Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
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
2.
Int J Mol Sci ; 23(21)2022 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-36361509

RESUMO

Ehrlichia chaffeensis, a tick-transmitted intraphagosomal bacterium, is the causative agent of human monocytic ehrlichiosis. The pathogen also infects several other vertebrate hosts. E. chaffeensis has a biphasic developmental cycle during its growth in vertebrate monocytes/macrophages and invertebrate tick cells. Host- and vector-specific differences in the gene expression from many genes of E. chaffeensis are well documented. It is unclear how the organism regulates gene expression during its developmental cycle and for its adaptation to vertebrate and tick host cell environments. We previously mapped promoters of several E. chaffeensis genes which are recognized by its only two sigma factors: σ32 and σ70. In the current study, we investigated in assessing five predicted E. chaffeensis transcription regulators; EcxR, CtrA, MerR, HU and Tr1 for their possible roles in regulating the pathogen gene expression. Promoter segments of three genes each transcribed with the RNA polymerase containing σ70 (HU, P28-Omp14 and P28-Omp19) and σ32 (ClpB, DnaK and GroES/L) were evaluated by employing multiple independent molecular methods. We report that EcxR binds to all six promoters tested. Promoter-specific binding of EcxR to several gene promoters results in varying levels of gene expression enhancement. This is the first detailed molecular characterization of transcription regulators where we identified EcxR as a gene regulator having multiple promoter-specific interactions.


Assuntos
Ehrlichia chaffeensis , Carrapatos , Animais , Humanos , Ehrlichia chaffeensis/genética , RNA Polimerases Dirigidas por DNA/metabolismo , Regulação da Expressão Gênica , Regiões Promotoras Genéticas , Monócitos/metabolismo , Fatores de Transcrição/metabolismo , Carrapatos/metabolismo
3.
J Bacteriol ; 203(13): e0002721, 2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-33875547

RESUMO

Ehrlichia chaffeensis causes human monocytic ehrlichiosis by replicating within phagosomes of monocytes/macrophages. A function disruption mutation within the pathogen's ECH_0660 gene, which encodes a phage head-to-tail connector protein, resulted in the rapid clearance of the pathogen in vivo, while aiding in induction of sufficient immunity in a host to protect against wild-type infection challenge. In this study, we describe the characterization of a cluster of seven genes spanning from ECH_0659 to ECH_0665, which contained four genes encoding bacterial phage proteins, including the ECH_0660 gene. Assessment of the promoter region upstream of the first gene of the seven genes (ECH_0659) in Escherichia coli demonstrated transcriptional enhancement under zinc and iron starvation conditions. Furthermore, transcription of the seven genes was significantly higher under zinc and iron starvation conditions for E. chaffeensis carrying a mutation in the ECH_0660 gene compared to the wild-type pathogen. In contrast, for the ECH_0665 gene mutant with the function disruption, transcription from the genes was mostly similar to that of the wild type or was moderately downregulated. Recently, we reported that this mutation caused a minimal impact on the pathogen's in vivo growth, as it persisted similarly to the wild type. The current study is the first to describe how zinc and iron contribute to E. chaffeensis biology. Specifically, we demonstrated that the functional disruption in the gene encoding the phage head-to-tail connector protein in E. chaffeensis results in the enhanced transcription of seven genes, including those encoding phage proteins, under zinc and iron limitation. IMPORTANCE Ehrlichia chaffeensis, a tick-transmitted bacterium, causes human monocytic ehrlichiosis by replicating within phagosomes of monocytes/macrophages. A function disruption mutation within the pathogen's gene encoding a phage head-to-tail connector protein resulted in the rapid clearance of the pathogen in vivo, while aiding in induction of sufficient immunity in a host to protect against wild-type infection challenge. In the current study, we investigated if the functional disruption in the phage head-to-tail connector protein gene caused transcriptional changes resulting from metal ion limitations. This is the first study describing how zinc and iron may contribute to E. chaffeensis replication.


