Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Acta Trop ; 220: 105933, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33895143

ABSTRACT

Anaplasmosis is a tick-borne disease caused by the intracellular rickettsia Anaplasma marginale, which affects cattle and other ruminants in both tropical and subtropical regions of the world, and also causing tremendous economic losses due to decreasing livestock production. The major surface protein 5 (MSP5) of A. marginale is an immunodominant and highly conserved protein encoding by a single gene. In the present study, the complete full-length of the msp5 coding sequence of A. marginale Thailand strain was cloned and determined at a size of 633 bp. Phylogenetic analysis based on neigh-joining (NJ) method showed that the msp5 sequence Thailand strains were clearly distributed in 3rd clade and conserved when compared with other strains. The results showed 9 haplotypes of the msp5 genes, and the entropy analysis of MSP5 amino acid sequences displayed 92 high entropy peaks with value ranging from 0.198 to 0.845 Additionally, a recombinant MSP5 of A. marginale (rAmMSP5) was over-expressed in the E. coli BL21 Star™ (DE3) host cell, affinity purified, and found in SDS-PAGE at a molecular weight of 26 kDa. The antigenicity of rAmMSP5 (26 kDa) and AmMSP5 (19 kDa) was recognized by rabbit anti-rAmMSP5 antisera and A. marginale-infected cattle sera. Both rAmMSP5 and AmMSP5 were perceived by these sera manifesting that recombinant and native AmMSP5 have conserved epitopes. Immunofluorescence technique using rabbit anti-rAmMSP5 antisera exhibited that the AmMSP5 is distributed on both the membrane and the outside of infected erythrocytes. Therefore, the recombinant MSP5 could be used for the development of immunodiagnostic assays and vaccine purposes for controlling anaplasmosis.


Subject(s)
Anaplasma marginale/genetics , Anaplasma marginale/metabolism , Bacterial Outer Membrane Proteins/genetics , Amino Acid Sequence , Anaplasmosis/microbiology , Animals , Epitopes , Phylogeny , Rabbits , Recombinant Proteins/immunology , Thailand , Tick-Borne Diseases
2.
Acta Trop ; 197: 105047, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31158343

ABSTRACT

Anaplasma marginale is the rickettsia which causes the bovine anaplasmosis. The distribution of A. marginale is both tropical and subtropical regions of the world. The major surface protein 4 (MSP4) of this parasite was identified as an immunodominant protein. In this study, the full length of DNA encoding A. marginale MSP4 (AmMSP4) was cloned from the parasites. The open reading frame of msp4 coding sequence of Thailand strain is 849 bp. Phylogenetic analysis revealed that the msp4 coding sequence of A. marginale was highly conserved when compared with Anaplasma phagocytophilum. The recombinant plasmid was further transformed into the BL21-CodonPlus (DE3)-RIPL competent cells for over-expression of the recombinant major surface protein 4 of A. marginale (rAmMSP4). Sera from rabbit immunized with rAmMSP4 and from cattle infected with A. marginale were used to study the antigenicity of rAmMSP4 (35 kDa) and AmMSP4 (31 kDa). Both rAmMSP4 and AmMSP4 were recognized by these sera showing that recombinant and native AmMSP4 have conserved epitopes. Localization of Anaplasma parasites by immunofluorescence showed these parasites are distributed on both the membrane and the outside of infected erythrocytes. Regarding antigenicity, recombinant MSP4 could be used for immunodiagnostic purposes and as a possible vaccine candidate against anaplasmosis.


