Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
6.
Ticks Tick Borne Dis ; 3(3): 147-53, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22534515

ABSTRACT

Anaplasma phagocytophilum, first identified as a pathogen of ruminants in Europe, has more recently been recognized as an emerging tick-borne pathogen of humans in the U.S. and Europe. A. phagocytophilum is transmitted by Ixodes spp., but the tick developmental cycle and pathogen/vector interactions have not been fully described. In this research, we report on the experimental infection of sheep with the human NY-18 isolate of A. phagocytophilum which then served as a host for infection of I. scapularis nymphs and adults. A. phagocytophilum was propagated in the human promyelocytic cell line, HL-60, and the infected cell cultures were then used to infect sheep by intravenous inoculation. Infections in sheep were confirmed by PCR and an Anaplasma-competitive ELISA. Clinical signs were not apparent in any of the infected sheep, and only limited hematologic and mild serum biochemical abnormalities were identified. While A. phagocytophilum morulae were rarely seen in neutrophils, blood film evaluation revealed prominent large granular lymphocytes, occasional plasma cells, and rare macrophages. Upon necropsy, gross lesions were restricted to the lymphoid system. Mild splenomegaly and lymphadenomegaly with microscopic evidence of lymphoid hyperplasia was observed in all infected sheep. Female I. scapularis that were allowed to feed and acquire infection on each of the 3 experimentally infected sheep became infected with A. phagocytophilum as determined by PCR of guts (80-87%) and salivary glands (67-100%). Female I. scapularis that acquired infection as nymphs on an experimentally infected sheep transmitted A. phagocytophilum to a susceptible sheep, thus confirming transstadial transmission. Sheep proved to be a good host for the production of I. scapularis infected with this human isolate of A. phagocytophilum, which can be used as a model for future studies of the tick/pathogen interface.


Subject(s)
Anaplasma phagocytophilum/physiology , Arachnid Vectors/microbiology , Ehrlichiosis/microbiology , Ixodes/microbiology , Tick Infestations/parasitology , Anaplasma phagocytophilum/genetics , Anaplasma phagocytophilum/immunology , Animals , Antigens, Bacterial/immunology , Arachnid Vectors/virology , Cell Line , DNA, Bacterial/genetics , Ehrlichiosis/complications , Enzyme-Linked Immunosorbent Assay , Female , Gastrointestinal Tract/microbiology , Humans , Ixodes/virology , Male , Models, Animal , Neutrophils/microbiology , Nymph , Polymerase Chain Reaction , Salivary Glands/microbiology , Sheep , Tick Infestations/complications
7.
Vet Parasitol ; 181(1): 17-22, 2011 Sep 08.
Article in English | MEDLINE | ID: mdl-21561715

ABSTRACT

Diseases caused by arthropod-borne pathogens greatly impact on human and animal health. Recent research has provided evidence that tick protective antigens can be used for development of vaccines with the dual target of controlling arthropod infestations and reducing their vector capacity for pathogens. As reviewed herein, protective antigens such as subolesin/akirin, which are highly conserved across vector species, show promise for use in development of a universal vaccine for the control of arthropod infestations and the reduction of pathogen transmission. However, further research is needed in critical areas towards achieving this goal.


Subject(s)
Animal Diseases/prevention & control , Antigens/immunology , Arthropod Proteins/immunology , Tick Infestations/veterinary , Ticks/immunology , Vaccines , Animal Diseases/parasitology , Animals , Arachnid Vectors/immunology , Humans , Tick Infestations/prevention & control
8.
Vector Borne Zoonotic Dis ; 11(7): 917-22, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21254834

ABSTRACT

Q fever, a zoonotic disease, is caused by a gram-negative intracellular bacterium, Coxiella burnetii. Although normally transmitted during exposure to infectious aerosols, C. burnetii is also found in arthropod vectors. In the environment, ticks are thought to play a crucial role in bacterial maintenance and transmission by infecting various mammalian species. However, the nature of the pathogen-tick relationship is not well defined. To determine C. burnetii's interactions with a cultured tick cell line, we introduced purified C. burnetii NMII into Ixodes scapularis-derived IDE8 cells and assayed for bacterial presence, replication, gene expression, and subsequent infectivity for mammalian cells. Tick cells were harvested at 24 h, 72 h, 7 days, and 11 days postinfection (PI). C. burnetii uptake and subsequent replication was demonstrated by indirect immunofluorescence assay, electron microscopy, and real-time polymerase chain reaction (PCR). When a genome equivalent multiplicity of infection of 30 was used, 30%-40% of exposed cells were seen to have small, rounded, vacuoles at 72 h PI, whereas at 7 and 11 days PI, 60%-70% of cells contained enlarged vacuoles harboring large numbers of bacteria. Quantitative PCR analysis of total genomic DNA confirmed that C. burnetii genome numbers increased significantly from 24 h to 11 days PI. Expression of C. burnetii type four secretion system homologs at 7 days PI was demonstrated by reverse transcriptase PCR. Finally, indirect immunofluorescence assay demonstrated that C. burnetii propagated within IDE8 cells were infectious for mammalian cells. These studies demonstrate the utility of cultured tick cell lines as a model to investigate C. burnetii's molecular interactions with its arthropod vectors.