Assuntos
Proteínas de Bactérias/genética , Ehrlichia chaffeensis/genética , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Genes Bacterianos/genética , Ferro/farmacologia , Mutação , Zinco/farmacologia , Animais , Bacteriófagos/genética , Ehrlichiose/microbiologia , Escherichia coli/genética , Humanos , Imunidade , Monócitos/microbiologia , Carrapatos/microbiologia , Transcrição Gênica
4.
Infect Immun ; 89(4)2021 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-33431703

RESUMO

Rickettsiae belong to the Anaplasmataceae family, which includes mostly tick-transmitted pathogens causing human, canine, and ruminant diseases. Biochemical characterization of the pathogens remains a major challenge because of their obligate parasitism. We investigated the use of an axenic medium for growth of two important pathogens-Anaplasma phagocytophilum and Ehrlichia chaffeensis-in host cell-free phagosomes. We recently reported that the axenic medium promotes protein and DNA biosynthesis in host cell-free replicating form of E. chaffeensis, although the bacterial replication is limited. We now tested the hypothesis that growth on axenic medium can be improved if host cell-free rickettsia-containing phagosomes are used. Purification of phagosomes from A. phagocytophilum- and E. chaffeensis-infected host cells was accomplished by density gradient centrifugation combined with magnet-assisted cell sorting. Protein and DNA synthesis was observed for both organisms in cell-free phagosomes with glucose-6-phosphate and/or ATP. The levels of protein and DNA synthesis were the highest for a medium pH of 7. The data demonstrate bacterial DNA and protein synthesis for the first time in host cell-free phagosomes for two rickettsial pathogens. The host cell support-free axenic growth of obligate pathogenic rickettsiae will be critical in advancing research goals in many important tick-borne diseases impacting human and animal health.


Assuntos
Anaplasma phagocytophilum/fisiologia , Cultura Axênica , Replicação do DNA , Ehrlichia chaffeensis/fisiologia , Fagossomos/microbiologia , Biossíntese de Proteínas , Sistema Livre de Células , Fracionamento Químico , Humanos , Concentração de Íons de Hidrogênio
5.
Biol Cell ; 112(9): 251-264, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32445585

RESUMO

Osteoclasts are multinucleated giant cells, responsible for bone resorption. Osteoclast differentiation and function requires a series of cytokines to remove the old bone, which coordinates with the induction of bone remodelling by osteoblast-mediated bone formation. Studies have demonstrated that AMP-activated protein kinase (AMPK) play a negative regulatory role in osteoclast differentiation and function. Research involving AMPK, a nutrient and energy sensor, has primarily focused on osteoclast differentiation and function; thus, its role in autophagy, inflammation and immunity remains poorly understood. Autophagy is a conservative homoeostatic mechanism of eukaryotic cells, and response to osteoclast differentiation and function; however, how it interacts with inflammation remains unclear. Additionally, based on the regulatory function of different AMPK subunits for osteoclast differentiation and function, its activation is regulated by upstream factors to perform bone metabolism. This review summarises the critical role of AMPK-mediated autophagy, inflammation and immunity by upstream and downstream signalling during receptor activator of nuclear factor kappa-B ligand-induced osteoclast differentiation and function. This pathway may provide therapeutic targets for bone-related diseases, as well as function as a biomarker for bone homoeostasis.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Doenças Ósseas , Osteoclastos , Animais , Autofagia , Doenças Ósseas/metabolismo , Doenças Ósseas/patologia , Diferenciação Celular , Linhagem Celular , Humanos , Imunidade , Inflamação/metabolismo , Osteoclastos/citologia , Osteoclastos/metabolismo
6.
Int J Mol Sci ; 22(16)2021 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-34445146

RESUMO

Ehrlichia chaffeensis causes human monocytic ehrlichiosis. Little is known about how this and other related tick-borne rickettsia pathogens maintain pH homeostasis in acidified phagosomes and the extracellular milieu. The membrane-bound sodium (cation)/proton antiporters are found in a wide range of organisms aiding pH homeostasis. We recently reported a mutation in an antiporter gene of E. chaffeensis (ECH_0379) which causes bacterial in vivo attenuation. The E. chaffeensis genome contains 10 protein coding sequences encoding for predicted antiporters. We report here that nine of these genes are transcribed during the bacterial growth in macrophages and tick cells. All E. chaffeensis antiporter genes functionally complemented antiporter deficient Escherichia coli. Antiporter activity for all predicted E. chaffeensis genes was observed at pH 5.5, while gene products of ECH_0179 and ECH_0379 were also active at pH 8.0, and ECH_0179 protein was complemented at pH 7.0. The antiporter activity was independently verified for the ECH_0379 protein by proteoliposome diffusion analysis. This is the first description of antiporters in E. chaffeensis and demonstrates that the pathogen contains multiple antiporters with varying biological functions, which are likely important for the pH homeostasis of the pathogen's replicating and infectious forms.