Subject(s)
Anaplasma marginale/metabolism , Anaplasmosis/microbiology , Bacterial Proteins/metabolism , Cattle Diseases/microbiology , Membrane Proteins/metabolism , Tick-Borne Diseases/microbiology , Anaplasma marginale/genetics , Anaplasmosis/genetics , Anaplasmosis/immunology , Animals , Cattle , Cattle Diseases/genetics , Cattle Diseases/immunology , Phylogeny , Rabbits , Sequence Analysis, DNA
3.
Mol Microbiol ; 107(2): 142-163, 2018 01.
Article in English | MEDLINE | ID: mdl-29178391

ABSTRACT

Peptidoglycan is the predominant stress-bearing structure in the cell envelope of most bacteria, and also a potent stimulator of the eukaryotic immune system. Obligate intracellular bacteria replicate exclusively within the interior of living cells, an osmotically protected niche. Under these conditions peptidoglycan is not necessarily needed to maintain the integrity of the bacterial cell. Moreover, the presence of peptidoglycan puts bacteria at risk of detection and destruction by host peptidoglycan recognition factors and downstream effectors. This has resulted in a selective pressure and opportunity to reduce the levels of peptidoglycan. In this review we have analysed the occurrence of genes involved in peptidoglycan metabolism across the major obligate intracellular bacterial species. From this comparative analysis, we have identified a group of predicted 'peptidoglycan-intermediate' organisms that includes the Chlamydiae, Orientia tsutsugamushi, Wolbachia and Anaplasma marginale. This grouping is likely to reflect biological differences in their infection cycle compared with peptidoglycan-negative obligate intracellular bacteria such as Ehrlichia and Anaplasma phagocytophilum, as well as obligate intracellular bacteria with classical peptidoglycan such as Coxiella, Buchnera and members of the Rickettsia genus. The signature gene set of the peptidoglycan-intermediate group reveals insights into minimal enzymatic requirements for building a peptidoglycan-like sacculus and/or division septum.


Subject(s)
Bacteria , Host Microbial Interactions , Intracellular Space/microbiology , Peptidoglycan/genetics , Peptidoglycan/metabolism , Anaplasma marginale/classification , Anaplasma marginale/genetics , Anaplasma marginale/immunology , Anaplasma marginale/metabolism , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/immunology , Bacteria/metabolism , Cell Wall/metabolism , Chlamydia/classification , Chlamydia/genetics , Chlamydia/immunology , Chlamydia/metabolism , Cytoplasm/metabolism , Genome, Bacterial/genetics , Humans , Immunity, Innate/immunology , Orientia tsutsugamushi/classification , Orientia tsutsugamushi/genetics , Orientia tsutsugamushi/immunology , Orientia tsutsugamushi/metabolism , Peptidoglycan/chemistry , Phylogeny , Wolbachia/classification , Wolbachia/genetics , Wolbachia/immunology , Wolbachia/metabolism
4.
Res Vet Sci ; 97(3): 514-20, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25458492

ABSTRACT

Anaplasma marginale is an intraerythrocytic vector-borne infectious agent of cattle. Immunization with the current vaccine, based on parasitized erythrocytes with live Anaplasma centrale, shows some constraints and confers partial protection, suggesting the feasibility for the development of new generation of vaccines. The aim of the present study was to assess the effect of sequential immunization of BALB/c mice, with herpesvirus amplicon vector-based vaccines combined with protein-based vaccines, on the quality of the immune response against the major surface protein 5 of A. marginale. The highest antibody titers against MSP5 were elicited in mice that received two doses of adjuvanted recombinant protein (p < 0.0001). Mice treated with a heterologous prime-boost strategy generated sustained antibody titers at least up to 200 days, and a higher specific cellular response. The results presented here showed that sequential immunization with HSV-based vectors and purified antigen enhances the quality of the immune response against A. marginale.


Subject(s)
Anaplasma marginale/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Immunity, Innate , Anaplasma marginale/genetics , Anaplasma marginale/metabolism , Anaplasmosis/prevention & control , Animals , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Bacterial Vaccines/virology , Cattle , Cattle Diseases/prevention & control , Cell Line, Tumor , Chlorocebus aethiops , Genetic Vectors/genetics , Herpesvirus 1, Human/genetics , Mice , Mice, Inbred BALB C , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Vero Cells
5.
PLoS One ; 7(3): e33605, 2012.
Article in English | MEDLINE | ID: mdl-22438962