Subject(s)
Coxiella burnetii/pathogenicity , Ixodes/microbiology , Animals , Arachnid Vectors/microbiology , Chlorocebus aethiops , Coxiella burnetii/genetics , Coxiella burnetii/growth & development , Fluorescent Antibody Technique, Indirect , Polymerase Chain Reaction , Vero Cells
9.
BMC Immunol ; 11: 7, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20170494

ABSTRACT

BACKGROUND: Ticks (Acari: Ixodidae) are vectors of pathogens worldwide that cause diseases in humans and animals. Ticks and pathogens have co-evolved molecular mechanisms that contribute to their mutual development and survival. Subolesin was discovered as a tick protective antigen and was subsequently shown to be similar in structure and function to akirins, an evolutionarily conserved group of proteins in insects and vertebrates that controls NF-kB-dependent and independent expression of innate immune response genes. The objective of this study was to investigate subolesin expression in several tick species infected with a variety of pathogens and to determine the effect of subolesin gene knockdown on pathogen infection. In the first experiment, subolesin expression was characterized in ticks experimentally infected with the cattle pathogen, Anaplasma marginale. Subolesin expression was then characterized in questing or feeding adult ticks confirmed to be infected with Anaplasma, Ehrlichia, Rickettsia, Babesia or Theileria spp. Finally, the effect of subolesin knockdown by RNA interference (RNAi) on tick infection was analyzed in Dermacentor variabilis males exposed to various pathogens by capillary feeding (CF). RESULTS: Subolesin expression increased with pathogen infection in the salivary glands but not in the guts of tick vector species infected with A. marginale. When analyzed in whole ticks, subolesin expression varied between tick species and in response to different pathogens. As reported previously, subolesin knockdown in D. variabilis infected with A. marginale and other tick-borne pathogens resulted in lower infection levels, while infection with Francisella tularensis increased in ticks after RNAi. When non-tick-borne pathogens were fed to ticks by CF, subolesin RNAi did not affect or resulted in lower infection levels in ticks. However, subolesin expression was upregulated in D. variabilis exposed to Escherichia coli, suggesting that although this pathogen may induce subolesin expression in ticks, silencing of this molecule reduced bacterial multiplication by a presently unknown mechanism. CONCLUSIONS: Subolesin expression in infected ticks suggested that subolesin may be functionally important for tick innate immunity to pathogens, as has been reported for the akirins. However, subolesin expression and consequently subolesin-mediated innate immunity varied with the pathogen and tick tissue. Subolesin may plays a role in tick innate immunity in the salivary glands by limiting pathogen infection levels, but activates innate immunity only for some pathogen in the guts and other tissues. In addition, these results provided additional support for the role of subolesin in other molecular pathways including those required for tissue development and function and for pathogen infection and multiplication in ticks. Consequently, RNAi experiments demonstrated that subolesin knockdown in ticks may affect pathogen infection directly by reducing tick innate immunity that results in higher infection levels and indirectly by affecting tissue structure and function and the expression of genes that interfere with pathogen infection and multiplication. The impact of the direct or indirect effects of subolesin knockdown on pathogen infection may depend on several factors including specific tick-pathogen molecular interactions, pathogen life cycle in the tick and unknown mechanisms affected by subolesin function in the control of global gene expression in ticks.