Assuntos
Antiporters/genética , Bactérias/genética , Proteínas de Bactérias/genética , Ehrlichia chaffeensis/genética , Genes Bacterianos/genética , Homeostase/genética , Sódio/metabolismo , Escherichia coli/genética , Concentração de Íons de Hidrogênio , Macrófagos/metabolismo , Mutação/genética , Prótons
7.
Infect Immun ; 88(10)2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32747600

RESUMO

Ehrlichia chaffeensis, a tick-transmitted obligate intracellular rickettsial agent, causes human monocytic ehrlichiosis. In recent reports, we described substantial advances in developing random and targeted gene disruption methods to investigate the functions of E. chaffeensis genes. We reported earlier that the Himar1 transposon-based random mutagenesis is a valuable tool in defining E. chaffeensis genes critical for its persistent growth in vivo in reservoir and incidental hosts. The method also aided in extending studies focused on vaccine development and immunity. Here, we describe the generation and mapping of 55 new mutations. To define the critical nature of the bacterial genes, infection experiments were carried out in the canine host with pools of mutant organisms. Infection evaluation in the physiologically relevant host by molecular assays and by xenodiagnoses allowed the identification of many proteins critical for the pathogen's persistent in vivo growth. Genes encoding proteins involved in biotin biosynthesis, protein synthesis and fatty acid biosynthesis, DNA repair, electron transfer, and a component of a multidrug resistance (MDR) efflux pump were concluded to be essential for the pathogen's in vivo growth. Three known immunodominant membrane proteins, i.e., two 28-kDa outer membrane proteins (P28/OMP) and a 120-kDa surface protein, were also recognized as necessary for the pathogen's obligate intracellular life cycle. The discovery of many E. chaffeensis proteins crucial for its continuous in vivo growth will serve as a major resource for investigations aimed at defining pathogenesis and developing novel therapeutics for this and related pathogens of the rickettsial family Anaplasmataceae.


Assuntos
Ehrlichia chaffeensis/genética , Ehrlichiose/microbiologia , Genes Bacterianos , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Cães , Ehrlichia chaffeensis/crescimento & desenvolvimento , Ehrlichia chaffeensis/patogenicidade , Ehrlichiose/transmissão , Biblioteca Gênica , Genoma Bacteriano/genética , Macrófagos/microbiologia , Mutagênese Insercional , Mutação , Carrapatos , Transcrição Gênica , Virulência/genética
8.
Infect Immun ; 87(2)2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30396898

RESUMO

Rocky Mountain spotted fever (RMSF) is a potentially fatal tick-borne disease in people and dogs. RMSF is reported in the United States and several countries in North, Central, and South America. The causative agent of this disease, Rickettsia rickettsii, is transmitted by several species of ticks, including Dermacentor andersoni, Rhipicephalus sanguineus, and Amblyomma americanum RMSF clinical signs generally include fever, headache, nausea, vomiting, muscle pain, lack of appetite, and rash. If untreated, it can quickly progress into a life-threatening illness in people and dogs, with high fatality rates ranging from 30 to 80%. While RMSF has been known for over a century, recent epidemiological data suggest that the numbers of documented cases and the fatality rates remain high in people, particularly during the last two decades in parts of North America. Currently, there are no vaccines available to prevent RMSF in either dogs or people. In this study, we investigated the efficacies of two experimental vaccines, a subunit vaccine containing two recombinant outer membrane proteins as recombinant antigens (RCA) and a whole-cell inactivated antigen vaccine (WCA), in conferring protection against virulent R. rickettsii infection challenge in a newly established canine model for RMSF. Dogs vaccinated with WCA were protected from RMSF, whereas those receiving RCA developed disease similar to that of nonvaccinated R. rickettsii-infected dogs. WCA also reduced the pathogen loads to nearly undetected levels in the blood, lungs, liver, spleen, and brain and induced bacterial antigen-specific immune responses. This study provides the first evidence of the protective ability of WCA against RMSF in dogs.


Assuntos
Antígenos de Bactérias/imunologia , Doenças do Cão , Rickettsia rickettsii/imunologia , Vacinas Antirrickéttsia/imunologia , Febre Maculosa das Montanhas Rochosas , Animais , Proteínas da Membrana Bacteriana Externa/imunologia , Doenças do Cão/imunologia , Doenças do Cão/microbiologia , Doenças do Cão/prevenção & controle , Cães , Proteínas Recombinantes/imunologia , Febre Maculosa das Montanhas Rochosas/imunologia , Febre Maculosa das Montanhas Rochosas/prevenção & controle , Febre Maculosa das Montanhas Rochosas/veterinária
9.
Infect Immun ; 83(7): 2827-35, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25916990

