Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Infect Immun ; 92(6): e0054023, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38727242

RESUMO

Anaplasma marginale is an obligate, intracellular, tick-borne bacterial pathogen that causes bovine anaplasmosis, an often severe, production-limiting disease of cattle found worldwide. Methods to control this disease are lacking, in large part due to major knowledge gaps in our understanding of the molecular underpinnings of basic host-pathogen interactions. For example, the surface proteins that serve as adhesins and, thus, likely play a role in pathogen entry into tick cells are largely unknown. To address this knowledge gap, we developed a phage display library and screened 66 A. marginale proteins for their ability to adhere to Dermacentor andersoni tick cells. From this screen, 17 candidate adhesins were identified, including OmpA and multiple members of the Msp1 family, including Msp1b, Mlp3, and Mlp4. We then measured the transcript of ompA and all members of the msp1 gene family through time, and determined that msp1b, mlp2, and mlp4 have increased transcript during tick cell infection, suggesting a possible role in host cell binding or entry. Finally, Msp1a, Msp1b, Mlp3, and OmpA were expressed as recombinant protein. When added to cultured tick cells prior to A. marginale infection, all proteins except the C-terminus of Msp1a reduced A. marginale entry by 2.2- to 4.7-fold. Except OmpA, these adhesins lack orthologs in related pathogens of humans and animals, including Anaplasma phagocytophilum and the Ehrlichia spp., thus limiting their utility in a universal tick transmission-blocking vaccine. However, this work greatly advances efforts toward developing methods to control bovine anaplasmosis and, thus, may help improve global food security.


Assuntos
Adesinas Bacterianas , Anaplasma marginale , Dermacentor , Animais , Anaplasma marginale/genética , Adesinas Bacterianas/metabolismo , Adesinas Bacterianas/genética , Dermacentor/microbiologia , Bovinos , Aderência Bacteriana/fisiologia , Anaplasmose/microbiologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas da Membrana Bacteriana Externa/genética , Técnicas de Visualização da Superfície Celular , Interações Hospedeiro-Patógeno , Doenças dos Bovinos/microbiologia
2.
PLoS Pathog ; 18(5): e1010540, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35576225

RESUMO

Tick-borne Anaplasma species are obligate, intracellular, bacterial pathogens that cause important diseases globally in people, agricultural animals, and dogs. Targeted mutagenesis methods are yet to be developed to define genes essential for these pathogens. In addition, vaccines conferring protection against diseases caused by Anaplasma species are not available. Here, we describe a targeted mutagenesis method for deletion of the phage head-to-tail connector protein (phtcp) gene in Anaplasma marginale. The mutant did not cause disease and exhibited attenuated growth in its natural host (cattle). We then assessed its ability to confer protection against wild-type A. marginale infection challenge. Additionally, we compared vaccine protection with the mutant to that of whole cell A. marginale inactivated antigens as a vaccine (WCAV) candidate. Upon infection challenge, non-vaccinated control cattle developed severe disease, with an average 57% drop in packed cell volume (PCV) between days 26-31 post infection, an 11% peak in erythrocytic infection, and apparent anisocytosis. Conversely, following challenge, all animals receiving the live mutant did not develop clinical signs or anemia, or erythrocyte infection. In contrast, the WCAV vaccinees developed similar disease as the non-vaccinees following A. marginale infection, though the peak erythrocyte infection reduced to 6% and the PCV dropped 43%. This is the first study describing targeted mutagenesis and its application in determining in vivo virulence and vaccine development for an Anaplasma species pathogen. This study will pave the way for similar research in related Anaplasma pathogens impacting multiple hosts.


Assuntos
Anaplasma marginale , Anaplasmose , Doenças dos Bovinos , Anaplasma , Anaplasma marginale/genética , Anaplasmose/genética , Anaplasmose/prevenção & controle , Animais , Bovinos , Doenças dos Bovinos/microbiologia , Cães , Humanos , Mutagênese , Desenvolvimento de Vacinas , Virulência
3.
Infect Immun ; 91(4): e0050122, 2023 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-36877065

RESUMO

Many vector-borne pathogens, including Anaplasma spp., Borrelia spp., Trypanosoma spp., and Plasmodium spp., establish persistent infection in the mammalian host by using antigenic variation. These pathogens are also able to establish strain superinfection, defined as infection of an infected host with additional strains of the same pathogen despite an adaptive immune response. The ability to establish superinfection results in a population of susceptible hosts even with high pathogen prevalence. It is likely that antigenic variation, responsible for persistent infection, also plays a role in the establishment of superinfection. Anaplasma marginale, an antigenically variable, obligate intracellular, tickborne bacterial pathogen of cattle, is well suited for the study of the role of antigenically variant surface proteins in the establishment of superinfection. Anaplasma marginale establishes persistent infection by variation in major surface protein 2 (msp2), which is encoded by approximately six donor alleles that recombine into a single expression site to produce immune escape variants. Nearly all cattle in regions of high prevalence are superinfected. By tracking the acquisition of strains in calves through time, the complement of donor alleles, and how those donor alleles are expressed, we determined that simple variants derived from a single donor allele, rather than multiple donor alleles, were predominant. Additionally, superinfection is associated with the introduction of new donor alleles, but these new donor alleles are not predominantly used to establish superinfection. These findings highlight the potential for competition among multiple strains of a pathogen for resources within the host and the balance between pathogen fitness and antigenic variation.


