首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
Anaplasma marginale, an ehrlichial pathogen of cattle and wild ruminants, is transmitted biologically by ticks. A developmental cycle of A. marginale occurs in a tick that begins in gut cells followed by infection of salivary glands, which are the site of transmission to cattle. Geographic isolates of A. marginale vary in their ability to be transmitted by ticks. In these experiments we studied transmission of two recent field isolates of A. marginale, an Oklahoma isolate from Wetumka, OK, and a Florida isolate from Okeechobee, FL, by two populations of Dermacentor variabilis males obtained from the same regions. The Florida and Oklahoma tick populations transmitted the Oklahoma isolate, while both tick populations failed to transmit the Florida isolate. Gut and salivary gland infections of A. marginale, as determined by quantitative PCR and microscopy, were detected in ticks exposed to the Oklahoma isolate, while these tissues were not infected in ticks exposed to the Florida isolate. An adhesion-recovery assay was used to study adhesion of the A. marginale major surface protein (MSP) 1a to gut cells from both tick populations and cultured tick cells. We demonstrated that recombinant Escherichia coli expressing Oklahoma MSP1a adhered to cultured and native D. variabilis gut cells, while recombinant E. coli expressing the Florida MSP1a were not adherent to either tick cell population. The MSP1a of the Florida isolate of A. marginale, therefore, was unable to mediate attachment to tick gut cells, thus inhibiting salivary gland infection and transmission to cattle. This is the first report of MSP1a being responsible for effecting infection and transmission of A. marginale by Dermacentor spp. ticks. The mechanism of tick infection and transmission of A. marginale is important in formulating control strategies and development of improved vaccines for anaplasmosis.  相似文献   

2.
Anaplasma marginale subsp. centrale is a naturally attenuated subtype that has been used as a vaccine for a century. We sequenced the genome of this organism and compared it to those of virulent senso stricto A. marginale strains. The comparison markedly narrows the number of outer membrane protein candidates for development of a safer inactivated vaccine and provides insight into the diversity among strains of senso lato A. marginale.Sir Arnold Theiler described Anaplasma marginale as the “cause of a specific tick-borne disease of cattle” in 1908 (14), providing the first identification of a rickettsial pathogen. Two years later, Theiler isolated a less virulent organism, which he designated A. marginale subtype centrale (15). This naturally attenuated strain has been used as a live vaccine to prevent severe disease due to A. marginale senso stricto strains for 100 years. Understanding the genetic similarities and differences between the vaccine strain and wild-type A. marginale strains will provide clues as to how the vaccine provides protection. To that end, we have sequenced the A. marginale subsp. centrale vaccine strain using a whole-genome shotgun sequencing strategy.Genomic DNA, obtained from Kimron Veterinary institute, was fragmented by hydroshearing and ligated into pSmartLCKan (Lucigen). A total of 10,752 paired-end sequence reads (∼6.5× coverage) were generated. Assembly with Phrap (www.phrap.org) resulted in 148 contigs. Closure was achieved by applying the genome walking method across gap-spanning subclones and genomic DNA amplicons. For polymorphic loci, the most frequently observed subclone sequence was selected.Coding sequences (CDSs) in the single, circular, 1,206,806-bp chromosome were predicted using Glimmer2 and Glimmer3 (4, 5, 12). Annotation was as described previously for A. marginale senso stricto genomes (2, 3). There are 925 predicted CDSs, 19 pseudogenes, 37 tRNA genes, and a single set of rRNA genes in the genome. A. marginale subsp. centrale contains 10 putative genes not found in the closed-core genomes of senso stricto strains (3). Similarly, 18 genes found in senso stricto strains are absent from A. marginale subsp. centrale. This divergence is consistent with the subspecies nomenclature (15), but the findings do not resolve whether these genetic differences warrant classification of the vaccine strain as a distinct species within the genus Anaplasma (6).The ability of live A. marginale subsp. centrale to protect against a diversity of A. marginale strains indicates that epitopes critical for protective immunity are broadly conserved (11). As immunity against A. marginale can be induced by immunization with purified outer membrane protein (OMP) complexes (8-10, 13), identification of OMPs conserved between A. marginale subsp. centrale and senso stricto A. marginale may narrow the vaccine candidate list. A. marginale OMPs cluster predominately into two protein superfamilies, major surface protein 1 (Msp1) and Pfam01617/Msp2 (2). Members of the Msp1 superfamily from senso stricto strains (1, 2) are not well conserved (e.g., Msp1a, Msp1b-1, Msp1b-2, and Mlp2 to Mlp4; 13 to 48% amino acid identity) or are nonexistent (e.g., the products of Msp1b partial genes 1 to 3) in A. marginale subsp. centrale, suggesting that immunity induced by the live vaccine strain is unlikely to be associated with the Msp1 superfamily.Comparative analysis of the Pfam01617/Msp2 superfamily (2, 8) reveals both conservation and diversity. OpAG1 to OpAG3 and Msp4 are generally well conserved, while the family comprising Omp1 to Omp15 found in senso stricto strains (2, 3, 8) is reduced in A. marginale subsp. centrale: genes for the closely related proteins Omp7 to Omp9 are collapsed into a single CDS, and genes for homologs of Omp2, Omp3, Omp6, and Omp15 are missing. The OMP complex capable of inducing protective immunity contains 11 proteins (7, 8). By excluding those without homologs in the vaccine strain and the highly variable Msp2 and Msp3, the number of candidates is narrowed to six: four Msp2 superfamily members (Msp4, Omp1, Omp7, and OpAG2) and two non-superfamily members (AM779/ACIS557 and AM854/ACIS486). The degree of identity among these candidates from the vaccine strain and senso stricto A. marginale strains ranges from 63% (for OpAG2 proteins) to 88% (for Msp4 homologs). While the next steps in vaccine development will require strain analysis for epitope conservation in these candidates and immunization trials to test in vivo efficacy, progress will be accelerated using the minimal candidate list defined by the comparative genomics approach.  相似文献   