RESUMO

Ehrlichia chaffeensis, a tick-borne rickettsial organism, causes the disease human monocytic ehrlichiosis. The pathogen also causes disease in several other vertebrates, including dogs and deer. In this study, we assessed two clonally purified E. chaffeensis mutants with insertions within the genes Ech_0379 and Ech_0660 as vaccine candidates in deer and dogs. Infection with the Ech_0379 mutant and challenge with wild-type E. chaffeensis 1 month following inoculation with the mutant resulted in the reduced presence of the organism in blood compared to the presence of wild-type infection in both deer and dogs. The Ech_0660 mutant infection resulted in its rapid clearance from the bloodstream. The wild-type infection challenge following Ech_0660 mutant inoculation also caused the pathogen's clearance from blood and tissue samples as assessed at the end of the study. The Ech_0379 mutant-infected and -challenged animals also remained positive for the organism in tissue samples in deer but not in dogs. This is the first study that documents that insertion mutations in E. chaffeensis that cause attenuated growth confer protection against wild-type infection challenge. This study is important in developing vaccines to protect animals and people against Ehrlichia species infections.


Assuntos
Vacinas Bacterianas/imunologia , Ehrlichia chaffeensis/imunologia , Ehrlichiose/prevenção & controle , Ehrlichiose/veterinária , Animais , Carga Bacteriana , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/genética , Sangue/microbiologia , Cervos , Cães , Ehrlichia chaffeensis/genética , Ehrlichiose/imunologia , Genes Bacterianos , Humanos , Mutagênese Insercional , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia
10.
PLoS Pathog ; 9(2): e1003171, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23459099

RESUMO

Ehrlichia chaffeensis is a tick transmitted pathogen responsible for the disease human monocytic ehrlichiosis. Research to elucidate gene function in rickettsial pathogens is limited by the lack of genetic manipulation methods. Mutational analysis was performed, targeting to specific and random insertion sites within the bacterium's genome. Targeted mutagenesis at six genomic locations by homologous recombination and mobile group II intron-based methods led to the consistent identification of mutants in two genes and in one intergenic site; the mutants persisted in culture for 8 days. Three independent experiments using Himar1 transposon mutagenesis of E. chaffeensis resulted in the identification of multiple mutants; these mutants grew continuously in macrophage and tick cell lines. Nine mutations were confirmed by sequence analysis. Six insertions were located within non-coding regions and three were present in the coding regions of three transcriptionally active genes. The intragenic mutations prevented transcription of all three genes. Transposon mutants containing a pool of five different insertions were assessed for their ability to infect deer and subsequent acquisition by Amblyomma americanum ticks, the natural reservoir and vector, respectively. Three of the five mutants with insertions into non-coding regions grew well in deer. Transposition into a differentially expressed hypothetical gene, Ech_0379, and at 18 nucleotides downstream to Ech_0230 gene coding sequence resulted in the inhibition of growth in deer, which is further evidenced by their failed acquisition by ticks. Similarly, a mutation into the coding region of ECH_0660 gene inhibited the in vivo growth in deer. This is the first study evaluating targeted and random mutagenesis in E. chaffeensis, and the first to report the generation of stable mutants in this obligate intracellular bacterium. We further demonstrate that in vitro mutagenesis coupled with in vivo infection assessment is a successful strategy in identifying genomic regions required for the pathogen's in vivo growth.


Assuntos
Proteínas de Bactérias/genética , Elementos de DNA Transponíveis/genética , Cervos/microbiologia , Ehrlichia chaffeensis/genética , Ehrlichiose/transmissão , Mutação/genética , Carrapatos/microbiologia , Sequência de Aminoácidos , Animais , Antibacterianos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Southern Blotting , Células Cultivadas , Cervos/genética , Ehrlichia chaffeensis/efeitos dos fármacos , Ehrlichia chaffeensis/patogenicidade , Ehrlichiose/genética , Ehrlichiose/veterinária , Genoma Bacteriano , Humanos , Macrófagos/microbiologia , Dados de Sequência Molecular , Mutagênese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Recombinação Genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Carrapatos/genética
11.
Cell Stress Chaperones ; 29(4): 540-551, 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38908470