ABSTRACT

The twin-arginine translocation (Tat) pathway exports fully folded proteins out of the cytoplasm of Gram-negative and Gram-positive bacteria. Although much progress has been made in unraveling the molecular mechanism and biochemical characterization of the Tat system, little is known concerning its functionality and biological role to confer adaptive skills, symbiosis or pathogenesis in the α-proteobacteria class. A comparative genomic analysis in the α-proteobacteria class confirmed the presence of tatA, tatB, and tatC genes in almost all genomes, but significant variations in gene synteny and rearrangements were found in the order Rickettsiales with respect to the typically described operon organization. Transcription of tat genes was confirmed for Anaplasma marginale str. St. Maries and Brucella abortus 2308, two α-proteobacteria with full and partial intracellular lifestyles, respectively. The tat genes of A. marginale are scattered throughout the genome, in contrast to the more generalized operon organization. Particularly, tatA showed an approximately 20-fold increase in mRNA levels relative to tatB and tatC. We showed Tat functionality in B. abortus 2308 for the first time, and confirmed conservation of functionality in A. marginale. We present the first experimental description of the Tat system in the Anaplasmataceae and Brucellaceae families. In particular, in A. marginale Tat functionality is conserved despite operon splitting as a consequence of genome rearrangements. Further studies will be required to understand how the proper stoichiometry of the Tat protein complex and its biological role are achieved. In addition, the predicted substrates might be the evidence of role of the Tat translocation system in the transition process from a free-living to a parasitic lifestyle in these α-proteobacteria.


Subject(s)
Alphaproteobacteria/genetics , Alphaproteobacteria/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Amino Acid Sequence , Anaplasma marginale/genetics , Anaplasma marginale/metabolism , Brucella abortus/genetics , Brucella abortus/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Genetic Complementation Test , Genetic Variation , Genome, Bacterial , Molecular Sequence Data , Multigene Family , Phenotype , Phylogeny , Protein Transport , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Species Specificity
6.
PLoS One ; 6(11): e27724, 2011.
Article in English | MEDLINE | ID: mdl-22140462

ABSTRACT

BACKGROUND: Anaplasma marginale, an obligate intracellular alphaproteobacterium in the order Rickettsiales, is a tick-borne pathogen and the leading cause of anaplasmosis in cattle worldwide. Complete genome sequencing of A. marginale revealed that it has a type IV secretion system (T4SS). The T4SS is one of seven known types of secretion systems utilized by bacteria, with the type III and IV secretion systems particularly prevalent among pathogenic Gram-negative bacteria. The T4SS is predicted to play an important role in the invasion and pathogenesis of A. marginale by translocating effector proteins across its membrane into eukaryotic target cells. However, T4SS effector proteins have not been identified and tested in the laboratory until now. RESULTS: By combining computational methods with phylogenetic analysis and sequence identity searches, we identified a subset of potential T4SS effectors in A. marginale strain St. Maries and chose six for laboratory testing. Four (AM185, AM470, AM705 [AnkA], and AM1141) of these six proteins were translocated in a T4SS-dependent manner using Legionella pneumophila as a reporter system. CONCLUSIONS: The algorithm employed to find T4SS effector proteins in A. marginale identified four such proteins that were verified by laboratory testing. L. pneumophila was shown to work as a model system for A. marginale and thus can be used as a screening tool for A. marginale effector proteins. The first T4SS effector proteins for A. marginale have been identified in this work.


Subject(s)
Anaplasma marginale/metabolism , Bacterial Proteins/metabolism , Bacterial Secretion Systems , Adenylate Cyclase Toxin/metabolism , Anaplasma marginale/genetics , Animals , Bacterial Proteins/genetics , Biological Assay , Cattle , Gene Order/genetics , Genome, Bacterial/genetics , Protein Transport , Reproducibility of Results
7.
Infect Immun ; 79(7): 2847-55, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21576345

ABSTRACT

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.