Assuntos
Anaplasma marginale , Anaplasmose , Doenças dos Bovinos , Superinfecção , Carrapatos , Bovinos , Animais , Anaplasma marginale/genética , Anaplasmose/microbiologia , Superinfecção/microbiologia , Gana/epidemiologia , Infecção Persistente , Antígenos de Bactérias , Mamíferos , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Doenças dos Bovinos/microbiologia
4.
Int J Mol Sci ; 23(7)2022 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-35409307

RESUMO

Anaplasma spp. are obligate intracellular, tick-borne, bacterial pathogens that cause bovine and human anaplasmosis. We lack tools to prevent these diseases in part due to major knowledge gaps in our fundamental understanding of the tick-pathogen interface, including the requirement for and molecules involved in iron transport during tick colonization. We determine that iron is required for the pathogen Anaplasma marginale, which causes bovine anaplasmosis, to replicate in Dermacentor andersoni tick cells. Using bioinformatics and protein modeling, we identified three orthologs of the Gram-negative siderophore-independent iron uptake system, FbpABC. Am069, the A. marginale ortholog of FbpA, lacks predicted iron-binding residues according to the NCBI conserved domain database. However, according to protein modeling, the best structural orthologs of Am069 are iron transport proteins from Cyanobacteria and Campylobacterjejuni. We then determined that all three A. marginale genes are modestly differentially expressed in response to altered host cell iron levels, despite the lack of a Ferric uptake regulator or operon structure. This work is foundational for building a mechanistic understanding of iron uptake, which could lead to interventions to prevent bovine and human anaplasmosis.


Assuntos
Anaplasma marginale , Anaplasmose , Dermacentor , Anaplasma , Anaplasma marginale/genética , Anaplasmose/microbiologia , Animais , Bovinos , Dermacentor/genética , Dermacentor/microbiologia , Humanos , Ferro
5.
Infect Immun ; 89(11): e0016621, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34338549

RESUMO

Vector-borne pathogens commonly establish multistrain infections, also called complex infections. How complex infections are established, either before or after the development of an adaptive immune response, termed coinfection or superinfection, respectively, has broad implications for the maintenance of genetic diversity, pathogen phenotype, epidemiology, and disease control strategies. Anaplasma marginale, a genetically diverse, obligate, intracellular, tick-borne bacterial pathogen of cattle, commonly establishes complex infections, particularly in regions with high transmission rates. Both coinfection and superinfection can be established experimentally; however, it is unknown how complex infections develop in a natural transmission setting. To address this question, we introduced naive animals into a herd in southern Ghana with a high infection prevalence and high transmission pressure and tracked the strain acquisition of A. marginale through time using multilocus sequence typing. As expected, the genetic diversity among strains was high, and 97% of animals in the herd harbored multiple strains. All the introduced naive animals became infected, and three to four strains were typically detected in an individual animal prior to seroconversion, while one to two new strains were detected in an individual animal following seroconversion. On average, the number of strains acquired via superinfection was 16% lower than the number acquired via coinfection. Thus, while complex infections develop via both coinfection and superinfection, coinfection predominates in this setting. These findings have broad implications for the development of control strategies in high-transmission settings.


Assuntos
Anaplasma marginale/genética , Anaplasmose/microbiologia , Coinfecção/microbiologia , Superinfecção/microbiologia , Alelos , Anaplasmose/etiologia , Anaplasmose/transmissão , Animais , Bovinos , Coinfecção/etiologia , Superinfecção/etiologia
6.
PLoS Pathog ; 10(10): e1004499, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25392914