Subject(s)
Antigens/metabolism , Bacteria/immunology , Bacterial Infections/immunology , Intestinal Mucosa/metabolism , Salivary Glands/metabolism , Ticks/metabolism , Animals , Antigens/genetics , Antigens/immunology , Arthropod Proteins , Bacteria/pathogenicity , Dermacentor/immunology , Drosophila Proteins/genetics , Evolution, Molecular , Host-Pathogen Interactions , Immunity, Innate , Insect Vectors , Intestines/immunology , Intestines/pathology , Life Cycle Stages , Nuclear Proteins , RNA, Small Interfering/genetics , Salivary Glands/immunology , Salivary Glands/pathology , Ticks/immunology , Ticks/microbiology , Virulence
10.
Int J Proteomics ; 2010: 657261, 2010.
Article in English | MEDLINE | ID: mdl-22084679

ABSTRACT

Ticks are ectoparasites of animals and humans that serve as vectors of Anaplasma and other pathogens that affect humans and animals worldwide. Ticks and the pathogens that they transmit have coevolved molecular interactions involving genetic traits of both the tick and the pathogen that mediate their development and survival. In this paper, the expression of heat shock proteins (HSPs) and other stress response proteins (SRPs) was characterized in ticks and cultured tick cells by proteomics and transcriptomics analyses in response to Anaplasma spp. infection and heat shock. The results of these studies demonstrated that the stress response was activated in ticks and cultured tick cells after Anaplasma spp. infection and heat shock. However, in the natural vector-pathogen relationship, HSPs and other SRPs were not strongly activated, which likely resulted from tick-pathogen coevolution. These results also demonstrated pathogen- and tick-specific differences in the expression of HSPs and other SRPs in ticks and cultured tick cells infected with Anaplasma spp. and suggested the existence of post-transcriptional mechanisms induced by Anaplasma spp. to control tick response to infection. These results illustrated the complexity of the stress response in ticks and suggested a function for the HSPs and other SRPs during Anaplasma spp. infection.

11.
Vet Parasitol ; 167(2-4): 175-86, 2010 Feb 10.
Article in English | MEDLINE | ID: mdl-19819630

ABSTRACT

The genus Anaplasma (Rickettsiales: Anaplasmataceae) includes several tick-transmitted pathogens that impact veterinary and human health. Tick-borne pathogens cycle between tick vectors and vertebrate hosts and their interaction is mediated by molecular mechanisms at the tick-pathogen interface. These mechanisms have evolved characteristics that involve traits from both the tick vector and the pathogen to insure their mutual survival. Herein, we review the information obtained from functional genomics and genetic studies to characterize the tick-Anaplasma interface and evolution of A. marginale and A. phagocytophilum. Anaplasma and tick genes and proteins involved in tick-pathogen interactions were characterized. The results of these studies demonstrated that common and Anaplasma species-specific molecular mechanism occur by which pathogen and tick cell gene expression mediates or limits Anaplasma developmental cycle and trafficking through ticks. These results have advanced our understanding of the biology of tick-Anaplasma interactions and have opened new avenues for the development of improved methods for the control of tick infestations and the transmission of tick-borne pathogens.


Subject(s)
Anaplasma/physiology , Genomics , Ticks/microbiology , Animals , Biological Evolution , Host-Pathogen Interactions/genetics
12.
Vaccine ; 28(7): 1786-95, 2010 Feb 17.
Article in English | MEDLINE | ID: mdl-20018267

ABSTRACT

The lone star tick, Amblyomma americanum, vectors pathogens of emerging diseases of humans and animals in the United States. Currently, measures are not available for effective control of A. americanum infestations. Development of vaccines directed against tick proteins may reduce tick infestations and the transmission of tick-borne pathogens. However, the limiting step in tick vaccine development has been the identification of tick protective antigens. Herein, we report the application of RNA interference (RNAi) for screening an A. americanum cDNA library for discovery of tick protective antigens that reduce tick survival and weights after feeding. Four cDNA clones, encoding for putative threonyl-tRNA synthetase (2C9), 60S ribosomal proteins L13a (2D10) and L13e (2B7), and interphase cytoplasm foci protein 45 (2G7), were selected for vaccine studies in cattle, along with subolesin, a tick protective protein identified previously. In vaccinated cattle, an overall efficacy (E)>30% was obtained when considering the vaccine effect on both nymphs and adults, but only 2D10, 2G7 and subolesin affected both tick stages. The highest efficacy of control for adult ticks (E>55%) was obtained in cattle vaccinated with recombinant 2G7 or subolesin. These collective results demonstrated the feasibility of developing vaccines for the control of lone star tick infestations. The use of RNAi for identification of tick protective antigens proved to be a rapid and cost-effective tool for discovery of candidate vaccine antigens, and this approach could likely be applied to other parasites of veterinary and medical importance.