Subject(s)
Anaplasma marginale/metabolism , Anaplasmosis/microbiology , Ankyrin Repeat , Arachnid Vectors/microbiology , Bacterial Proteins/genetics , Erythrocytes/microbiology , Ticks/microbiology , Anaplasma marginale/genetics , Anaplasma marginale/growth & development , Anaplasmosis/transmission , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cell Line , Dermacentor/microbiology , Ehrlichia chaffeensis/genetics , Ehrlichia chaffeensis/growth & development , Ehrlichia chaffeensis/metabolism , Gene Expression Regulation, Bacterial , Mice , Molecular Sequence Data , Synteny
8.
Genet Mol Res ; 7(2): 460-6, 2008.
Article in English | MEDLINE | ID: mdl-18561379

ABSTRACT

Anaplasma marginale, a tick-borne bacterium, causes bovine anaplasmosis responsible for significant economic losses in tropical and subtropical regions worldwide. Various major outer membranes have been described, and VirB9, a type IV secretion system protein, has been recently indicated as a candidate in vaccine development against anaplasmosis. The virB9 gene of an A. marginale strain isolated in Paraná, Brazil, was cloned by polymerase chain reaction and sequenced; its cloning into the pETSUMO vector produced a virB9-SUMO-6x His fusion gene construct. This recombinant clone was over-expressed in Escherichia coli BL21 (DE3), and the expressed fusion protein was solubilized with urea and purified with an Ni-NTA column. This method produced a relatively high yield of rVirB9. The deduced amino acid sequence encoded by VirB9 showed 99% homology to A. marginale isolates from St. Maries. rVirB9 was recognized by serum from cattle immunized with PR1 strain and by bovine sera infected with heterologous strains, showing that rVirB9 has conserved epitopes, which suggests that rVirB9 could be useful for the development of a vaccine against anaplasmosis.


Subject(s)
Anaplasma marginale/genetics , Antigens, Bacterial/genetics , Bacterial Outer Membrane Proteins/genetics , Anaplasma marginale/isolation & purification , Anaplasma marginale/metabolism , Anaplasmosis/immunology , Anaplasmosis/microbiology , Animals , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/metabolism , Blotting, Western , Brazil , Cattle , Cattle Diseases/immunology , Cattle Diseases/microbiology , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Sequence Analysis, DNA
9.
Genet. mol. res. (Online) ; 7(2): 460-466, 2008. ilus
Article in English | LILACS | ID: lil-640992

ABSTRACT

Anaplasma marginale, a tick-borne bacterium, causes bovine anaplasmosis responsible for significant economic losses in tropical and subtropical regions worldwide. Various major outer membranes have been described, and VirB9, a type IV secretion system protein, has been recently indicated as a candidate in vaccine development against anaplasmosis. The virB9 gene of an A. marginale strain isolated in Paraná, Brazil, was cloned by polymerase chain reaction and sequenced; its cloning into the pETSUMO vector produced a virB9-SUMO-6x His fusion gene construct. This recombinant clone was over-expressed in Escherichia coli BL21 (DE3), and the expressed fusion protein was solubilized with urea and purified with an Ni-NTA column. This method produced a relatively high yield of rVirB9. The deduced amino acid sequence encoded by VirB9 showed 99% homology to A. marginale isolates from St. Maries. rVirB9 was recognized by serum from cattle immunized with PR1 strain and by bovine sera infected with heterologous strains, showing that rVirB9 has conserved epitopes, which suggests that rVirB9 could be useful for the development of a vaccine against anaplasmosis.


Subject(s)
Animals , Anaplasma marginale/genetics , Antigens, Bacterial/genetics , Bacterial Outer Membrane Proteins/genetics , Anaplasma marginale/isolation & purification , Anaplasma marginale/metabolism , Anaplasmosis/immunology , Anaplasmosis/microbiology , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Blotting, Western , Brazil , Cloning, Molecular , Cattle Diseases/immunology , Cattle Diseases/microbiology , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/metabolism , Sequence Analysis, DNA
10.
Infect Immun ; 75(6): 2959-64, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17420231

ABSTRACT

Understanding the determinants of efficient tick-borne microbial transmission is needed to better predict the emergence of highly transmissible pathogen strains and disease outbreaks. Although the basic developmental cycle of Anaplasma and Ehrlichia spp. within the tick has been delineated, there are marked differences in the ability of specific strains to be efficiently tick transmitted. Using the highly transmissible St. Maries strain of Anaplasma marginale in Dermacentor andersoni as a positive control and two unrelated nontransmissible strains, we identified distinct barriers to efficient transmission within the tick. The Mississippi strain was unable to establish infection at the level of the midgut epithelium despite successful ingestion of infected blood following acquisition feeding on a bacteremic animal host. This inability to colonize the midgut epithelium prevented subsequent development within the salivary glands and transmission. In contrast, A. marginale subsp. centrale colonized the midgut and then the salivary glands, replicating to a titer indistinguishable from that of the highly transmissible St. Maries strain and at least 100 times greater than that previously associated with successful transmission. Nonetheless, A. marginale subsp. centrale was not transmitted, even when a large number of infected ticks was used for transmission feeding. These results establish that there are at least two specific barriers to efficient tick-borne transmission, the midgut and salivary glands, and highlight the complexity of the pathogen-tick interaction.


Subject(s)
Anaplasma marginale/physiology , Anaplasmosis/transmission , Arachnid Vectors/metabolism , Salivary Glands/physiology , Ticks/microbiology , Anaplasma marginale/genetics , Anaplasma marginale/metabolism , Anaplasmosis/microbiology , Animals , Arachnid Vectors/microbiology , Cattle , Digestive System/physiopathology
11.
Infect Immun ; 72(7): 3883-9, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15213131

ABSTRACT

The msp2 and p44 genes encode polymorphic major outer membrane proteins that are considered unique to the intraerythrocytic agent of Anaplasma marginale and the intragranulocytic agent of Anaplasma phagocytophilum, respectively. In the present study, however, we found an msp2 gene in A. phagocytophilum that was remarkably conserved among A. phagocytophilum strains from human granulocytic anaplasmosis (HGA) patients, ticks, and a horse from various regions in the United States, but the gene was different in a sheep isolate from the United Kingdom. The msp2 gene in the A. phagocytophilum strain HZ genome was a single-copy gene and was located downstream of two Ehrlichia chaffeensis omp-1 homologs and a decarboxylase gene (ubiD). The msp2 gene was expressed by A. phagocytophilum in the blood from HGA patients NY36 and NY37 and by A. phagocytophilum isolates from these patients cultured in HL-60 cells at 37 degrees C. The msp2 gene was also expressed in a DBA/2 mouse infected by attaching ticks infected with strain NTN-1 and in a horse experimentally infected by attaching strain HZ-infected ticks. However, the transcript of the msp2 gene was undetectable in A. phagocytophilum strain HZ in SCID mice and Ixodes scapularis ticks infected with strain NTN-1. These results indicate that msp2 is functional in various strains of A. phagocytophilum, and relative expression ratios of msp2 to p44 vary in different infected hosts. These findings may be important in understanding roles that Msp2 proteins play in granulocytic ehrlichia infection and evolution of the polymorphic major outer membrane protein gene families in Anaplasma species.


Subject(s)
Anaplasma phagocytophilum/genetics , Bacterial Outer Membrane Proteins/genetics , Anaplasma marginale/genetics , Anaplasma marginale/metabolism , Anaplasma phagocytophilum/metabolism , Anaplasmataceae Infections/metabolism , Anaplasmataceae Infections/veterinary , Animals , Bacterial Outer Membrane Proteins/metabolism , Horses/microbiology , Humans , Mice , Molecular Sequence Data , Multigene Family , Phylogeny , Promoter Regions, Genetic , Ticks/microbiology , Transcription, Genetic
12.
Vet Microbiol ; 100(1-2): 55-64, 2004 May 20.
Article in English | MEDLINE | ID: mdl-15135513

ABSTRACT

Anaplasma centrale msp4 and msp5 genes were cloned and sequenced, and the recombinant proteins were expressed. The identity between Anaplasma marginale and A. centrale MSP4 was 83% in the nucleotide sequences and 91.7% in the encoded protein sequences. A. centrale msp5 nucleotide sequences shared 86.8% identity with A. marginale msp5, and there was 92.9% homology between A. centrale and A. marginale encoded amino acids of the MSP5 protein. Southern blots hybridized with probes derived from the msp4 and msp5 central regions indicate that msp4 and msp5 of A. centrale are encoded by single copy genes. Recombinant MSP4 and MSP5 fusion proteins reacted with anti-A. marginale monoclonal antibodies ANAR76A1 and ANAF16C, respectively, demonstrating the conservation of conformation-sensitive B-cell epitopes between A. centrale and A. marginale. These data demonstrate the structural and antigenic conservation of MSP4 and MSP5 in A. centrale and A. marginale. This conservation is consistent with the cross-protective immunity between A. marginale and A. centrale and supports the development of improved vaccines based upon common outer membrane proteins.


Subject(s)
Anaplasma centrale/genetics , Anaplasma marginale/genetics , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Bacterial Vaccines/genetics , Membrane Proteins/genetics , Amino Acid Sequence , Anaplasma centrale/immunology , Anaplasma centrale/metabolism , Anaplasma marginale/immunology , Anaplasma marginale/metabolism , Anaplasmosis/immunology , Anaplasmosis/microbiology , Animals , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/immunology , Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Bacterial Vaccines/chemistry , Bacterial Vaccines/immunology , Base Sequence , Blotting, Southern , Cattle , Cloning, Molecular , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Escherichia coli/genetics , Immunoblotting , Membrane Proteins/chemistry , Membrane Proteins/immunology , Molecular Sequence Data , Polymerase Chain Reaction , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sequence Analysis, DNA
13.
Exp Appl Acarol ; 28(1-4): 9-25, 2002.
Article in English | MEDLINE | ID: mdl-14570114

ABSTRACT

The tick-borne cattle pathogen Anaplasma marginale (Rickettsiales: Anaplasmataceae) multiplies within membrane-bound inclusions in host cell cytoplasm. Many geographic isolates of A. marginale occur that vary in genotype, antigenic composition, morphology and infectivity for ticks. A tick cell culture system for propagation of A. marginale proved to be a good model for study of tick-pathogen interactions. Six major surface proteins (MSPs) identified on A. marginale from bovine erythrocytes were conserved on A. marginale derived from tick cells. MSP1a and MSP1b were adhesins for bovine erythrocytes, while only MSP1a was bound to be an adhesin for tick cells. The tandemly repeated portion of MSP1a was found to be necessary and sufficient for adhesion to both tick cells and bovine erythrocytes. Infectivity of A. marginale isolates for ticks was dependent on the adhesive capacity of the isolate MSP1a, which was found to involve both the adhesive properties and sequence of the repeated peptides. Cattle immunized with A. marginale derived from bovine erythrocytes or tick cells demonstrated a differential antibody response to MSP1a and MSP1b that resulted from the differential expression of these proteins in cattle and ticks cells. MSP2, derived from a multigene family, was found to undergo antigenic variation in cattle and ticks and may contribute to establishment of persistent A. marginale infections. MSP1a has been used as a stable genetic marker for geographic isolates because the molecular weight varies due to differing numbers of the tandem repeats. However, phylogenetic studies of A. marginale isolates from North America using MSP1a and MSP4 demonstrated that MSP4 was a good biogeographic marker, while MSP1a varied greatly among and within geographic areas. Infection and development of A. marginale in cattle and tick cells appears to differ and to be mediated by several surface proteins encoded from the small genome.


Subject(s)
Anaplasma marginale/physiology , Anaplasmosis/microbiology , Cattle Diseases/microbiology , Ticks/microbiology , Anaplasma marginale/genetics , Anaplasma marginale/metabolism , Anaplasmosis/transmission , Animals , Bacterial Adhesion/physiology , Bacterial Outer Membrane Proteins/metabolism , Cattle , Cattle Diseases/transmission , Phylogeny , Ticks/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...