RESUMO

The genetic diversity of pathogens, and interactions between genotypes, can strongly influence pathogen phenotypes such as transmissibility and virulence. For vector-borne pathogens, both mammalian hosts and arthropod vectors may limit pathogen genotypic diversity (number of unique genotypes circulating in an area) by preventing infection or transmission of particular genotypes. Mammalian hosts often act as "ecological filters" for pathogen diversity, where novel variants are frequently eliminated because of stochastic events or fitness costs. However, whether vectors can serve a similar role in limiting pathogen diversity is less clear. Here we show using Francisella novicida and a natural tick vector of Francisella spp. (Dermacentor andersoni), that the tick vector acted as a stronger ecological filter for pathogen diversity compared to the mammalian host. When both mice and ticks were exposed to mixtures of F. novicida genotypes, significantly fewer genotypes co-colonized ticks compared to mice. In both ticks and mice, increased genotypic diversity negatively affected the recovery of available genotypes. Competition among genotypes contributed to the reduction of diversity during infection of the tick midgut, as genotypes not recovered from tick midguts during mixed genotype infections were recovered from tick midguts during individual genotype infection. Mediated by stochastic and selective forces, pathogen genotype diversity was markedly reduced in the tick. We incorporated our experimental results into a model to demonstrate how vector population dynamics, especially vector-to-host ratio, strongly affected pathogen genotypic diversity in a population over time. Understanding pathogen genotypic population dynamics will aid in identification of the variables that most strongly affect pathogen transmission and disease ecology.


Assuntos
Vetores Aracnídeos/microbiologia , Dermacentor/microbiologia , Francisella/genética , Variação Genética , Animais , Trato Gastrointestinal/microbiologia , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Fenótipo , Coelhos , Virulência
7.
Appl Environ Microbiol ; 82(15): 4715-4731, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27235428

RESUMO

UNLABELLED: Tick-borne transmission of bacterial pathogens in the order Rickettsiales is responsible for diverse infectious diseases, many of them severe, in humans and animals. Transmission dynamics differ among these pathogens and are reflected in the pathogen-vector interaction. Anaplasma marginale has been shown to establish and maintain infectivity within Dermacentor spp. for weeks to months while escaping the complex network of vacuolar peptidases that are responsible for digestion of the tick blood meal. How this prolonged maintenance of infectivity in a potentially hostile environment is achieved has been unknown. Using the natural vector Dermacentor andersoni, we demonstrated that A. marginale-infected tick vacuoles (AmVs) concurrently recruit markers of the early endosome (Rab5), recycling endosome (Rab4 and Rab11), and late endosome (Rab7), are maintained near neutral pH, do not fuse with lysosomes, exclude the protease cathepsin L, and engage the endoplasmic reticulum and Golgi apparatus for up to 21 days postinfection. Maintenance of this safe vacuolar niche requires active A. marginale protein synthesis; in its absence, the AmVs mature into acidic, protease-active phagolysosomes. Identification of this bacterially directed modeling of the tick midgut endosome provides a mechanistic basis for examination of the differences in transmission efficiency observed among A. marginale strains and among vector populations. IMPORTANCE: Ticks transmit a variety of intracellular bacterial pathogens that cause significant diseases in humans and animals. For successful transmission, these bacterial pathogens must first gain entry into the tick midgut digestive cells, avoid digestion, and establish a replicative niche without harming the tick vector. Little is known about how this replicative niche is established and maintained. Using the ruminant pathogen A. marginale and its natural tick vector, D. andersoni, this study characterized the features of the A. marginale niche in the tick midgut and demonstrates that A. marginale protein synthesis is required for the maintenance of this niche. This work opens a new line of inquiry about the pathogen effectors and their targets within the tick that mediate tick-pathogen interactions and ultimately serve as the determinants of pathogen success.


Assuntos
Anaplasma marginale/fisiologia , Vetores Aracnídeos/microbiologia , Dermacentor/microbiologia , Anaplasma marginale/genética , Anaplasma marginale/isolamento & purificação , Animais , Vetores Aracnídeos/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Citoplasma/metabolismo , Citoplasma/microbiologia , Dermacentor/metabolismo , Vacúolos/metabolismo , Vacúolos/microbiologia
8.
Appl Environ Microbiol ; 82(11): 3217-3224, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-26994084

RESUMO

UNLABELLED: The remarkable genetic diversity of vector-borne pathogens allows for the establishment of superinfection in the mammalian host. To have a long-term impact on population strain structure, the introduced strains must also be transmitted by a vector population that has been exposed to the existing primary strain. The sequential exposure of the vector to multiple strains frequently prevents establishment of the second strain, a phenomenon termed superinfection exclusion. As a consequence, superinfection exclusion may greatly limit genetic diversity in the host population, which is difficult to reconcile with the high degree of genetic diversity maintained among vector-borne pathogens. Using Anaplasma marginale, a tick-borne bacterial pathogen of ruminants, we hypothesized that superinfection exclusion is temporally dependent and that longer intervals between strain exposures allow successful acquisition and transmission of a superinfecting strain. To test this hypothesis, we sequentially exposed Dermacentor andersoni ticks to two readily tick-transmissible strains of A. marginale The tick feedings were either immediately sequential or 28 days apart. Ticks were allowed to transmission feed and were individually assessed to determine if they were infected with one or both strains. The second strain was excluded from the tick when the exposure interval was brief but not when it was prolonged. Midguts and salivary glands of individual ticks were superinfected and transmission of both strains occurred only when the exposure interval was prolonged. These findings indicate that superinfection exclusion is temporally dependent, which helps to account for the differences in pathogen strain structure in tropical compared to temperate regions. IMPORTANCE: Many vector-borne pathogens have marked genetic diversity, which influences pathogen traits such as transmissibility and virulence. The most successful strains are those that are preferentially transmitted by the vector. However, the factors that determine successful transmission of a particular strain are unknown. In the case of intracellular, bacterial, tick-borne pathogens, one potential factor is superinfection exclusion, in which colonization of ticks by the first strain of a pathogen it encounters prevents the transmission of a second strain. Using A. marginale, the most prevalent tick-borne pathogen of cattle worldwide, and its natural tick vector, we determined that superinfection exclusion occurs when the time between exposures to two strains is brief but not when it is prolonged. These findings suggest that superinfection exclusion may influence strain transmission in temperate regions, where tick activity is limited by season, but not in tropical regions, where ticks are active for long periods.


Assuntos
Anaplasma marginale/crescimento & desenvolvimento , Anaplasma marginale/isolamento & purificação , Antibiose , Vetores Aracnídeos/microbiologia , Dermacentor/microbiologia , Anaplasma marginale/classificação , Animais , Trato Gastrointestinal/microbiologia , Glândulas Salivares/microbiologia , Fatores de Tempo
9.
Infect Immun ; 83(11): 4178-84, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26259814

RESUMO

Antigenic variation allows microbial pathogens to evade immune clearance and establish persistent infection. Anaplasma marginale utilizes gene conversion of a repertoire of silent msp2 alleles into a single active expression site to encode unique Msp2 variants. As the genomic complement of msp2 alleles alone is insufficient to generate the number of variants required for persistence, A. marginale uses segmental gene conversion, in which oligonucleotide segments from multiple alleles are recombined into the expression site to generate a novel msp2 mosaic not represented elsewhere in the genome. Whether these segmental changes are sufficient to evade a broad antibody response is unknown. We addressed this question by identifying Msp2 variants that differed in primary structure within the immunogenic hypervariable region microdomains and tested whether they represented true antigenic variants. The minimal primary structural difference between variants was a single amino acid resulting from a codon insertion, and overall, the amino acid identity among paired microdomains ranged from 18 to 92%. Collectively, 89% of the expressed structural variants were also antigenic variants across all biological replicates, independent of a specific host major histocompatibility complex haplotype. Biological relevance is supported by the following: (i) all structural variants were expressed during infection of a natural host, (ii) the structural variation observed in the microdomains corresponded to the mean length of variants generated by segmental gene conversion, and (iii) antigenic variants were identified using a broad antibody response that developed during infection of a natural host. The findings demonstrate that segmental gene conversion efficiently generates Msp2 antigenic variants.


Assuntos
Anaplasma marginale/imunologia , Anaplasmose/imunologia , Variação Antigênica , Antígenos de Bactérias/química , Antígenos de Bactérias/imunologia , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/imunologia , Sequência de Aminoácidos , Anaplasma marginale/química , Anaplasma marginale/genética , Anaplasmose/microbiologia , Anticorpos Antibacterianos/imunologia , Antígenos de Bactérias/genética , Proteínas da Membrana Bacteriana Externa/genética , Humanos , Evasão da Resposta Imune , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Alinhamento de Sequência
10.
BMC Genomics ; 15: 278, 2014 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-24725301

RESUMO

BACKGROUND: The large amounts of data generated by genomics, transcriptomics and proteomics have increased our understanding of the biology of Anaplasma marginale. However, these data have also led to new assumptions that require testing, ideally through classical genetic mutation. One example is the definition of genes associated with virulence. Here we describe the molecular characterization of a red fluorescent and spectinomycin and streptomycin resistant A. marginale mutant generated by Himar1 transposon mutagenesis. RESULTS: High throughput genome sequencing to determine the Himar1-A. marginale genome junctions established that the transposon sequences were integrated within the coding region of the omp10 gene. This gene is arranged within an operon with AM1225 at the 5' end and with omp9, omp8, omp7 and omp6 arranged in tandem at the 3' end. RNA analysis to determine the effects of the transposon insertion on the expression of omp10 and downstream genes revealed that the Himar1 insertion not only reduced the expression of omp10 but also that of downstream genes. Transcript expression from omp9, and omp8 dropped by more than 90% in comparison with their counterparts in wild-type A. marginale. Immunoblot analysis showed a reduction in the production of Omp9 protein in these mutants compared to wild-type A. marginale. CONCLUSIONS: These results demonstrate that transposon mutagenesis in A. marginale is possible and that this technology can be used for the creation of insertional gene knockouts that can be evaluated in natural host-vector systems.