Subject(s)
Antigens/genetics , Ixodidae/genetics , RNA Interference , Tick Infestations/prevention & control , Animals , Antibody Formation , Antigens/immunology , Cattle , Female , Gene Expression Profiling , Gene Library , Ixodidae/immunology , Male , Polymorphism, Genetic , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Proteins/genetics , Ribosomal Proteins/immunology , Sequence Analysis, DNA , Threonine-tRNA Ligase/genetics , Threonine-tRNA Ligase/immunology , Tick Infestations/immunology , Tick Infestations/veterinary , Vaccines/immunology
13.
Vet Parasitol ; 167(2-4): 95-107, 2010 Feb 10.
Article in English | MEDLINE | ID: mdl-19811876

ABSTRACT

The intracellular pathogen Anaplasma marginale (Rickettsiales: Anaplasmataceae), described by Sir Arnold Theiler in 1910, is endemic worldwide in tropical and subtropical areas. Infection of cattle with A. marginale causes bovine anaplasmosis, a mild to severe hemolytic disease that results in considerable economic loss to both dairy and beef industries. Transmission of A. marginale to cattle occurs biologically by ticks and mechanically by biting flies and by blood-contaminated fomites. Both male ticks and cattle hosts become persistently infected with A. marginale and serve as reservoirs of infection. While erythrocytes are the major site of infection in cattle, A. marginale undergoes a complex developmental cycle in ticks that begins by infection of gut cells, and transmission to susceptible hosts occurs from salivary glands during feeding. Major surface proteins (MSPs) play a crucial role in the interaction of A. marginale with host cells, and include adhesion proteins and MSPs from multigene families that undergo antigenic change and selection in cattle, thus contributing to maintenance of persistent infections. Many geographic strains of A. marginale have been identified worldwide, which vary in genotype, antigenic composition, morphology and infectivity for ticks. Isolates of A. marginale may be maintained by independent transmission events and a mechanism of infection/exclusion in cattle and ticks. The increasing numbers of A. marginale genotypes identified in some geographic regions most likely resulted from intensive cattle movement. However, concurrent A. marginale strain infections in cattle was reported, but these strains were more distantly related. Phylogenetic studies of selected geographic isolates of A. marginale, using msp4 and msp1alpha, provided information about the biogeography and evolution of A. marginale, and msp1alpha genotypes appear to have evolved under positive selection pressure. Live and killed vaccines have been used for control of anaplasmosis and both types of vaccines have advantages and disadvantages. Vaccines have effectively prevented clinical anaplasmosis in cattle but have failed to block A. marginale infection. Vaccines are needed that can prevent clinical disease and, simultaneously, prevent infection in cattle and ticks, thus eliminating these hosts as reservoirs of infection. Advances in genomics, proteomics, immunology and biochemical and molecular technologies during the last decade have been applied to research on A. marginale and related organisms, and the recent development of a cell culture system for A. marginale has provided a format for studying the pathogen/tick interface. Recent advancements and new research methodologies should provide additional opportunities for development of new strategies for control and prevention of bovine anaplasmosis.


Subject(s)
Anaplasma marginale/physiology , Anaplasmosis/epidemiology , Anaplasmosis/microbiology , Animals , Cattle , Male , Ticks/microbiology
14.
BMC Dev Biol ; 9: 42, 2009 Jul 16.
Article in English | MEDLINE | ID: mdl-19607704

ABSTRACT

BACKGROUND: The cattle pathogen, Anaplasma marginale, undergoes a developmental cycle in ticks that begins in gut cells. Transmission to cattle occurs from salivary glands during a second tick feeding. At each site of development two forms of A. marginale (reticulated and dense) occur within a parasitophorous vacuole in the host cell cytoplasm. However, the role of tick genes in pathogen development is unknown. Four genes, found in previous studies to be differentially expressed in Dermacentor variabilis ticks in response to infection with A. marginale, were silenced by RNA interference (RNAi) to determine the effect of silencing on the A. marginale developmental cycle. These four genes encoded for putative glutathione S-transferase (GST), salivary selenoprotein M (SelM), H+ transporting lysosomal vacuolar proton pump (vATPase) and subolesin. RESULTS: The impact of gene knockdown on A. marginale tick infections, both after acquiring infection and after a second transmission feeding, was determined and studied by light microscopy. Silencing of these genes had a different impact on A. marginale development in different tick tissues by affecting infection levels, the densities of colonies containing reticulated or dense forms and tissue morphology. Salivary gland infections were not seen in any of the gene-silenced ticks, raising the question of whether these ticks were able to transmit the pathogen. CONCLUSION: The results of this RNAi and light microscopic analyses of tick tissues infected with A. marginale after the silencing of genes functionally important for pathogen development suggest a role for these molecules during pathogen life cycle in ticks.