3.
The sequencing of the complete genome of Anaplasma marginale has enabled the identification of several genes that encode membrane proteins, thereby increasing the chances of identifying candidate immunogens. Little is known regarding the genetic variability of genes that encode membrane proteins in A. marginale isolates. The aim of the present study was to determine the degree of conservation of the predicted amino acid sequences of OMP1, OMP4, OMP5, OMP7, OMP8, OMP10, OMP14, OMP15, SODb, OPAG1, OPAG3, VirB3, VirB9-1, PepA, EF-Tu and AM854 proteins in a Brazilian isolate of A. marginale compared to other isolates. Hence, primers were used to amplify these genes: omp1, omp4, omp5, omp7, omp8, omp10, omp14, omp15, sodb, opag1, opag3, virb3, VirB9-1, pepA, ef-tu and am854. After polimerase chain reaction amplification, the products were cloned and sequenced using the Sanger method and the predicted amino acid sequence were multi-aligned using the CLUSTALW and MEGA 4 programs, comparing the predicted sequences between the Brazilian, Saint Maries, Florida and A. marginale centrale isolates. With the exception of outer membrane protein (OMP) 7, all proteins exhibited 92-100% homology to the other A. marginale isolates. However, only OMP1, OMP5, EF-Tu, VirB3, SODb and VirB9-1 were selected as potential immunogens capable of promoting cross-protection between isolates due to the high degree of homology (over 72%) also found with A. (centrale) marginale.  相似文献   

4.
5.
Anaplasma marginale is the causative agent of anaplasmosis in cattle. Transposon mutagenesis of this pathogen using the Himar1 system resulted in the isolation of an omp10 operon insertional mutant referred to as the omp10::himar1 mutant. The work presented here evaluated if this mutant had morphological and/or growth rate defects compared to wild-type A. marginale. Results showed that the morphology, developmental cycle, and growth in tick and mammalian cell cultures are similar for the mutant and the wild type. Tick transmission experiments established that tick infection levels with the mutant were similar to those with wild-type A. marginale and that infected ticks successfully infected cattle. However, this mutant exhibited reduced infectivity and growth in cattle. The possibility of transforming A. marginale by transposon mutagenesis coupled with in vitro and in vivo assessment of altered phenotypes can aid in the identification of genes associated with virulence. The isolation of deliberately attenuated organisms that can be evaluated in their natural biological system is an important advance for the rational design of vaccines against this species.  相似文献   

6.
Anaplasmosis, a persistent intraerythrocytic infection of cattle by Anaplasma marginale, causes severe anemia and a higher rate of abortion, resulting in significant loss to both dairy and beef industries. Clinical diagnosis is based on symptoms and confirmatory laboratory tests are required. Currently, all the diagnostic assays have been developed with whole antigens with indirect ELISA based on multiple epitopes. In a pioneer investigation we demonstrated the use of critical motifs of an epitope as biomarkers for immunosensor applications. Mimotopes of the MSP1a protein functional epitope were obtained through Phage Display after three cycles of selection of a 12-mer random peptide library against the neutralizing monoclonal antibody 15D2. Thirty-nine clones were randomly selected, sequenced, translated and aligned with the native sequence. The consensus sequence SxSSQSEASTSSQLGA was obtained, which is located in C-terminal end of the 28-aa repetitive motif of the MSP1a protein, but the alignment and sequences' variation among mimotopes allowed us to map the critical motif STSSxL within the consensus sequence. Based on these results, two peptides were chemically synthesized: one based on the critical motif (STSSQL, Am1) and the other based on the consensus sequence aligned with the native epitope (SEASTSSQLGA, Am2). Sera from 24 infected and 52 healthy animals were tested by ELISA for reactivity against Am1 and Am2, which presented sensitivities of 96% and 100%, respectively. The Am1 peptide was incorporated onto a biolectrode (graphite modified with poly-3-hydroxyphenylacetic acid) and direct serum detection was demonstrated by impedance, differential pulse voltammetry, and atomic force microscopy. The electrochemical sensor system proved to be highly effective in discriminating sera from positive and negative animals. These immunosensors were highly sensitive and selective for positive IgG, contaminants did not affect measurements, and were based on a simple, fast and reproducible electrochemical system.  相似文献   

7.
The infectivity of Anaplasma marginale was maintained in liquid nitrogen storage throughout a 4-yr test period.  相似文献   

8.
The protective major surface protein 1 (MSP1) complex of Anaplasma marginale is a heteromer of MSP1a and MSP1b, encoded by a multigene family. The msp1beta sequences were highly conserved throughout infection. However, liquid chromatography-tandem mass spectrometry analysis identified only a single MSP1b protein, MSP1b1, within the MSP1 complex.  相似文献   

9.
Bacterial pathogens in the genus Anaplasma generate surface coat variants by gene conversion of chromosomal pseudogenes into single-expression sites. These pseudogenes encode unique surface-exposed hypervariable regions flanked by conserved domains, which are identical to the expression site flanking domains. In addition, Anaplasma marginale generates variants by recombination of oligonucleotide segments derived from the pseudogenes into the existing expression site copy, resulting in a combinatorial increase in variant diversity. Using the A. marginale genome sequence to track the origin of sequences recombined into the msp2 expression site, we demonstrated that the complexity of the expressed msp2 increases during infection, reflecting a shift from recombination of the complete hypervariable region of a given pseudogene to complex mosaics with segments derived from hypervariable regions of different pseudogenes. Examination of the complete set of 1183 variants with segmental changes revealed that 99% could be explained by one of the recombination sites occurring in the conserved flanking domains and the other within the hypervariable region. Consequently, we propose an 'anchoring' model for segmental gene conversion whereby the conserved flanking sequences tightly align and anchor the expression site sequence to the pseudogene. Associated with the recombination sites were deletions, insertions and substitutions; however, these are a relatively minor contribution to variant generation as these occurred in less than 2% of the variants. Importantly, the anchoring model, which can account for more variants than a strict segmental sequence identity mechanism, is consistent with the number of msp2 variants predicted and empirically identified during persistent infection.  相似文献   

10.
Indirect enzyme-linked immunosorbent assays (ELISAs) based on recombinant MSP1a and MSP2 from a Brazilian isolate of Anaplasma marginale were developed to detect antibodies against this rickettsia in cattle. The high sensitivities (99% for both tests) and specificities (100% for both tests) were confirmed with sera from cattle positive or negative for A. marginale antibodies, respectively, by immunofluorescent antibody test. By the analysis of 583 sera from cattle of three regions of the state of Pernambuco, Brazil, the agreement between both tests was high, with a kappa index of 0.89. The similar performances of the ELISAs suggest that both tests can be used in epidemiological surveys for detection of antibodies to A. marginale in cattle.  相似文献   

11.
12.
The coevolution of ticks and the pathogens that they transmit has ensured their mutual survival. In these studies, we used a functional genomics approach to characterize tick genes regulated in response to Anaplasma marginale infection. Differentially regulated genes/proteins were identified by suppression-subtractive hybridization and differential in-gel electrophoresis analyses of cultured IDE8 tick cells infected with A. marginale. Nine of 17 of these genes were confirmed by real-time RT-PCR to be differentially regulated in ticks and/or IDE8 tick cells in response to A. marginale infection. RNA interference was used for functional studies. Six genes, which encode putative selenoprotein W2a, hematopoietic stem/progenitor cells protein-like, proteasome 26S subunit, ferritin, GST, and subolesin control, were found to affect A. marginale infection in IDE8 tick cells. Four genes, which encode putative GST, salivary selenoprotein M, vATPase, and ubiquitin, affected A. marginale infection in different sites of development in ticks. The results of these studies demonstrated that a molecular mechanism occurs by which tick cell gene expression mediates the A. marginale developmental cycle and trafficking through ticks.  相似文献   

13.
Anaplasma marginale is a tick-borne rickettsial pathogen of cattle with a worldwide distribution. Currently a safe and efficacious vaccine is unavailable. Outer membrane protein (OMP) extracts or a defined surface protein complex reproducibly induce protective immunity. However, there are several knowledge gaps limiting progress in vaccine development. First, are these OMPs conserved among the diversity of A. marginale strains circulating in endemic regions? Second, are the most highly conserved outer membrane proteins in the immunogens recognized by immunized and protected animals? Lastly, can this subset of OMPs recognized by antibody from protected vaccinates and conserved among strains recapitulate the protection of outer membrane vaccines? To address the first goal, genes encoding OMPs AM202, AM368, AM854, AM936, AM1041, and AM1096, major subdominant components of the outer membrane, were cloned and sequenced from geographically diverse strains and isolates. AM202, AM936, AM854, and AM1096 share 99.9 to 100% amino acid identity. AM1041 has 97.1 to 100% and AM368 has 98.3 to 99.9% amino acid identity. While all four of the most highly conserved OMPs were recognized by IgG from animals immunized with outer membranes, linked surface protein complexes, or unlinked surface protein complexes and shown to be protected from challenge, the highest titers and consistent recognition among vaccinates were to AM854 and AM936. Consequently, animals were immunized with recombinant AM854 and AM936 and challenged. Recombinant vaccinates and purified outer membrane vaccinates had similar IgG and IgG2 responses to both proteins. However, the recombinant vaccinates developed higher bacteremia after challenge as compared to adjuvant-only controls and outer membrane vaccinates. These results provide the first evidence that vaccination with specific antigens may exacerbate disease. Progressing from the protective capacity of outer membrane formulations to recombinant vaccines requires testing of additional antigens, optimization of the vaccine formulation and a better understanding of the protective immune response.  相似文献   

14.
Mess1是新近鉴定的 STE2 0家族的蛋白激酶 .对 Mess1的基因表达和蛋白功能进行研究 ,发现其 m RNA在鼠组织中广泛分布 ,但在不同细胞系中表达显著不同 ;结构分析表明 ,Mess1蛋白N端是保守的 STE2 0样激酶催化区 ,C端是高度亲水的酸性调节区 ,包含多个潜在的丝氨酸 /苏氨酸磷酸化调节位点 .哺乳动物细胞表达的 Mess1对 MBP显示出激酶活性 ,并发生自主磷酸化 .Mess1可被砷酸盐应激激活 ,但丝裂原 EGF刺激无活化效应 .表明 Mess1可能在蛋白磷酸化的早期过程中发挥作用 ,介导细胞对严重应激刺激引起的特异性反应 .  相似文献   

15.
Mess1是新近鉴定的 STE2 0家族的蛋白激酶 .对 Mess1的基因表达和蛋白功能进行研究 ,发现其 m RNA在鼠组织中广泛分布 ,但在不同细胞系中表达显著不同 ;结构分析表明 ,Mess1蛋白N端是保守的 STE2 0样激酶催化区 ,C端是高度亲水的酸性调节区 ,包含多个潜在的丝氨酸 /苏氨酸磷酸化调节位点 .哺乳动物细胞表达的 Mess1对 MBP显示出激酶活性 ,并发生自主磷酸化 .Mess1可被砷酸盐应激激活 ,但丝裂原 EGF刺激无活化效应 .表明 Mess1可能在蛋白磷酸化的早期过程中发挥作用 ,介导细胞对严重应激刺激引起的特异性反应 .  相似文献   

16.
SIRT1 is a NAD+-dependent deacetylase that plays important roles in many cellular processes. SIRT1 activity is uniquely controlled by a C-terminal regulatory segment (CTR). Here we present crystal structures of the catalytic domain of human SIRT1 in complex with the CTR in an open apo form and a closed conformation in complex with a cofactor and a pseudo-substrate peptide. The catalytic domain adopts the canonical sirtuin fold. The CTR forms a β hairpin structure that complements the β sheet of the NAD+-binding domain, covering an essentially invariant hydrophobic surface. The apo form adopts a distinct open conformation, in which the smaller subdomain of SIRT1 undergoes a rotation with respect to the larger NAD+-binding subdomain. A biochemical analysis identifies key residues in the active site, an inhibitory role for the CTR, and distinct structural features of the CTR that mediate binding and inhibition of the SIRT1 catalytic domain.  相似文献   

17.

Background

Surfactant protein B (SP-B; 79 residues) belongs to the saposin protein superfamily, and plays functional roles in lung surfactant. The disulfide cross-linked, N- and C-terminal domains of SP-B have been theoretically predicted to fold as charged, amphipathic helices, suggesting their participation in surfactant activities. Earlier structural studies with Mini-B, a disulfide-linked construct based on the N- and C-terminal regions of SP-B (i.e., ∼residues 8–25 and 63–78), confirmed that these neighboring domains are helical; moreover, Mini-B retains critical in vitro and in vivo surfactant functions of the native protein. Here, we perform similar analyses on a Super Mini-B construct that has native SP-B residues (1–7) attached to the N-terminus of Mini-B, to test whether the N-terminal sequence is also involved in surfactant activity.

Methodology/Results

FTIR spectra of Mini-B and Super Mini-B in either lipids or lipid-mimics indicated that these peptides share similar conformations, with primary α-helix and secondary β-sheet and loop-turns. Gel electrophoresis demonstrated that Super Mini-B was dimeric in SDS detergent-polyacrylamide, while Mini-B was monomeric. Surface plasmon resonance (SPR), predictive aggregation algorithms, and molecular dynamics (MD) and docking simulations further suggested a preliminary model for dimeric Super Mini-B, in which monomers self-associate to form a dimer peptide with a “saposin-like” fold. Similar to native SP-B, both Mini-B and Super Mini-B exhibit in vitro activity with spread films showing near-zero minimum surface tension during cycling using captive bubble surfactometry. In vivo, Super Mini-B demonstrates oxygenation and dynamic compliance that are greater than Mini-B and compare favorably to full-length SP-B.

Conclusion

Super Mini-B shows enhanced surfactant activity, probably due to the self-assembly of monomer peptide into dimer Super Mini-B that mimics the functions and putative structure of native SP-B.  相似文献   

18.
19.
20.
Purified immunoglobulin from each of two hybridoma cell lines (ANA 15D2 and ANA 22B1) significantly neutralized the infectivity of 108Anaplasma marginale initial bodies for cattle. Both cell lines produce antibody to the same Am 105 epitope as they inhibited the binding of each other to Am 105 in a competition radioimmunoassay. Complete digestion of Am 105 with proteinase K, pronase, or trypsin prevented monoclonal antibody binding indicating that the epitope was protein in nature rather than surface polysaccharide. In addition, evidence that the neutralization-sensitive epitope was not membrane-protein-bound polysaccharide included: [1] 35S-methionine, but not 3H-glucosamine, was metabolically incorporated into Am 105 during short-term in vitro culture; [2] Am 105 was surface radiolabeled using 125I in a lactoperoxidase mediated reaction, but not labeled using a galactose oxidase-NaB[3H]4 mediated reaction with or without neuraminidase pretreatment; and [3] Am 105 did not bind to concanavalin A, Helix pomatia lectin, peanut lectin, soybean lectin, or wheat germ lectin.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号