RESUMO

Anaplasma phagocytophilum is an intracellular tick-transmitted bacterial pathogen that infects neutrophils in mammals and causes granulocytic anaplasmosis. In this study, we investigated the molecular chaperones ClpB and DnaK from A. phagocytophilum. In Escherichia coli, ClpB cooperates with DnaK and its co-chaperones DnaJ and GrpE in ATP-dependent reactivation of aggregated proteins. Since ClpB is not produced in metazoans, it is a promising target for developing antimicrobial therapies, which generates interest in studies on that chaperone's role in pathogenic bacteria. We found that ClpB and DnaK are transcriptionally upregulated in A. phagocytophilum 3-5 days after infection of human HL-60 and tick ISE6 cells, which suggests an essential role of the chaperones in supporting the pathogen's intracellular life cycle. Multiple sequence alignments show that A. phagocytophilum ClpB and DnaK contain all structural domains that were identified in their previously studied orthologs from other bacteria. Both A. phagocytophilum ClpB and DnaK display ATPase activity, which is consistent with their participation in the ATP-dependent protein disaggregation system. However, despite a significant sequence similarity between the chaperones from A. phagocytophilum and those from E. coli, the former were not as effective as their E. coli orthologs during reactivation of aggregated proteins in vitro and in supporting the survival of E. coli cells under heat stress. We conclude that the A. phagocytophilum chaperones might have evolved with distinct biochemical properties to maintain the integrity of pathogenic proteins under unique stress conditions of an intracellular environment of host cells.

12.
Vaccine ; 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38880695

RESUMO

Anaplasma marginale is a tick-borne pathogen of cattle that causes bovine anaplasmosis in tropical and subtropical regions throughout the world. Killed vaccines derived from infected erythrocytes have been used for control of this disease with limited success. Recently, we described a targeted deletion mutation in the phage head-to-tail connector protein gene of A. marginale which caused bacterial attenuation in vivo and provided protection as a modified live vaccine (MLAV). Following intravenous injection of susceptible steers, the MLAV induced protective immunity against disease progression. In the current study, we demonstrated that the immunity resulting from MLAV in cattle prevents the disease progression resulting from virulent A. marginale intrastadial transmission from infected Dermacentor variabilis male ticks. The nonimmunized control steers receiving the infection from ticks developed fever, lethargy, and inappetence for several days post tick exposure with significant decreases in the packed cell volume and increases in bacteremia. In contrast, the MLAV immunized steers remained healthy after being challenged with infected ticks and this group of animals had a significant reduction in bacteremia as compared with the controls. This study demonstrated that the A. marginale MLAV provided protection against acute tick-transmitted anaplasmosis, in addition to protection documented in steers challenge-exposed with infected blood as reported previously.

13.
Front Cell Infect Microbiol ; 13: 1220025, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37457955

RESUMO

Ehrlichia chaffeensis is a tick-transmitted monocytic ehrlichiosis agent primarily causing the disease in people and dogs. We recently described the development and characterization of 55 random mutations in E. chaffeensis, which aided in defining the critical nature of many bacterial genes for its growth in a physiologically relevant canine infection model. In the current study, we tested 45 of the mutants for their infectivity ability to the pathogen's tick vector; Amblyomma americanum. Four mutations resulted in the pathogen's replication deficiency in the tick, similar to the vertebrate host. Mutations causing growth defects in both vertebrate and tick hosts included in genes coding for a predicted alpha/beta hydrolase, a putative dicarboxylate amino acid:cation symporter, a T4SS protein, and predicted membrane-bound proteins. Three mutations caused the bacterial defective growth only in the tick vector, which represented putative membrane proteins. Ten mutations causing no growth defect in the canine host similarly grew well in the tick vector. Mutations in 28 genes/genomic locations causing E. chaffeensis growth attenuation in the canine host were recognized as non-essential for its growth in the tick vector. The tick non-essential genes included genes coding for many metabolic pathway- and outer membrane-associated proteins. This study documents novel vector- and host-specific differences in E. chaffeensis for its functional gene requirements.


Assuntos
Ehrlichia chaffeensis , Ehrlichiose , Carrapatos , Animais , Cães , Carrapatos/microbiologia , Amblyomma , Ehrlichia chaffeensis/metabolismo , Infecção Persistente , Vertebrados , Ehrlichiose/veterinária , Ehrlichiose/microbiologia
14.
Infect Immun ; 80(10): 3576-86, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22851751