Assuntos
Anaplasma marginale/genética , Proteínas da Membrana Bacteriana Externa/genética , Elementos de DNA Transponíveis , Óperon , Sequência de Bases , Western Blotting , Cromossomos Bacterianos , DNA Bacteriano , Técnicas de Silenciamento de Genes , Genes Bacterianos , Dados de Sequência Molecular , Mutagênese
11.
Res Vet Sci ; 174: 105278, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38759348

RESUMO

Little research is available on acquired immunity to rabies in dogs and cats from Central Africa, particularly regarding the legal movements of pets. Movement of domestic animals from rabies-endemic countries like Cameroon to rabies free areas poses one of the main risks for rabies introduction into rabies-free areas. Thus, the aim of this study was to assess the effect of various risk factors on rabies vaccine efficacy in Cameroonian. Since the dependent variable, rabies neutralizing titres, were censored from above (right-censoring), Generalized Additive Model for Location, Scale and Shape (GAMLSS) was used in the analysis. Overall, 85.7% of dogs and 100% of cats had titres greater than or equal to 0.5 IU/mL, which is considered protective. Additionally, compared to cats, the value of the rabies-neutralizing serum titres in dogs was on average smaller by 2.3 IU/mL. For each additional year of age, the value of the rabies-neutralizing serum titre, on average, increased by approximately 0.14 IU/mL. Finally, for each 30 additional days between the date of the last rabies vaccination and the date of the sampling, the value the rabies neutralizing titre, on average, decreased by approximately 0.10 IU/mL, given the species and age at sampling were equivalent. These results are useful for assessing risk and improving surveillance to prevent the introduction of rabies into a country via the international movement of animals.


Assuntos
Doenças do Gato , Doenças do Cão , Vacina Antirrábica , Raiva , Animais , Cães , Gatos , Vacina Antirrábica/imunologia , Vacina Antirrábica/administração & dosagem , Doenças do Cão/prevenção & controle , Doenças do Cão/imunologia , Doenças do Gato/prevenção & controle , Doenças do Gato/imunologia , Doenças do Gato/virologia , Raiva/prevenção & controle , Raiva/veterinária , Fatores de Risco , Camarões , Viagem , Masculino , Feminino , Vacinação/veterinária
12.
Infect Immun ; 81(5): 1852-8, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23509140

RESUMO

Anaplasma marginale subsp. centrale was the first vaccine used to protect against a rickettsial disease and is still in widespread use a century later. As its use preceded development of either cryopreservation or cell culture, the vaccine strain was maintained for decades by sequential passage among donor animals, excluding the natural tick-borne transmission cycle that provides a selective pressure or population "bottleneck." We demonstrated that the vaccine strain is genetically heterogeneous at 46 chromosomal loci and that heterogeneity was maintained upon inoculation into recipient animals. The number of variants per site ranged from 2 to 11 with a mean of 2.8/locus and a mode and median of 2/locus; variants included single-nucleotide polymorphisms, insertions/deletions, polynucleotide tracts, and different numbers of perfect repeats. The genetic heterogeneity is highly unlikely to be a result of strain contamination based on analysis using a panel of eight gene markers with a high power for strain discrimination. In contrast, heterogeneity appears to be a result of genetic drift in the absence of the restriction of tick passage. Heterogeneity could be reduced following tick passage, and the reduced heterogeneity could be maintained in sequential intravenous and tick-borne passages. The reduction in vaccine strain heterogeneity following tick passage did not confer an enhanced transmission phenotype, indicating that a stochastically determined population bottleneck was likely responsible as opposed to a positive selective pressure. These findings demonstrate the plasticity of an otherwise highly constrained genome and highlight the role of natural transmission cycles in shaping and maintaining the bacterial genome.


Assuntos
Anaplasma marginale/genética , Anaplasma marginale/imunologia , Anaplasmose/transmissão , Vacinas Bacterianas/genética , Heterogeneidade Genética , Anaplasmose/prevenção & controle , Animais , Carrapatos/microbiologia
13.
Microorganisms ; 11(5)2023 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-37317070

RESUMO

Anaplasma marginale is a tick-borne pathogen that causes bovine anaplasmosis, which affects cattle around the world. Despite its broad prevalence and severe economic impacts, limited treatments exist for this disease. Our lab previously reported that a high proportion of Rickettsia bellii, a tick endosymbiont, in the microbiome of a population of Dermacentor andersoni ticks negatively impacts the ticks' ability to acquire A. marginale. To better understand this correlation, we used mixed infection of A. marginale and R. bellii in D. andersoni cell culture. We assessed the impacts of different amounts of R. bellii in coinfections, as well as established R. bellii infection, on the ability of A. marginale to establish an infection and grow in D. andersoni cells. From these experiments, we conclude that A. marginale is less able to establish an infection in the presence of R. bellii and that an established R. bellii infection inhibits A. marginale replication. This interaction highlights the importance of the microbiome in preventing tick vector competence and may lead to the development of a biological or mechanistic control for A. marginale transmission by the tick.

14.
Front Cell Infect Microbiol ; 12: 877525, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35711652

RESUMO

Tick midgut is the primary infection site required by tick-borne pathogens to initiate their development for transmission. Despite the biological significance of this organ, cell cultures derived exclusively from tick midgut tissues are unavailable and protocols for generating primary midgut cell cultures have not been described. To study the mechanism of Anaplasma marginale-tick cell interactions, we successfully developed an in vitro Dermacentor andersoni primary midgut cell culture system. Midgut cells were maintained for up to 120 days. We demonstrated the infection of in vitro midgut cells by using an A. marginale omp10::himar1 mutant with continued replication for up to 10 days post-infection. Anaplasma marginale infection of midgut cells regulated the differential expression of tick α-(1,3)-fucosyltransferases A1 and A2. Silencing of α-(1,3)-fucosyltransferase A2 in uninfected midgut cells reduced the display of fucosylated glycans and significantly lowered the susceptibility of midgut cells to A. marginale infection, suggesting that the pathogen utilized core α-(1,3)-fucose of N-glycans to infect tick midgut cells. This is the first report using in vitro primary D. andersoni midgut cells to study A. marginale-tick cell interactions at the molecular level. The primary midgut cell culture system will further facilitate the investigation of tick-pathogen interactions, leading to the development of novel intervention strategies for tick-borne diseases.


Assuntos
Anaplasma marginale , Anaplasmose , Dermacentor , Anaplasma , Anaplasma marginale/genética , Animais , Técnicas de Cultura de Células , Dermacentor/metabolismo , Polissacarídeos/metabolismo
15.
mBio ; 13(4): e0070322, 2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-35862781

RESUMO

The insect immune deficiency (IMD) pathway is a defense mechanism that senses and responds to Gram-negative bacteria. Ticks lack genes encoding upstream components that initiate the IMD pathway. Despite this deficiency, core signaling molecules are present and functionally restrict tick-borne pathogens. The molecular events preceding activation remain undefined. Here, we show that the unfolded-protein response (UPR) initiates the IMD network. The endoplasmic reticulum (ER) stress receptor IRE1α is phosphorylated in response to tick-borne bacteria but does not splice the mRNA encoding XBP1. Instead, through protein modeling and reciprocal pulldowns, we show that Ixodes IRE1α complexes with TRAF2. Disrupting IRE1α-TRAF2 signaling blocks IMD pathway activation and diminishes the production of reactive oxygen species. Through in vitro, in vivo, and ex vivo techniques, we demonstrate that the UPR-IMD pathway circuitry limits the Lyme disease-causing spirochete Borrelia burgdorferi and the rickettsial agents Anaplasma phagocytophilum and A. marginale (anaplasmosis). Altogether, our study uncovers a novel linkage between the UPR and the IMD pathway in arthropods. IMPORTANCE The ability of an arthropod to harbor and transmit pathogens is termed "vector competency." Many factors influence vector competency, including how arthropod immune processes respond to the microbe. Divergences in innate immunity between arthropods are increasingly being reported. For instance, although ticks lack genes encoding key upstream molecules of the immune deficiency (IMD) pathway, it is still functional and restricts causative agents of Lyme disease (Borrelia burgdorferi) and anaplasmosis (Anaplasma phagocytophilum). How the IMD pathway is activated in ticks without classically defined pathway initiators is not known. Here, we found that a cellular stress response network, the unfolded-protein response (UPR), functions upstream to induce the IMD pathway and restrict transmissible pathogens. Collectively, this explains how the IMD pathway can be activated in the absence of canonical pathway initiators. Given that the UPR is highly conserved, UPR-initiated immunity may be a fundamental principle impacting vector competency across arthropods.


Assuntos
Anaplasma phagocytophilum , Anaplasmose , Artrópodes , Borrelia burgdorferi , Ixodes , Doença de Lyme , Anaplasma phagocytophilum/fisiologia , Animais , Endorribonucleases , Ixodes/genética , Ixodes/microbiologia , Proteínas Serina-Treonina Quinases , Fator 2 Associado a Receptor de TNF
16.
Infect Immun ; 79(12): 4941-6, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21930762