Subject(s)
Anaplasma marginale/growth & development , Anaplasma marginale/genetics , Arthropod Vectors/parasitology , Dermacentor/parasitology , Gene Silencing , Animals , Cattle , Glutathione Transferase/genetics , Host-Parasite Interactions , Male , Microscopy , RNA Interference , Selenoproteins/genetics , Vacuolar Proton-Translocating ATPases/genetics
15.
Comp Funct Genomics ; : 705034, 2009.
Article in English | MEDLINE | ID: mdl-19636428

ABSTRACT

The genus Anaplasma (Rickettsiales: Anaplasmataceae) includes obligate tick-transmitted intracellular organisms, Anaplasma phagocytophilum and Anaplasma marginale that multiply in both vertebrate and tick host cells. Recently, we showed that A. marginale affects the expression of tick genes that are involved in tick survival and pathogen infection and multiplication. However, the gene expression profile in A. phagocytophilum-infected tick cells is currently poorly characterized. The objectives of this study were to characterize tick gene expression profile in Ixodes scapularis ticks and cultured ISE6 cells in response to infection with A. phagocypthilum and to compare tick gene expression responses in A. phagocytophilum- and A. marginale-infected tick cells by microarray and real-time RT-PCR analyses. The results of these studies demonstrated modulation of tick gene expression by A. phagocytophilum and provided evidence of different gene expression responses in tick cells infected with A. phagocytophilum and A. marginale. These differences in Anaplasma-tick interactions may reflect differences in pathogen life cycle in the tick cells.

16.
Vet Parasitol ; 161(1-2): 110-5, 2009 Apr 06.
Article in English | MEDLINE | ID: mdl-19168288

ABSTRACT

Cytauxzoon felis was transmitted to a domestic cat by Amblyomma americanum. The infection was produced by the bite of A. americanum adults that were acquisition fed as nymphs on a domestic cat that naturally survived infection of C. felis. Fever, inappetence, depression, and lethargy were first noted 11 days post-infestation (dpi). Pale mucus membranes, splenomegaly, icterus, and dyspnea were also observed during the course of the disease. The body temperature of the experimentally infected C. felis cat was subnormal from 16 dpi until 24 dpi when it returned to within normal limits. All clinical signs of cytauxzoonsis began to resolve by 23 dpi when the cat became subclinically infected with C. felis. The cat developed a marked, regenerative anemia beginning by 13 dpi and reached a nadir at 20 dpi before recovering. A moderate neutrophilia and marked lymphocytosis also developed between 18 and 26 dpi. Schizonts of C. felis were observed in spleen aspirates of the infected cat at 15 dpi. DNA of C. felis was amplified by real-time PCR starting 17 dpi and piroplasms of C. felis were first noted by light microscopy 18 dpi. Dermacentor variabilis, Ixodes scapularis, and Rhipicephalus sanguineus were also tested in a similar manner at the same time but did not transmit C. felis. Prior to the present study, only D. variabilis had been shown experimentally to transmit infection of C. felis. This is the first report of C. felis being transmitted by A. americanum. The transmission of C. felis infection from one domestic cat to another indicates that domestic cats subclinically infected with C. felis may be a reservoir of infection for naive domestic cats.


Subject(s)
Arachnid Vectors , Cat Diseases/transmission , Ixodidae/parasitology , Protozoan Infections, Animal/transmission , Animals , Cat Diseases/parasitology , Cats , Female , Protozoan Infections, Animal/parasitology
17.
Vector Borne Zoonotic Dis ; 9(5): 565-8, 2009 Oct.
Article in English | MEDLINE | ID: mdl-18973438

ABSTRACT

Four white-tailed deer were inoculated with either the Ap-V1 or NY-18 strain of Anaplasma phagocytophilum. Ixodes scapularis nymphs were then allowed to acquistion feed on the inoculated deer and molt to adults. Only an Ap-V1 infected deer was infected persistently and able to infect nymphal Ixodes scapularis. Molted adult ticks maintained Ap-V1 infection as demonstrated by PCR and microscopy. We report, for the first time, a morphologic description of A. phagocytophilum in I. scapularis.