RESUMO

Ehrlichia chaffeensis is an obligate intracellular bacterium that causes human monocytic ehrlichiosis (HME). To determine what host components are important for bacterial replication, we performed microarray analysis on Drosophila melanogaster S2 cells by comparing host gene transcript levels between permissive and nonpermissive conditions for E. chaffeensis growth. Five-hundred twenty-seven genes had increased transcript levels unique to permissive growth conditions 24 h postinfection. We screened adult flies that were mutants for several of the "permissive" genes for the ability to support Ehrlichia replication. Three additional D. melanogaster fly lines with putative mutations in pyrimidine metabolism were also tested. Ten fly lines carrying mutations in the genes CG6479, separation anxiety, chitinase 11, CG6364 (Uck2), CG6543 (Echs1), withered (whd), CG15881 (Ccdc58), CG14806 (Apop1), CG11875 (Nup37), and dumpy (dp) had increased resistance to infection with Ehrlichia. Analysis of RNA by quantitative real-time reverse transcription-PCR (qRT-PCR) confirmed that the bacterial load was decreased in these mutant flies compared to wild-type infected control flies. Seven of these genes (san, Cht11, Uck2, Echs1, whd, Ccdc58, and Apop1) encoded proteins that had mitochondrial functions or could be associated with proteins with mitochondrial functions. Treatment of THP-1 cells with double-stranded RNA to silence the human UCK2 gene indicates that the disruption of the uridine-cytidine kinase affects E. chaffeensis replication in human macrophages. Experiments with cyclopentenyl cytosine (CPEC), a CTP synthetase inhibitor and cytosine, suggest that the nucleotide salvage pathway is essential for E. chaffeensis replication and that it may be important for the provision of CTP, uridine, and cytidine nucleotides.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Ehrlichia chaffeensis/fisiologia , Ehrlichiose/imunologia , Regulação da Expressão Gênica/imunologia , Animais , Diferenciação Celular , Linhagem Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/microbiologia , Ehrlichiose/genética , Técnicas de Silenciamento de Genes , Inativação Gênica , Células Precursoras de Granulócitos/metabolismo , Humanos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Análise Serial de Proteínas , Reação em Cadeia da Polimerase em Tempo Real/métodos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Uridina Quinase/genética , Uridina Quinase/metabolismo
15.
Front Cell Infect Microbiol ; 12: 1070356, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36619760

RESUMO

Tick-transmitted Ehrlichia chaffeensis, the causative agent for human monocytic ehrlichiosis, resides and multiplies within a host cell phagosome. Infection progression of E. chaffeensis includes internalization into a host cell by host cell membrane fusion events following engulfment leading to the formation of E. chaffeensis containing vacuole (ECV). Revealing the molecular composition of ECV is important in understanding the host cellular processes, evasion of host defense pathways and in defining host-pathogen interactions. ECVs purified from infected host cells were analyzed to define both host and bacterial proteomes associated with the phagosome membranes. About 160 bacterial proteins and 2,683 host proteins were identified in the ECV membranes. The host proteins included predominantly known phagosome proteins involved in phagocytic trafficking, fusion of vesicles, protein transport, Ras signaling pathway and pathogenic infection. Many highly expressed proteins were similar to the previously documented proteins of phagosome vacuole membranes containing other obligate pathogenic bacteria. The finding of many bacterial membrane proteins is novel; they included multiple outer membrane proteins, such as the p28-Omps, the 120 kDa protein, preprotein translocases, lipoproteins, metal binding proteins, and chaperonins, although the presence of ankyrin repeat proteins, several Type I and IV secretion system proteins is anticipated. This study demonstrates that ECV membrane is extensively modified by the pathogen. This study represents the first and the most comprehensive description of ECV membrane proteome. The identity of many host and Ehrlichia proteins in the ECV membrane will be a valuable to define pathogenic mechanisms critical for the replication of the pathogen within macrophages.


Assuntos
Ehrlichia chaffeensis , Ehrlichiose , Humanos , Proteoma/análise , Ehrlichia chaffeensis/metabolismo , Proteínas de Bactérias/metabolismo , Fagossomos/química , Proteínas de Membrana/metabolismo , Ehrlichiose/microbiologia
16.
BMC Microbiol ; 11: 83, 2011 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-21513529

RESUMO

BACKGROUND: Ehrlichia chaffeensis is a tick-transmitted rickettsial pathogen responsible for an important emerging disease, human monocytic ehrlichiosis. To date how E. chaffeensis and many related tick-borne rickettsial pathogens adapt and persist in vertebrate and tick hosts remain largely unknown. In recent studies, we demonstrated significant host-specific differences in protein expression in E. chaffeensis originating from its tick and vertebrate host cells. The adaptive response of the pathogen to different host environments entails switch of gene expression regulated at the level of transcription, possibly by altering RNA polymerase activity. RESULTS: In an effort to understand the molecular basis of pathogen gene expression differences, we isolated native E. chaffeensis RNA polymerase using a heparin-agarose purification method and developed an in vitro transcription system to map promoter regions of two differentially expressed genes of the p28 outer membrane protein locus, p28-Omp14 and p28-Omp19. We also prepared a recombinant protein of E. chaffeensis σ70 homologue and used it for in vitro promoter analysis studies. The possible role of one or more proteins presents in E. chaffeensis lysates in binding to the promoter segments and on the modulation of in vitro transcription was also assessed. CONCLUSIONS: Our experiments demonstrated that both the native and recombinant proteins are functional and have similar enzyme properties in driving the transcription from E. chaffeensis promoters. This is the first report of the functional characterization of E. chaffeensis RNA polymerase and in vitro mapping of the pathogen promoters using the enzyme. This study marks the beginning to broadly characterize the mechanisms controlling the transcription by Anaplasmataceae pathogens.


Assuntos
Proteínas da Membrana Bacteriana Externa/genética , RNA Polimerases Dirigidas por DNA/genética , RNA Polimerases Dirigidas por DNA/metabolismo , Ehrlichia chaffeensis/enzimologia , Ehrlichia chaffeensis/genética , Perfilação da Expressão Gênica , Regiões Promotoras Genéticas , Sequência de Aminoácidos , Cromatografia Líquida/métodos , RNA Polimerases Dirigidas por DNA/isolamento & purificação , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Dados de Sequência Molecular , Ligação Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Transcrição Gênica
17.
Pathogens ; 10(4)2021 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-33918856

RESUMO

Ehrlichia ruminantium, a tick-borne rickettsial, causes heartwater in ruminants resulting from vascular damage. Severity of heartwater varies greatly in ruminant species and breeds, age of animals and for diverse geographic E. ruminantium strains. E. ruminantium and a tick vector, Amblyomma variegatum, originating from Africa, are well established in certain Caribbean islands two centuries ago. Besides the possibility of introduction of heartwater through African exotic animal importation, presence of the pathogen, and the tick vector in the Caribbean pose a high risk to ruminants in the USA and other western hemisphere countries. Scientific evidence supporting the heartwater threat to nonendemic regions, however, is lacking. We describe the first infection study in sheep reared in the USA with seven E. ruminantium strains. All infected sheep exhibited clinical signs characteristic of subacute to subclinical disease, which included labored breathing, depression, coughing, and nasal discharges. Gross and microscopic lesions consistent with heartwater disease including edema and hemorrhage were observed in several organs. Pathogen-specific IgG antibody response was detected in animals infected with all seven strains, while molecular analysis confirmed the pathogen presence only when infected with in vitro cultures. This is the first infection study demonstrating severe heartwater in sheep reared in North America.

18.
Ticks Tick Borne Dis ; 12(4): 101727, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33865177

RESUMO

Anaplasma platys is a tick-transmitted rickettsial pathogen, which is known to be the etiologic agent for cyclic thrombocytopenia in its primary canine host. Infections with this pathogen are also reported in cats, cattle and people. Similarly, Ehrlichia canis is another tick-borne rickettsial pathogen responsible for canine monocytic ehrlichiosis and is also reported to cause infections in people. We describe infections in dogs with these two pathogens on the Caribbean island of Grenada, West Indies by detection using molecular methods. We utilized a 16S rRNA gene-based PCR assay to detect both Ehrlichia and Anaplasma species by screening 155 canine blood samples from asymptomatic dogs. We found 18.7 % of the dogs to be positive for A. platys and 16.8 % for E. canis. Samples that tested positive for A. platys were further assessed by sequence analysis targeting 16S rRNA, 23S rRNA, citrate synthase (gltA) and heat shock protein (groEL) genes. Phylogenetic analysis revealed high correlation of A. platys 16S rRNA and gltA gene sequences with the geographic origins, while 23S rRNA and groEL gene sequences clustered independent of the geographic origins. This study represents an important step in defining the widespread distribution of active rickettsial infections in Caribbean dogs with no apparent clinical signs, thus posing a high risk for canine health and to a lesser extent to humans, as most dogs in the Caribbean are free-roaming.