RESUMO

Ticks serve as biological vectors for a wide variety of bacterial pathogens which must be able to efficiently colonize specific tick tissues prior to transmission. The bacterial determinants of tick colonization are largely unknown, a knowledge gap attributed in large part to the paucity of tools to genetically manipulate these pathogens. In this study, we demonstrated that Francisella tularensis subsp. novicida, for which a complete two-allele transposon mutant library has been constructed, initially infects the midguts of 100% of acquisition-fed Dermacentor andersoni nymphs, with stable colonization and replication during a subsequent molt. Increased dissemination to and marked replication within the salivary gland was closely linked to a second (transmission) feed and culminated in secretion of bacteria into the saliva and successful transmission. Simultaneous testing of multiple mutants resulted in total bacterial levels similar to those observed for single mutants. However, there was evidence of a bottleneck during colonization, resulting in a founder effect in which the most successful mutant varied when comparing individual ticks. Thus, it is essential to assess mutant success at the level of the tick population rather than in individual ticks. The ability of F. tularensis subsp. novicida to recapitulate the key physiological events by which bacteria colonize and are transmitted by ixodid ticks provides a new genome-wide approach to identify the required pathogen molecules and pathways. The identification of specific genes and, more importantly, conserved pathways that function at the tick-pathogen interface will accelerate the development of new methods to block transmission.


Assuntos
Vetores Aracnídeos/microbiologia , Dermacentor/microbiologia , Francisella tularensis/fisiologia , Tularemia/transmissão , Animais , Vetores Aracnídeos/fisiologia , Quitinases/genética , Dermacentor/fisiologia , Comportamento Alimentar/fisiologia , Feminino , Francisella tularensis/genética , Francisella tularensis/crescimento & desenvolvimento , Regulação Bacteriana da Expressão Gênica/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Ninfa/microbiologia , Ninfa/fisiologia , Coelhos , Tularemia/microbiologia
17.
Infect Immun ; 79(7): 2847-55, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21576345

RESUMO

Transmission of tick-borne pathogens requires transition between distinct host environments with infection and replication in host-specific cell types. Anaplasma marginale illustrates this transition: in the mammalian host, the bacterium infects and replicates in mature (nonnucleated) erythrocytes, while in the tick vector, replication occurs in nucleated epithelial cells. We hypothesized that proteins containing ankyrin motifs would be expressed by A. marginale only in tick cells and would traffic to the infected host cell nucleus. A. marginale encodes three proteins containing ankyrin motifs, an AnkA orthologue (the AM705 protein), AnkB (the AM926 protein), and AnkC (the AM638 protein). All three A. marginale Anks were confirmed to be expressed during intracellular infection: AnkA is expressed at significantly higher levels in erythrocytes, AnkB is expressed equally by both infected erythrocytes and tick cells, and AnkC is expressed exclusively in tick cells. There was no evidence of any of the Ank proteins trafficking to the nucleus. Thus, the hypothesis that ankyrin-containing motifs were predictive of cell type expression and nuclear localization was rejected. In contrast, AnkA orthologues in the closely related A. phagocytophilum and Ehrlichia chaffeensis have been shown to localize to the host cell nucleus. This difference, together with the lack of a nuclear localization signal in any of the AnkA orthologues, suggests that trafficking may be mediated by a separate transporter rather than by endogenous signals. Selection for divergence in Ank function among Anaplasma and Ehrlichia spp. is supported by both locus and allelic analyses of genes encoding orthologous proteins and their ankyrin motif compositions.


Assuntos
Anaplasma marginale/metabolismo , Anaplasmose/microbiologia , Repetição de Anquirina , Vetores Aracnídeos/microbiologia , Proteínas de Bactérias/genética , Eritrócitos/microbiologia , Carrapatos/microbiologia , Anaplasma marginale/genética , Anaplasma marginale/crescimento & desenvolvimento , Anaplasmose/transmissão , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Linhagem Celular , Dermacentor/microbiologia , Ehrlichia chaffeensis/genética , Ehrlichia chaffeensis/crescimento & desenvolvimento , Ehrlichia chaffeensis/metabolismo , Regulação Bacteriana da Expressão Gênica , Camundongos , Dados de Sequência Molecular , Sintenia
18.
Appl Environ Microbiol ; 77(1): 330-4, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21057014

RESUMO

We tested the stability and tick transmission phenotype of transformed Anaplasma marginale through a complete in vivo infection cycle. Similar to the wild type, the gfp-transformed A. marginale strain established infection in cattle, a natural reservoir host, and persisted in immune competent animals. The tick infection rates for the transformed A. marginale and the wild type were the same. However, there were significantly lower levels of the transformed A. marginale than of the wild type in the tick. Despite the lower levels of replication, ticks transmitted the transformant. Transformants can serve as valuable tools to dissect the molecular requirements of tick colonization and pathogen transmission.