Subject(s)
Anaplasma phagocytophilum/classification , Anaplasma phagocytophilum/physiology , Anaplasmosis/microbiology , Deer/microbiology , Ixodes/microbiology , Anaplasmosis/transmission , Animals , Nymph/microbiology
18.
BMC Genomics ; 9: 372, 2008 Aug 02.
Article in English | MEDLINE | ID: mdl-18673577

ABSTRACT

BACKGROUND: Subolesin is an evolutionary conserved protein that was discovered recently in Ixodes scapularis as a tick protective antigen and has a role in tick blood digestion, reproduction and development. In other organisms, subolesin orthologs may be involved in the control of developmental processes. Because of the profound effect of subolesin knockdown in ticks and other organisms, we hypothesized that subolesin plays a role in gene expression, and therefore affects multiple cellular processes. The objective of this study was to provide evidence for the role of subolesin in gene expression. RESULTS: Two subolesin-interacting proteins were identified and characterized by yeast two-hybrid screen, co-affinity purification and RNA interference (RNAi). The effect of subolesin knockdown on the tick gene expression pattern was characterized by microarray analysis and demonstrated that subolesin RNAi affects the expression of genes involved in multiple cellular pathways. The analysis of subolesin and interacting protein sequences identified regulatory motifs and predicted the presence of conserved protein kinase C (PKC) phosphorylation sites. CONCLUSION: Collectively, these results provide evidence that subolesin plays a role in gene expression in ticks.


Subject(s)
Gene Expression Regulation , Proteins/metabolism , Ticks/genetics , Animals , Base Sequence , Feeding Behavior , Female , Gene Expression Profiling , Molecular Sequence Data , Oviposition/genetics , Ovum/growth & development , Protein Processing, Post-Translational , Proteins/genetics , Ticks/cytology , Ticks/physiology , Two-Hybrid System Techniques
19.
Front Biosci ; 13: 6947-56, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18508707

ABSTRACT

Ticks are ectoparasites of wild and domestic animals and humans that most notably impact global health by transmitting disease-causing pathogens. While information on the molecular interactions between ticks and pathogens that facilitate pathogen infection, development and transmission is limited, a comprehensive understanding of the tick-pathogen interface would be fundamental toward development of new and novel measures for control of both tick infestations and tick-borne pathogens. Recently, vaccine studies using key tick antigens and characterization of tick gene function by RNA interference (RNAi) have provided new information on genes that impact the tick-pathogen interface. In this review we summarize current research and prospects of tick vaccines and genetic manipulation of ticks targeted to the tick-pathogen interface. The knowledge gained from these collective studies will be fundamental toward understanding of tick-pathogen interactions and for formulation of control methods targeted at both ticks and tick-borne pathogens. Use of these molecular approaches will likely contribute to control measures that will notably reduce tick populations and tick-borne diseases in the future.


Subject(s)
Tick Infestations/immunology , Tick-Borne Diseases/immunology , Ticks/immunology , Vaccines/therapeutic use , Animals , Borrelia burgdorferi/immunology , Humans , Lyme Disease/immunology , Lyme Disease/prevention & control , RNA Interference/immunology , Tick Infestations/prevention & control , Tick-Borne Diseases/prevention & control , Ticks/genetics
20.
Front Biosci ; 13: 7032-45, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18508714

ABSTRACT

The genus Anaplasma includes a diverse group of tick-borne pathogens found exclusively within membrane-bound vacuoles in host cells. While A. marginale, A. centrale and A. ovis, vectored by Dermacentor and Rhipicephalus ticks, are host-specific for ruminants, A. phagocytophilum, vectored by Ixodes spp., infects a wide range of hosts. In ticks Anaplasma undergoes a developmental cycle that is coordinated with the tick feeding cycle. Although research at the tick/Anaplasma interface is in its infancy, recent studies have provided evidence that Anaplasma infection and transmission is mediated by a molecular mechanism involving both tick cell and pathogen genes. Application of a growing array of molecular approaches, such as RNA interference, genomics and proteomics, are rapidly expanding our knowledge of the tick/pathogen interface. Targeting key tick cell molecules required for pathogen development in vaccine strategies may compromise the vector capacity of ticks for Anaplasma, thus reducing transmission and infection of vertebrates. Collectively, this information will likely lead to the development of dual target vaccines designed to protect vertebrates against tick infestations and prevent the transmission of pathogens.


Subject(s)
Anaplasma/physiology , Ticks/microbiology , Anaplasma/genetics , Animals , Genomics , Ixodes , Molecular Biology/methods , Proteomics , RNA Interference , Tick Infestations/immunology , Tick Infestations/physiopathology , Tick Infestations/prevention & control , Ticks/genetics , Vaccines
SELECTION OF CITATIONS
SEARCH DETAIL
...