戻る
「早戻しボタン」を押すと検索画面に戻ります。

今後説明を表示しない

[OK]

コーパス検索結果 (1語後でソート)

通し番号をクリックするとPubMedの該当ページを表示します
1 k of the tick transmitted pathogen Anaplasma marginale.
2  at the protein level) was higher than in A. marginale.
3 e outer membrane of the rickettsia Anaplasma marginale.
4  90-kb region of the St. Maries strain of A. marginale.
5 and transmitted the Puerto Rico strain of A. marginale.
6 ion strains of the cattle pathogen Anaplasma marginale.
7                  Maries (Idaho) strain of A. marginale.
8 ly and phenotypically distinct strains of A. marginale.
9 wide economic importance caused by Anaplasma marginale.
10 entia tsutsugamushi, Wolbachia and Anaplasma marginale.
11 ain (Florida) and heterologous strains of A. marginale.
12 ce variants in bloodstream populations of A. marginale.
13 s infected with the South Idaho strain of A. marginale.
14 nes encoding MSP1b in a Florida strain of A. marginale.
15  calves following recall stimulation with A. marginale.
16  genes encoding major surface proteins of A. marginale.
17  would augment type 1 recall responses to A. marginale.
18 d antigens are conserved among strains of A. marginale.
19  outer membranes of the Florida strain of A. marginale.
20 omologous to the MSP-2 proteins of Anaplasma marginale.
21 ized all bovine blood-passaged strains of A. marginale.
22  species but distinct from that of Anaplasma marginale.
23 y important antigenic surface proteins of A. marginale.
24 cally polymorphic among strains of Anaplasma marginale.
25  ewingii reacted with the MSP3 antigen of A. marginale.
26 sial agents Anaplasma phagocytophilum and A. marginale.
27 sting as a linked protein vaccine against A. marginale.
28 unized with the protective OM fraction of A. marginale.
29 the diversity among strains of senso lato A. marginale.
30 of cultured IDE8 tick cells infected with A. marginale.
31 pp. (79 to 83% similarity) than to Anaplasma marginale (72 to 75% similarity).
32 e examined the strain structure of Anaplasma marginale, a genogroup II ehrlichial pathogen, in both a
33 pulation immunity develops against Anaplasma marginale, a highly antigenically variant rickettsial pa
34 nant surface proteins, we examined Anaplasma marginale, a rickettsia with two highly immunogenic oute
35                                    Anaplasma marginale, a rickettsial pathogen, evades clearance in t
36 gated this by using infection with Anaplasma marginale, a ruminant pathogen that replicates to levels
37                                    Anaplasma marginale, a tick-borne rickettsial pathogen of cattle,
38 his study, the surface proteome of Anaplasma marginale, a tick-transmitted bacterial pathogen, was ta
39 genotypically and phenotypically distinct A. marginale, A. ovis, and A. centrale strains.
40     These data suggest that, similarly to A. marginale, A. phagocytophilum uses combinatorial mechani
41 tor variabilis persistently infected with A. marginale after feeding successively on one susceptible
42                                    Anaplasma marginale, an intraerythrocytic ehrlichial pathogen of c
43 ginale strains, as has been reported with A. marginale and A. marginale subsp. centrale strains.
44 -time PCR assays to simultaneously detect A. marginale and A. marginale subsp. centrale.
45 PCR (qRT-PCR) assays for the detection of A. marginale and A. phagocytophilum 16S rRNA in plasma-free
46                     The promoters in both A. marginale and A. phagocytophilum have similar structure
47  as few as 100 copies of 16S rRNA of both A. marginale and A. phagocytophilum in the same reaction.
48 for the accurate and precise detection of A. marginale and A. phagocytophilum infections in cattle.
49 rface protein 2 (MSP2) and MSP3 of Anaplasma marginale and Anaplasma ovis, Anaplasma phagocytophilum
50 dominant outer membrane protein of Anaplasma marginale and Anaplasma phagocytophilum pathogens that c
51 e protein expression sites in both Anaplasma marginale and Anaplasma phagocytophilum.
52  mRNA transcript in erythrocyte stages of A. marginale and defined the structure of the genomic expre
53 ed the major surface protein 2 (MSP-2) in A. marginale and HGE and OMP-1 in E. chaffeensis.
54 s superinfected with different strains of A. marginale and hypothesized that the msp2 pseudogene repe
55  proliferated specifically in response to A. marginale and produced high titers of gamma interferon.
56 ed for vaccine-induced protection against A. marginale and provides clear direction for development o
57                                           A. marginale and related ehrlichiae express immunoprotectiv
58            Thus, an effective vaccine for A. marginale and related organisms should contain both T- a
59 oteins in development of vaccines against A. marginale and related pathogens.
60 tence in the chronic infections caused by A. marginale and related rickettsiae.
61 servoirs harbor genetically heterogeneous A. marginale and suggest that different genotypes are maint
62 s (MSPs) between the cell culture-derived A. marginale and the bovine erythrocytic stage, currently t
63 el obligate intracellular pathogen Anaplasma marginale and the human pathogen Anaplasma phagocytophil
64  to the intraerythrocytic agent of Anaplasma marginale and the intragranulocytic agent of Anaplasma p
65  of epitopes common to several strains of A. marginale and the related pathogen A. ovis.
66 t conserved between different isolates of A. marginale, and at least in the isolate from Florida, wha
67 ehrlichiosis, msp-2 and msp-4 from Anaplasma marginale, and map-1 from Cowdria ruminantium.
68 on does not apply to two other strains of A. marginale, and that different variants are also expresse
69                                 All three A. marginale Anks were confirmed to be expressed during int
70 ythrocytic stage, currently the source of A. marginale antigen, was determined.
71 which we hypothesize reflect emergence of A. marginale antigenic variants.
72 per (Th) cell response to these and other A. marginale antigens and to determine conservation of Th c
73 sistent bovine ehrlichial pathogen Anaplasma marginale are immunodominant proteins that undergo antig
74  data show that MSPs of erythrocyte-stage A. marginale are present on culture stages and may be struc
75          Major surface proteins of Anaplasma marginale are vaccine candidates.
76  The suppressed response was specific for A. marginale, as responses to Clostridium vaccine Ag were c
77           The rickettsial pathogen Anaplasma marginale assembles an actin filament bundle during intr
78 imit is the minimum infective unit of one A. marginale bacterium.
79 nce variants did not change on passage of A. marginale between culture, acute erythrocyte stage infec
80  unrelated vector-borne pathogens, Anaplasma marginale, Borrelia hermsii and Trypanosoma brucei, illu
81                                    Anaplasma marginale causes bovine anaplasmosis, a debilitating and
82 e tick-borne rickettsial organism, Anaplasma marginale, causes a disease in cattle of world-wide econ
83 ly immunized with outer membranes against A. marginale challenge and identify three MSPs that are rec
84 nst high-level bacteremia and anemia upon A. marginale challenge of cattle and effectively recapitula
85  induction of protective immunity against A. marginale challenge.
86 -3(+) exhausted T cells were monitored in A. marginale-challenged cattle previously immunized with OM
87                                    Anaplasma marginale colonies differed in their development and app
88  major surface protein 1 (MSP1) of Anaplasma marginale, composed of covalently associated MSP1a and M
89 SP1b) of the erythrocytic stage of Anaplasma marginale conferred protection against homologous challe
90 he cell-type (tick or mammalian) in which A. marginale developed.
91 ch tick cell gene expression mediates the A. marginale developmental cycle and trafficking through ti
92 rains of the tick-borne rickettsia Anaplasma marginale differ markedly in transmission efficiency, wi
93 scovered that several surface proteins of A. marginale encode polymorphic multigene families.
94                                           A. marginale encodes three proteins containing ankyrin moti
95                                           A. marginale establishes lifelong persistence in infected r
96           The rickettsial pathogen Anaplasma marginale establishes lifelong persistent infection in t
97                                    Anaplasma marginale establishes persistent infection characterized
98 cytophila, and the bovine pathogen Anaplasma marginale, express a markedly immunodominant outer membr
99           The rickettsial pathogen Anaplasma marginale expresses a variable immunodominant outer memb
100                   Under immune selection, A. marginale expresses complex major surface protein 2 mosa
101           Within each rickettsemic cycle, A. marginale expressing antigenically variant major surface
102 esis that challenge of cattle with Anaplasma marginale expressing MSP2 variants to which the animals
103 o most immunodominant surface proteins of A. marginale followed by emergence of unique variants indic
104 opes conserved among different strains of A. marginale for inclusion in a nucleic acid or recombinant
105     Immunization of cattle with an Anaplasma marginale fraction enriched in outer membranes (OM) can
106 nst erythrocyte stages, also reacted with A. marginale from cell culture and tick salivary glands.
107                               Isolates of A. marginale from the southern United States (Florida, Miss
108 zed and used to amplify msp1 sequences of A. marginale from tick cell cultures, from cattle during ac
109   The results support the hypothesis that A. marginale gene expression is regulated by the specific h
110                Within the mammalian host, A. marginale generates antigenic variants by changing a sur
111                       In addition, Anaplasma marginale generates variants by recombination of oligonu
112 tigenic proteins, 14 are annotated in the A. marginale genome and include type IV secretion system pr
113    A key question is how the small 1.2 Mb A. marginale genome generates sufficient variants to allow
114      Recently, complete sequencing of the A. marginale genome has identified an expanded set of genes
115                  A database search of the A. marginale genome identified 24 antigenic proteins that w
116   Recent sequencing and annotation of the A. marginale genome predicts at least 62 outer membrane pro
117                                 Using the A. marginale genome sequence to track the origin of sequenc
118  not a marker for the characterization of A. marginale geographic isolates and suggest that the genet
119 several other bacterial pathogens, Anaplasma marginale has an outer membrane that induces complete pr
120 ty against the ehrlichial pathogen Anaplasma marginale has been hypothesized to require induction of
121 ilum, MSP2(P44), is homologous to MSP2 of A. marginale, has a similar organization of conserved and v
122 zation, proliferated specifically against A. marginale homogenate and purified MSP1 in a dose-depende
123 erated strongly in response to both whole A. marginale homogenates and purified outer membranes, and
124                                    Anaplasma marginale illustrates this transition: in the mammalian
125  as that found on the Virginia isolate of A. marginale in bovine erythrocytes and tick salivary gland
126                            Development of A. marginale in cell culture was morphologically similar to
127 transmissible St. Maries strain of Anaplasma marginale in Dermacentor andersoni as a positive control
128 sion that allows long-term persistence of A. marginale in the mammalian reservoir.
129 ination, selection for sub-populations of A. marginale in the vertebrate host and/or PCR errors.
130 portion of erythrocytes containing viable A. marginale in vitro, indicating that an antibody-independ
131  (MSP1) of the ehrlichial pathogen Anaplasma marginale induces protective immunity in calves challeng
132  coli were also shown to be functional in A. marginale infected cells, as determined by quantificatio
133 als, similar to the pattern described for A. marginale-infected cattle, while in the second pattern,
134 hese superfamily genes are transcribed in A. marginale-infected erythrocytes, tick midgut and salivar
135  expression site is utilized in stages of A. marginale infecting tick salivary glands.
136 rickettsemia that characterize persistent A. marginale infection and control of each rickettsemic cyc
137 s induces protection against acute Anaplasma marginale infection and disease, and a proteomic and gen
138  during cyclic rickettsemia in persistent A. marginale infection and suggest that emergent variants p
139 onse is central for the control of Anaplasma marginale infection in cattle.
140 otein M, vATPase, and ubiquitin, affected A. marginale infection in different sites of development in
141 d subolesin control, were found to affect A. marginale infection in IDE8 tick cells.
142                                Persistent A. marginale infection is characterized by repetitive ricke
143 loss of memory T cell responses following A. marginale infection is due to a mechanism other than ind
144 itive samples from tropical regions where A. marginale infection is endemic identified individual inf
145 mOmpA and ApOmpA competitively antagonize A. marginale infection of host cells, but a monoclonal anti
146  or its predicted binding domain inhibits A. marginale infection of host cells.
147 nsignificant during subsequent persistent A. marginale infection up to 1 year.
148 vity and specificity for the detection of A. marginale infection were found to be 65.2% (95% CI, 55.3
149                                    During A. marginale infection, dynamic and extensive amino acid se
150             Antibody was induced early in A. marginale infection, predominately against the surface-e
151                         Throughout Anaplasma marginale infection, recombination results in the sequen
152 e(x) fails to inhibit AmOmpA adhesion and A. marginale infection.
153 ick genes regulated in response to Anaplasma marginale infection.
154 cks and/or IDE8 tick cells in response to A. marginale infection.
155  persistently infected with one strain of A. marginale, infection with a second strain (superinfectio
156 ogens that generate actin filament tails, A. marginale infects mature erythrocytes, and the F-actin a
157 ter percentage of infected ticks secreted A. marginale into the saliva and did so at a significantly
158 urface protein 1 (MSP1) complex of Anaplasma marginale is a heteromer of MSP1a and MSP1b, encoded by
159           The St. Maries strain of Anaplasma marginale is a high-transmission-efficiency strain that
160                                    Anaplasma marginale is a tick-borne pathogen, one of several close
161                                    Anaplasma marginale is a tick-transmitted pathogen of cattle close
162                                    Anaplasma marginale is an ehrlichial pathogen of cattle that estab
163                                    Anaplasma marginale is an ehrlichial pathogen of cattle, in the or
164                                    Anaplasma marginale is an intraerythrocytic rickettsial pathogen o
165                                   Because A. marginale is an obligate intracellular organism, its adh
166 aerythrocytic rickettsial pathogen Anaplasma marginale is endemic in South Africa.
167  cattle from east-central Oklahoma, where A. marginale is endemic.
168 cted reservoir herd within a region where A. marginale is endemic.
169  major surface protein 2 (MSP2) of Anaplasma marginale is expressed from a 3.5-kb operon that contain
170                                           A. marginale is present in the tick salivary glands before
171                     The rickettsia Anaplasma marginale is the most prevalent tick-borne livestock pat
172 (Msp2) of the tick-borne pathogen, Anaplasma marginale, is thought to be involved in antigenic variat
173                  The surface complexes of A. marginale isolated from erythrocytes of the mammalian ho
174      In contrast, the surface proteome of A. marginale isolated from tick cells was much less complex
175                                    All 11 A. marginale isolates collected from Oklahoma had different
176                    MSP1a among geographic A. marginale isolates is variable in size because of differ
177            Thus, infection of cattle with A. marginale leads to the rapid loss of Ag-specific T cells
178                  The cell culture-derived A. marginale maintained the same-size MSP1a as that found o
179 he CD4(+) T lymphocyte response to Anaplasma marginale major surface protein 1a (MSP1a).
180                           Specific Anaplasma marginale major surface protein 2 (MSP2) variants are se
181  in this species, the homologue of Anaplasma marginale major surface protein 4 gene (msp4) was identi
182                               To identify A. marginale molecules associated with these filaments, two
183 y, with sequence similarity to the Anaplasma marginale msp-2 genes.
184  the Cowdria ruminantium MAP-1 and Anaplasma marginale MSP-4 proteins.
185 ed strong CD4(+) T cell responses against A. marginale, MSP1a, and specific MHC class II DR-restricte
186 w that the single hypervariable region of A. marginale MSP2 encodes epitopes that are immunogenic and
187   Interestingly, the 5' structure of this A. marginale msp2 locus is conserved in the omp1 gene locus
188  that the major outer membrane protein of A. marginale, MSP2, is encoded on a polycistronic mRNA.
189                                       The A. marginale msp3 gene msp3-12 was cloned and expressed in
190                                           A. marginale numbers per tick increase gradually in salivar
191  the genetic variations among isolates of A. marginale obtained during 2001 from infected cattle from
192 strains of the tick-borne pathogen Anaplasma marginale occur and are transmitted within regions where
193               Many geographic isolates of A. marginale occur in the United States and have been ident
194 ning ankyrin motifs would be expressed by A. marginale only in tick cells and would traffic to the in
195                                    Anaplasma marginale (order Rickettsiales, family Anaplasmataceae),
196 the msp2 mRNA and MSP2 protein levels per A. marginale organism increase only minimally and transient
197 -feeding ticks, and whether the number of A. marginale organisms per salivary gland is significantly
198 ce of physiologically relevant numbers of A. marginale organisms.
199 ute anaplasmosis and cultured with Anaplasma marginale organisms.
200                 Here, we demonstrate that A. marginale outer membrane protein A (AmOmpA; AM854) contr
201 c CD4(+) T cells in cattle immunized with A. marginale outer membrane proteins or purified outer memb
202  this study we demonstrate that in Anaplasma marginale outer membrane-vaccinated cattle, VirB9, VirB1
203 as naturally complexed OMPs in the Anaplasma marginale outer membrane.
204         Immunization with purified Anaplasma marginale outer membranes induces complete protection ag
205 SP2 antigenic variation as a mechanism of A. marginale persistence.
206            In the related organism Anaplasma marginale, persistence is associated with antigenic vari
207 l antibody and expression screening of an A. marginale phage library.
208 enically diverse and continually changing A. marginale population within the blood.
209        New MSP2 variants appeared in each A. marginale population, and sequence alignment of the MSP2
210 ferent MSP2 variants were encoded in each A. marginale population.
211 ire responses to an antigenically diverse A. marginale population.
212         In this study we analyzed MSP2 in A. marginale populations from the salivary glands of male D
213                                Testing of A. marginale-positive samples from tropical regions where A
214 ections of the tick-borne pathogen Anaplasma marginale predominate.
215    One-dimensional gel electrophoresis of A. marginale proteins demonstrated size polymorphism of MSP
216 ctrometry and tandem mass spectrometry of A. marginale proteins identified with an appendage-specific
217   Comparison between A. platys and Anaplasma marginale proteins showed sequence identities of 73.1% f
218 iased proteomic screen to identify Anaplasma marginale proteins specifically upregulated in the tick
219  nine pseudogenes from a single strain of A. marginale provides for a combinatorial number of possibl
220                                 In Anaplasma marginale pseudogenes for two antigenically variable gen
221  outer membranes of the Florida strain of A. marginale resulted in protective immunity that correlate
222 ein antigenic variation during persistent A. marginale rickettsemia, were identified in the A. ovis g
223 gainst severe disease upon challenge with A. marginale sensu stricto strains.
224 st, we tested whether the presence of two A. marginale (sensu lato) strains that differed in transmis
225 ls contributes to the rapid exhaustion of A. marginale-specific T cells following infection and that
226 ain (low transmission efficiency) and the A. marginale St. Maries strain (high transmission efficienc
227 ults indicate that a genetically distinct A. marginale strain capable of superinfecting the mammalian
228 0 of 75) were infected with only a single A. marginale strain, five animals each carried two strains
229 ttle with persistent infections of either A. marginale strain.
230 y, we tested the hypothesis that specific A. marginale strains are preferentially transmitted.
231  and OpAG3 were expressed by all examined A. marginale strains during the acute rickettsemia in the m
232                     In contrast, the same A. marginale strains expressed only OpAG2 in two different
233 that transmission of genomically distinct A. marginale strains predominates in high-prevalence areas
234 ked variation in the abilities of diverse A. marginale strains to assemble the F-actin appendages.
235                    We have identified two A. marginale strains with significant differences in the tr
236 Florida strain, four genetically distinct A. marginale strains, and Anaplasma ovis.
237 t superinfection does occur with distinct A. marginale strains, as has been reported with A. marginal
238 fic CD4+ T-cell clones responded to three A. marginale strains, confirming the VirB9-specific T-cell
239  cell epitopes among genetically distinct A. marginale strains, Th cell clones obtained prior to chal
240 acid sequences are highly conserved among A. marginale strains, with identities ranging from 95 to 99
241 s were identified in the genomes of three A. marginale strains.
242 red it to those of virulent senso stricto A. marginale strains.
243                              In contrast, A. marginale subsp. centrale (Israel vaccine strain) has an
244              Live vaccination with Anaplasma marginale subsp. centrale (synonym for Anaplasma central
245                              In contrast, A. marginale subsp. centrale colonized the midgut and then
246 were identified, which corresponded to 32 A. marginale subsp. centrale genotypes detected in cattle,
247  PCR results indicated high occurrence of A. marginale subsp. centrale infections, ranging from 25 to
248                                    Anaplasma marginale subsp. centrale is a naturally attenuated subt
249 ility of msp1aS as a genotypic marker for A. marginale subsp. centrale strain diversity.
250    Our results demonstrate a diversity of A. marginale subsp. centrale strains from cattle and wildli
251 s has been reported with A. marginale and A. marginale subsp. centrale strains.
252  on animals superinfected with the Anaplasma marginale subsp. centrale vaccine strain (low transmissi
253                              Nonetheless, A. marginale subsp. centrale was not transmitted, even when
254                                    Anaplasma marginale subsp. centrale was the first vaccine used to
255                      Samples positive for A. marginale subsp. centrale were further characterized usi
256 been less interest in the epidemiology of A. marginale subsp. centrale, and, as a result, there are f
257 to simultaneously detect A. marginale and A. marginale subsp. centrale.
258 e induced by live vaccination with Anaplasma marginale subsp. centrale.
259                                    Anaplasma marginale subspecies centrale also infects cattle; howev
260                                    Anaplasma marginale superinfection occurs when the second strain c
261                                           A. marginale T4SS proteins VirB2, VirB4-1, VirB4-2, VirB6-1
262 strains of the tick-borne pathogen Anaplasma marginale that encode distinctly different surface prote
263  region bound a unique MSP-2 expressed on A. marginale that was not recognized by antibody generated
264                                    Anaplasma marginale, the causative agent of bovine anaplasmosis, i
265                                     As in A. marginale, the msp2(p44) gene in this expression site is
266  DNA from closely related species (Anaplasma marginale, the white-tailed deer agent, and additional E
267                          Maries strain of A. marginale; the correct identification was confirmed by e
268 sis and is used as a live vaccine against A. marginale There has been less interest in the epidemiolo
269                                           A. marginale thus maintains two large, separate systems wit
270  also homologous to the MSP-2 proteins of A. marginale; thus, they were designated GE MSP-2A (45 kDa)
271                            The ability of A. marginale to persist in cattle has been shown to be due,
272 cosylation may play a role in adhesion of A. marginale to tick cells because chemical deglycosylation
273  of MSP1a plays a role in the adhesion of A. marginale to tick cells.
274 tion of MSP1a plays a role in adhesion of A. marginale to tick cells.
275 he United States would be associated with A. marginale transmission.
276 enomic expression site in Oklahoma strain A. marginale transmitted from in vitro cultures to cattle a
277  which encodes the complete msp1a gene of A. marginale under the control of human cytomegalovirus imm
278 ber of variants required for persistence, A. marginale uses segmental gene conversion, in which oligo
279                                    Anaplasma marginale utilizes gene conversion of a repertoire of si
280 tion was addressed by tracking the Anaplasma marginale variant population and corresponding segment-s
281 rized by rickettsemic cycles in which new A. marginale variant types, defined by the sequence of the
282           This restriction of transmitted A. marginale variant types, in contrast to the marked diver
283 amined allelic usage in generating Anaplasma marginale variants during in vivo infection in the mamma
284  are highly conserved among the expressed A. marginale variants, and similar sequences define the MSP
285 e ratio of 16S rRNA to 16S DNA copies for A. marginale was determined to be 117.9:1 (95% confidence i
286         Recently, the Virginia isolate of A. marginale was propagated in a continuous tick cell line,
287 iants revealed a change in structure when A. marginale was transferred from one cell-type to another,
288                 The South Idaho strain of A. marginale was used, as MSP2 expression is restricted to
289                          Maries strain of A. marginale, we show that this surface coat is dominated b
290 unctional pseudogenes from two strains of A. marginale were detected and extracted from the phi29-amp
291               Two calves seronegative for A. marginale were immunized four times, at weeks 0, 3, 7, a
292                           Five strains of A. marginale were selected in order to identify and compare
293 nce, Anaplasma phagocytophilum and Anaplasma marginale were successfully localized in situ within int
294 he msp1aS gene, a homolog of msp1alpha of A. marginale, which contains repeats at the 5' ends that ar
295     We examined this balance using Anaplasma marginale, which generates antigenic variants in the out
296 MSP1a) and MSP1b form the MSP1 complex of A. marginale, which is involved in adhesion of the pathogen
297  confirmed to be expressed as proteins by A. marginale within infected erythrocytes, with expression
298  heterogeneity observed among isolates of A. marginale within Oklahoma could be explained by cattle m
299 2 (MSP2) variants are expressed by Anaplasma marginale within the tick salivary gland and, following
300 cases, it is reported as coinfection with A. marginale without characterization of the strain.

WebLSDに未収録の専門用語(用法)は "新規対訳" から投稿できます。
 
Page Top