Assuntos
Anaplasma marginale/isolamento & purificação , Anaplasmose/microbiologia , Doenças dos Bovinos/microbiologia , Reservatórios de Doenças , Carrapatos/crescimento & desenvolvimento , Carrapatos/microbiologia , Anaplasma marginale/genética , Anaplasmose/transmissão , Animais , Bovinos , Doenças dos Bovinos/transmissão , Transmissão de Doença Infecciosa , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Coloração e Rotulagem/métodos
19.
Ticks Tick Borne Dis ; 12(1): 101584, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33059171

RESUMO

For most organisms, iron is an essential nutrient due to its role in fundamental cellular processes. Insufficient iron causes sub-optimal metabolism with potential effects on viability, while high levels of iron are toxic due to the formation of oxidative radicals, which damage cellular components. Many molecules and processes employed in iron uptake, storage, transport and metabolism are conserved, however significant knowledge gaps remain regarding these processes in ticks due to their unique physiology. In this study, we first identified and sequenced 13 genes likely to be involved in iron metabolism in Dermacentor andersoni cells. We then developed a method to reduce iron levels in D. andersoni cells using the iron chelator 2,2'-bipyridyl and measured the transcriptional response of these genes to iron reduction. The genes include a putative transferrin receptor, divalent metal transporter 1, duodenal cytochrome b, zinc/iron transporters zip7, zip13, zip14, mitoferrin, ferrochelatase, iron regulatory protein 1, ferritin1, ferritin2, transferrin and poly r(C)-binding protein. Overall, the transcriptional response of the target genes to iron reduction was modest. The most marked changes were a decrease in ferritin2, which transports iron through the tick hemolymph, the mitochondrial iron transporter mitoferrin, and the mitochondrial enzyme ferrochelatase. Iron regulatory protein1 was the only gene with an overall increase in transcript in response to reduced iron levels. This work lays the foundation for an improved understanding of iron metabolism in ticks which may provide molecular targets for the development of novel tick control methods and aid in the understanding of tick-pathogen interactions.


Assuntos
Proteínas de Artrópodes/genética , Dermacentor/genética , Ferro/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Artrópodes/química , Proteínas de Artrópodes/metabolismo , Dermacentor/metabolismo , Perfilação da Expressão Gênica , Alinhamento de Sequência
20.
Parasit Vectors ; 14(1): 157, 2021 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-33726815

RESUMO

BACKGROUND: Theileria orientalis is a tick-borne hemoparasite that causes anemia, ill thrift, and death in cattle globally. The Ikeda strain of T. orientalis is more virulent than other strains, leading to severe clinical signs and death of up to 5% of affected animals. Within the Asia-Pacific region, where it affects 25% of Australian cattle, T. orientalis Ikeda has a significant economic impact on the cattle industry. In 2017, T. orientalis Ikeda was detected in a cattle herd in Albermarle County, Virginia, United States. Months earlier, the U.S. was alerted to the invasion of the Asian longhorned tick, Haemaphysalis longicornis, throughout the eastern U.S. Abundant H. longicornis ticks were identified on cattle in the T. orientalis-affected herd in VA, and a subset of ticks from the environment were PCR-positive for T. orientalis Ikeda. A strain of T. orientalis from a previous U.S. outbreak was not transmissible by H. longicornis; however, H. longicornis is the primary tick vector of T. orientalis Ikeda in other regions of the world. Thus, the objective of this study was to determine whether invasive H. longicornis ticks in the U.S. are competent vectors of T. orientalis Ikeda. METHODS: Nymphal H. longicornis ticks were fed on a splenectomized calf infected with the VA-U.S.-T. orientalis Ikeda strain. After molting, a subset of adult ticks from this cohort were dissected, and salivary glands assayed for T. orientalis Ikeda via qPCR. The remaining adult ticks from the group were allowed to feed on three calves. Calves were subsequently monitored for T. orientalis Ikeda infection via blood smear cytology and PCR. RESULTS: After acquisition feeding on a VA-U.S.-T. orientalis Ikeda-infected calf as nymphs, a subset of molted adult tick salivary glands tested positive by qPCR for T. orientalis Ikeda. Adult ticks from the same cohort successfully transmitted T. orientalis Ikeda to 3/3 naïve calves, each of which developed parasitemia reaching 0.4-0.9%. CONCLUSIONS: Our findings demonstrate that U.S. H. longicornis ticks are competent vectors of the VA-U.S.-T. orientalis Ikeda strain. This data provides important information for the U.S. cattle industry regarding the potential spread of this parasite and the necessity of enhanced surveillance and control measures.


Assuntos
Doenças dos Bovinos/parasitologia , Doenças dos Bovinos/transmissão , Surtos de Doenças/veterinária , Genótipo , Theileria/genética , Theileriose/transmissão , Carrapatos/parasitologia , Animais , Ásia , Bovinos , Masculino , Parasitemia/epidemiologia , Theileria/isolamento & purificação , Theileriose/parasitologia , Estados Unidos/epidemiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA