, but the pathogen was not recovered from the subsequently-hatched larvae. CONCLUSIONS/SIGNIFICANCE: This study demonstrates that D. variabilis can be efficiently colonized with F. tularensis using artificial methods. The persistence of F. tularensis in D. variabilis suggests that this tick species may be involved in the maintenance of enzootic foci of tularemia in the central United States.


Subject(s)
Arthropod Vectors/microbiology , Dermacentor/microbiology , Francisella tularensis/physiology , Tularemia/transmission , Animals , Arthropod Vectors/growth & development , Dermacentor/growth & development , Female , Larva/microbiology , Male , Mice , Mice, Inbred BALB C , Nymph/microbiology , Oocytes/microbiology , Salivary Glands/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...