Assuntos
Anaplasma/isolamento & purificação , Anaplasmose/epidemiologia , Doenças do Cão/epidemiologia , Ehrlichia canis/isolamento & purificação , Ehrlichiose/veterinária , Anaplasma/enzimologia , Anaplasma/genética , Anaplasmose/microbiologia , Animais , Proteínas de Bactérias/análise , Chaperonina 60/análise , Citrato (si)-Sintase/análise , Doenças do Cão/microbiologia , Cães , Ehrlichia canis/enzimologia , Ehrlichia canis/genética , Ehrlichiose/epidemiologia , Ehrlichiose/microbiologia , Granada/epidemiologia , Prevalência , RNA Bacteriano/análise , RNA Ribossômico 16S/análise , RNA Ribossômico 23S/análise
19.
J Clin Microbiol ; 48(7): 2424-32, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20463162

RESUMO

Insufficient diagnostic sensitivity and specificity coupled with the potential for cross-reactivity among closely related Anaplasma species has made the accurate determination of infection status problematic. A method for the development of simplex and duplex real-time quantitative reverse transcriptase PCR (qRT-PCR) assays for the detection of A. marginale and A. phagocytophilum 16S rRNA in plasma-free bovine peripheral blood samples is described. The duplex assay was able to detect as few as 100 copies of 16S rRNA of both A. marginale and A. phagocytophilum in the same reaction. The ratio of 16S rRNA to 16S DNA copies for A. marginale was determined to be 117.9:1 (95% confidence interval [95% CI], 100.7:1, 135.2:1). Therefore, the detection limit is the minimum infective unit of one A. marginale bacterium. The duplex assay detected nonequivalent molar ratios as high as 100-fold. Additionally, the duplex assay and a competitive enzyme-linked immunosorbent assay (cELISA) were used to screen 237 samples collected from herds in which anaplasmosis was endemic. When the cELISA was evaluated by the results of the qRT-PCR, its sensitivity and specificity for the detection of A. marginale infection were found to be 65.2% (95% CI, 55.3%, 75.1%) and 97.3% (95% CI, 94.7%, 99.9%), respectively. A. phagocytophilum infection was not detected in the samples analyzed. One- and two-way receiver operator characteristic curves were constructed in order to recommend the optimum negative cutoff value for the cELISA. Percentages of inhibition of 20 and 15.3% were recommended for the one- and two-way curves, respectively. In conclusion, the duplex real-time qRT-PCR assay is a highly sensitive and specific diagnostic tool for the accurate and precise detection of A. marginale and A. phagocytophilum infections in cattle.


Assuntos
Anaplasma marginale/isolamento & purificação , Anaplasma phagocytophilum/isolamento & purificação , DNA Bacteriano/sangue , Técnicas de Diagnóstico Molecular/métodos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Análise de Variância , Anaplasmose/diagnóstico , Anaplasmose/microbiologia , Animais , Bovinos , RNA Ribossômico 16S/genética , Curva ROC , Análise de Regressão
20.
Am J Vet Res ; 71(10): 1178-88, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20919904

RESUMO

OBJECTIVE: To compare iatrogenic transmission of Anaplasma marginale during sham vaccination between needle and needle-free injection techniques. ANIMALS: 26 Holstein steers confirmed negative for anaplasmosis by use of a competitive ELISA (cELISA) and an A marginale-specific reverse transcription (RT)-PCR assay. PROCEDURES: An isolate of A marginale was propagated to a circulating parasitemia of 2.0% in a splenectomized steer. Sham vaccination was performed in the left cervical muscles of the splenectomized parasitemic steer with a hypodermic needle fitted to a multiple-dose syringe. The same needle and syringe were used to sham vaccinate a naïve steer. This 2-step procedure was repeated until 10 naïve steers (group ND) were injected. Similarly, sham vaccination of the left cervical muscles of the splenectomized parasitemic steer and another group of 10 naïve steers (group NF) was performed by use of a needle-free injection system. Five control steers were not injected. Disease status was evaluated twice weekly for 61 days by use of light microscopy, a cELISA, and an A marginale-specific RT-PCR assay. RESULTS: Iatrogenic transmission was detected in 6 of 10 steers in group ND. Disease status did not change in the NF or control steers. Sensitivity of light microscopy, cELISA, and RT-PCR assay was 100% on days 41, 41, and 20 after sham vaccination, respectively; however, only cELISA and RT-PCR assay sustained a sensitivity of 100% thereafter. CONCLUSIONS AND CLINICAL RELEVANCE: Needle-free injection was superior to needle injection for the control of iatrogenic transmission of A marginale.


Assuntos
Anaplasma marginale , Anaplasmose/transmissão , Doenças dos Bovinos/transmissão , Doença Iatrogênica/veterinária , Agulhas/veterinária , Vacinação/veterinária , Animais , Bovinos , Doenças dos Bovinos/microbiologia , Injeções a Jato/veterinária , Masculino , Agulhas/efeitos adversos , Vacinação/instrumentação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA