Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
mBio ; 13(6): e0296122, 2022 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-36409075

RESUMO

Anaplasma phagocytophilum is the etiologic agent of the emerging infection, granulocytic anaplasmosis. This obligate intracellular bacterium lives in a host cell-derived vacuole that receives membrane traffic from multiple organelles to fuel its proliferation and from which it must ultimately exit to disseminate infection. Understanding of these essential pathogenic mechanisms has remained poor. Multivesicular bodies (MVBs) are late endosomal compartments that receive biomolecules from other organelles and encapsulate them into intralumenal vesicles (ILVs) using endosomal sorting complexes required for transport (ESCRT) machinery and ESCRT-independent machinery. Association of the ESCRT-independent protein, ALIX, directs MVBs to the plasma membrane where they release ILVs as exosomes. We report that the A. phagocytophilum vacuole (ApV) is acidified and enriched in lysobisphosphatidic acid, a lipid that is abundant in MVBs. ESCRT-0 and ESCRT-III components along with ALIX localize to the ApV membrane. siRNA-mediated inactivation of ESCRT-0 and ALIX together impairs A. phagocytophilum proliferation and infectious progeny production. RNA silencing of ESCRT-III, which regulates ILV scission, pronouncedly reduces ILV formation in ApVs and halts infection by arresting bacterial growth. Rab27a and its effector Munc13-4, which drive MVB trafficking to the plasma membrane and subsequent exosome release, localize to the ApV. Treatment with Nexinhib20, a small molecule inhibitor that specifically targets Rab27a to block MVB exocytosis, abrogates A. phagocytophilum infectious progeny release. Thus, A. phagocytophilum exploits MVB biogenesis and exosome release to benefit each major stage of its intracellular infection cycle: intravacuolar growth, conversion to the infectious form, and exit from the host cell. IMPORTANCE Anaplasma phagocytophilum causes granulocytic anaplasmosis, a globally emerging zoonosis that can be severe, even fatal, and for which antibiotic treatment options are limited. A. phagocytophilum lives in an endosomal-like compartment that interfaces with multiple organelles and from which it must ultimately exit to spread within the host. How the bacterium accomplishes these tasks is poorly understood. Multivesicular bodies (MVBs) are intermediates in the endolysosomal pathway that package biomolecular cargo from other organelles as intralumenal vesicles for release at the plasma membrane as exosomes. We discovered that A. phagocytophilum exploits MVB biogenesis and trafficking to benefit all aspects of its intracellular infection cycle: proliferation, conversion to its infectious form, and release of infectious progeny. The ability of a small molecule inhibitor of MVB exocytosis to impede A. phagocytophilum dissemination indicates the potential of this pathway as a novel host-directed therapeutic target for granulocytic anaplasmosis.


Assuntos
Anaplasma phagocytophilum , Anaplasmose , Proliferação de Células , Corpos Multivesiculares , Biogênese de Organelas , Animais , Anaplasma phagocytophilum/patogenicidade , Anaplasma phagocytophilum/fisiologia , Anaplasmose/metabolismo , Anaplasmose/microbiologia , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Corpos Multivesiculares/metabolismo , Transporte Proteico
2.
Sci Rep ; 9(1): 13236, 2019 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-31520000

RESUMO

Aerobic organisms evolved conserved mechanisms controlling the generation of reactive oxygen species (ROS) to maintain redox homeostasis signaling and modulate signal transduction, gene expression and cellular functional responses under physiological conditions. The production of ROS by mitochondria is essential in the oxidative stress associated with different pathologies and in response to pathogen infection. Anaplasma phagocytophilum is an intracellular pathogen transmitted by Ixodes scapularis ticks and causing human granulocytic anaplasmosis. Bacteria multiply in vertebrate neutrophils and infect first tick midgut cells and subsequently hemocytes and salivary glands from where transmission occurs. Previous results demonstrated that A. phagocytophilum does not induce the production of ROS as part of its survival strategy in human neutrophils. However, little is known about the role of ROS during pathogen infection in ticks. In this study, the role of tick oxidative stress during A. phagocytophilum infection was characterized through the function of different pathways involved in ROS production. The results showed that tick cells increase mitochondrial ROS production to limit A. phagocytophilum infection, while pathogen inhibits alternative ROS production pathways and apoptosis to preserve cell fitness and facilitate infection. The inhibition of NADPH oxidase-mediated ROS production by pathogen infection appears to occur in both neutrophils and tick cells, thus supporting that A. phagocytophilum uses common mechanisms for infection of ticks and vertebrate hosts. However, differences in ROS response to A. phagocytophilum infection between human and tick cells may reflect host-specific cell tropism that evolved during pathogen life cycle.


Assuntos
Anaplasma phagocytophilum/patogenicidade , Anaplasmose/microbiologia , Vetores de Doenças , Interações Hospedeiro-Patógeno , Ixodes/microbiologia , Redes e Vias Metabólicas , Neutrófilos/microbiologia , Anaplasma phagocytophilum/metabolismo , Anaplasmose/metabolismo , Anaplasmose/transmissão , Animais , Regulação da Expressão Gênica , Células HL-60 , Humanos , Neutrófilos/metabolismo , Oxirredução , Estresse Oxidativo , Coelhos , Ovinos , Transdução de Sinais
3.
Sci Rep ; 9(1): 4235, 2019 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-30862835

RESUMO

Anaplasma phagocytophilum, the aetiologic agent of human granulocytic anaplasmosis (HGA) is an obligate intracellular Gram-negative bacterium with the genome size of 1.47 megabases. The intracellular life style and small size of genome suggest that A. phagocytophilum has to modulate a multitude of host cell physiological processes to facilitate its replication. One strategy employed by A. phagocytophilum is through its type IV secretion system (T4SS), which translocates bacterial effectors into target cells to disrupt normal cellular activities. In this study we developed a TEM-1 ß-lactamase based protein translocation assay and applied this assay for identification of A. phagocytophilum T4SS effectors. An A. phagocytophilum hypothetical protein, APH0215 is identified as a T4SS effector protein and found interacting with trans-Golgi network in transfected cells. Hereby, this protein translocation assay developed in this study will facilitate the identification of A. phagocytophilum T4SS effectors and elucidation of HGA pathogenesis.


Assuntos
Anaplasma phagocytophilum , Anaplasmose , Proteínas de Bactérias , Bioensaio , beta-Lactamases , Anaplasma phagocytophilum/genética , Anaplasma phagocytophilum/metabolismo , Anaplasmose/diagnóstico , Anaplasmose/genética , Anaplasmose/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Células CHO , Cricetinae , Cricetulus , Humanos , Sistemas de Secreção Tipo IV/genética , Sistemas de Secreção Tipo IV/metabolismo , beta-Lactamases/genética , beta-Lactamases/metabolismo
4.
PLoS One ; 12(6): e0179655, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28628633

RESUMO

Human anaplasmosis (HA) is an emerging tick-borne disease that may present as a mild flu-like illness or a life threatening, sepsis-like condition. Although disease severity is hypothesized to relate to immunopathology and immune dysfunction in humans, studies to directly measure immune responses in infected humans have been very limited. We quantified cytokines in 80 confirmed HA patients using a multiplex chemiluminescence immunoassay system and compared similarly measured responses in 1000 control subjects. Pro-inflammatory cytokines were significantly elevated in HA patients (all seven p<0.0001). Interferon gamma (IFN-γ) concentrations were particularly high, with average concentrations 7.8 times higher in the HA patients than the controls. A subset of cytokines consisting of IL-1ß, IL-8, IL-6, TNF-α, and IL-10 was also coordinately high and significantly associated with severity of thrombocytopenia in HA patients. Patients with infections in the very acute stage (≤ 4 days ill) tended to have the highest IFN-γ, IL-12p70, and IL-2 levels. Higher concentrations of IL-13 and IL-5 were associated with diarrhea and vomiting. Our findings support a pathophysiological role for a pro-inflammatory response in HA, especially with regard to the modulation of hematopoiesis and subsequent hematopoietic complications.


Assuntos
Anaplasmose/patologia , Citocinas/análise , Imunoensaio , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Anaplasmose/complicações , Anaplasmose/imunologia , Anaplasmose/metabolismo , Estudos de Casos e Controles , Criança , Pré-Escolar , Diarreia/etiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Contagem de Plaquetas , Índice de Gravidade de Doença , Células Th1/citologia , Células Th1/metabolismo , Células Th2/citologia , Células Th2/metabolismo , Vômito/etiologia , Adulto Jovem
5.
Artigo em Inglês | MEDLINE | ID: mdl-28229048

RESUMO

The obligate intracellular pathogen, Anaplasma phagocytophilum, is the causative agent of human, equine, and canine granulocytic anaplasmosis and tick-borne fever (TBF) in ruminants. A. phagocytophilum has become an emerging tick-borne pathogen in the United States, Europe, Africa, and Asia, with increasing numbers of infected people and animals every year. It has been recognized that intracellular pathogens manipulate host cell metabolic pathways to increase infection and transmission in both vertebrate and invertebrate hosts. However, our current knowledge on how A. phagocytophilum affect these processes in the tick vector, Ixodes scapularis is limited. In this study, a genome-wide search for components of major carbohydrate metabolic pathways was performed in I. scapularis ticks for which the genome was recently published. The enzymes involved in the seven major carbohydrate metabolic pathways glycolysis, gluconeogenesis, pentose phosphate, tricarboxylic acid cycle (TCA), glyceroneogenesis, and mitochondrial oxidative phosphorylation and ß-oxidation were identified. Then, the available transcriptomics and proteomics data was used to characterize the mRNA and protein levels of I. scapularis major carbohydrate metabolic pathway components in response to A. phagocytophilum infection of tick tissues and cultured cells. The results showed that major carbohydrate metabolic pathways are conserved in ticks. A. phagocytophilum infection inhibits gluconeogenesis and mitochondrial metabolism, but increases the expression of glycolytic genes. A model was proposed to explain how A. phagocytophilum could simultaneously control tick cell glucose metabolism and cytoskeleton organization, which may be achieved in part by up-regulating and stabilizing hypoxia inducible factor 1 alpha in a hypoxia-independent manner. The present work provides a more comprehensive view of the major carbohydrate metabolic pathways involved in the response to A. phagocytophilum infection in ticks, and provides the basis for further studies to develop novel strategies for the control of granulocytic anaplasmosis.


Assuntos
Anaplasmose/metabolismo , Metabolismo dos Carboidratos/genética , Interações Hospedeiro-Patógeno/genética , Ixodes/genética , Ixodes/metabolismo , Redes e Vias Metabólicas/genética , Anaplasma phagocytophilum/patogenicidade , Anaplasma phagocytophilum/fisiologia , Animais , Proteínas de Artrópodes/química , Proteínas de Artrópodes/genética , Proteínas de Artrópodes/metabolismo , Carboidratos , Linhagem Celular , Ciclo do Ácido Cítrico/genética , Feminino , Regulação da Expressão Gênica/fisiologia , Gluconeogênese/genética , Glicólise/genética , Ixodes/enzimologia , Ixodes/microbiologia , Mitocôndrias/genética , Mitocôndrias/metabolismo , Via de Pentose Fosfato/genética , Estrutura Terciária de Proteína , Proteômica/métodos , Glândulas Salivares/microbiologia , Transcriptoma/genética
7.
J Bacteriol ; 191(3): 693-700, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18978058

RESUMO

Anaplasma phagocytophilum, the etiologic agent of human granulocytic anaplasmosis (HGA), has genes predicted to encode three sensor kinases, one of which is annotated PleC, and three response regulators, one of which is PleD. Prior to this study, the roles of PleC and PleD in the obligatory intracellular parasitism of A. phagocytophilum and their biochemical activities were unknown. The present study illustrates the relevance of these factors by demonstrating that both pleC and pleD were expressed in an HGA patient. During A. phagocytophilum development in human promyelocytic HL-60 cells, PleC and PleD were synchronously upregulated at the exponential growth stage and downregulated prior to extracellular release. A recombinant PleC kinase domain (rPleCHKD) has histidine kinase activity; no activity was observed when the conserved site of phosphorylation was replaced with alanine. A recombinant PleD (rPleD) has autokinase activity using phosphorylated rPleCHKD as the phosphoryl donor but not with two other recombinant histidine kinases. rPleCHKD could not serve as the phosphoryl donor for a mutant rPleD (with a conserved aspartic acid, the site of phosphorylation, replaced by alanine) or two other A. phagocytophilum recombinant response regulators. rPleD had diguanylate cyclase activity to generate cyclic (c) di-GMP from GTP in vitro. UV cross-linking of A. phagocytophilum lysate with c-di-[(32)P]GMP detected an approximately 47-kDa endogenous protein, presumably c-di-GMP downstream receptor. A new hydrophobic c-di-GMP derivative, 2'-O-di(tert-butyldimethylsilyl)-c-di-GMP, inhibited A. phagocytophilum infection in HL-60 cells. Our results suggest that the two-component PleC-PleD system is a diguanylate cyclase and that a c-di-GMP-receptor complex regulates A. phagocytophilum intracellular infection.


Assuntos
Anaplasma phagocytophilum/enzimologia , Anaplasmose/metabolismo , Anaplasmose/patologia , GMP Cíclico/análogos & derivados , Fósforo-Oxigênio Liases/metabolismo , Proteínas Quinases/metabolismo , Anaplasmose/enzimologia , Western Blotting , GMP Cíclico/metabolismo , Ehrlichiose/enzimologia , Proteínas de Escherichia coli , Células HL-60 , Histidina Quinase , Humanos , Modelos Genéticos , Fósforo-Oxigênio Liases/genética , Fosforilação , Proteínas Quinases/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
J Immunol ; 179(7): 4783-91, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17878377

RESUMO

Anaplasma phagocytophilum is an obligate intracellular pathogen that resides within neutrophils and can cause fever, pancytopenia, or death. IFN-gamma plays a critical role in the control of A. phagocytophilum; however, the mechanisms that regulate IFN-gamma production remain unclear. In this study, we demonstrate that apoptotic specklike protein with a caspase-activating recruiting domain (ASC)/PYCARD, a central adaptor molecule in the Nod-like receptor (NLR) pathway, regulates the IL-18/IFN-gamma axis during A. phagocytophilum infection through its effect on caspase-1. Caspase-1- and asc-null mice were more susceptible than control animals to A. phagocytophilum infection due to the absence of IL-18 secretion and reduced IFN-gamma levels in the peripheral blood. Moreover, caspase-1 and ASC deficiency reduced CD4+ T cell-mediated IFN-gamma after in vitro restimulation with A. phagocytophilum. The NLR family member IPAF/NLRC4, but not NALP3/NLRP3, was partially required for IFN-gamma production in response to A. phagocytophilum. Taken together, our data demonstrate that ASC and caspase-1 are critical for IFN-gamma-mediated control of A. phagocytophilum infection.


Assuntos
Anaplasma/imunologia , Caspase 1/metabolismo , Interferon gama/metabolismo , Interleucina-18/metabolismo , Fagocitose , Motivos de Aminoácidos , Anaplasmose/imunologia , Anaplasmose/metabolismo , Anaplasmose/microbiologia , Anaplasmose/patologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Caspase 1/deficiência , Caspase 1/genética , Suscetibilidade a Doenças , Ativação Enzimática , Células HL-60 , Humanos , Interleucina-18/deficiência , Interleucina-18/genética , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Th1/imunologia
9.
BMC Infect Dis ; 7: 110, 2007 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-17883869

RESUMO

BACKGROUND: The tick-borne intracellular pathogen, Anaplasma phagocytophilum (Rickettsiales: Anaplasmataceae) causes human granulocytic anaplasmosis after infection of polymorphonuclear leucocytes. The human Sp110 gene is a member of the nuclear body (NB) components that functions as a nuclear hormone receptor transcriptional coactivator and plays an important role in immunoprotective mechanisms against pathogens in humans. In this research, we hypothesized that Sp110 may be involved in the infection of human promyelocytic HL-60 cells with A. phagocytophilum. METHODS: The human Sp110 and A. phagocytophilum msp4 mRNA levels were evaluated by real-time RT-PCR in infected human HL-60 cells sampled at 0, 12, 24, 48, 72 and 96 hours post-infection. The effect of Sp110 expression on A. phagocytophilum infection was determined by RNA interference (RNAi). The expression of Sp110 was silenced in HL-60 cells by RNAi using pre-designed siRNAs using the Nucleofector 96-well shuttle system (Amaxa Biosystems, Gaithersburg, MD, USA). The A. phagocytophilum infection levels were evaluated in HL-60 cells after RNAi by real-time PCR of msp4 and normalizing against human Alu sequences. RESULTS: While Sp110 mRNA levels increased concurrently with A. phagocytophilum infections in HL-60 cells, the silencing of Sp110 expression by RNA interference resulted in decreased infection levels. CONCLUSION: These results demonstrated that Sp110 expression is required for A. phagocytophilum infection and multiplication in HL-60 cells, and suggest a previously undescribed mechanism by which A. phagocytophilum modulates Sp110 mRNA levels to facilitate establishment of infection of human HL-60 cells.


Assuntos
Anaplasma phagocytophilum/genética , Anaplasmose/metabolismo , Células Precursoras de Granulócitos/metabolismo , Células Precursoras de Granulócitos/microbiologia , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Anaplasma phagocytophilum/metabolismo , Anaplasma phagocytophilum/patogenicidade , Anaplasmose/genética , Animais , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Células Precursoras de Granulócitos/fisiologia , Células HL-60 , Humanos , Antígenos de Histocompatibilidade Menor , Proteínas Nucleares/metabolismo , Interferência de RNA , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Transcrição Gênica , Zoonoses
10.
Vet Microbiol ; 91(2-3): 265-83, 2003 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-12458174

RESUMO

The major surface protein (MSP) 1a of the genus type species Anaplasma marginale (Rickettsiales: Anaplasmataceae) has been shown to mediate adhesion, infection and transmission of the organism, as well as to contribute to protective immunity in cattle. MSP1a contains a variable number of tandemly repeated peptides in the amino-terminal region, while the remainder of the protein is highly conserved among isolates. The number of repeats varies among geographic isolates of A. marginale but is constant within an isolate and has been used as a stable genetic marker of isolate identity. Because the sequence of the tandem repeats is the most variable part of the protein among isolates, this region of the protein is most likely to be involved in adhesion to host cells, a prerequisite to infection. The purpose of this study was to characterize the organization and function of the MSP1a tandem repeats of A. marginale in adhesion to host cells. We demonstrated by use of recombinant mutant proteins that the tandemly repeated region of MSP1a was necessary and sufficient to mediate adhesion of MSP1a to tick cells and bovine erythrocytes. Synthetic peptides representing the predominant sequences of individual repeats were tested for their adhesive capacity for tick cell extract (TCE). Peptides containing acidic amino acids D or E at position 20 bound to TCE, while peptides with a G as the 20th amino acid were not adhesive to TCE. Antibodies produced in rabbits against a synthetic repeat peptide neutralized A. marginale infection of cultured tick cells, and the neutralization observed was similar to that effected by antibodies produced against the whole MSP1a recombinant protein. Analysis of tandemly repeated MSP1a peptides of several geographic isolates of A. marginale revealed a complex relationship between the msp1alpha genotype and the tick-transmissible phenotype of the isolate and suggested that both the sequence and conformation of the repeated peptides influenced the adhesive properties of MSP1a. These studies demonstrated that the tandemly repeated region of the protein mediates the adhesive function of MSP1a.


Assuntos
Anaplasma/metabolismo , Aderência Bacteriana/fisiologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Doenças dos Bovinos/microbiologia , Sequência de Aminoácidos , Anaplasma/genética , Anaplasmose/metabolismo , Anaplasmose/microbiologia , Animais , Anticorpos Antibacterianos/imunologia , Aderência Bacteriana/genética , Proteínas da Membrana Bacteriana Externa/genética , Bovinos , Células Cultivadas/microbiologia , Eritrócitos/microbiologia , Escherichia coli/genética , Ixodes , Dados de Sequência Molecular , Mutagênese , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Análise de Sequência de DNA , Sequências de Repetição em Tandem/genética , Sequências de Repetição em Tandem/fisiologia
11.
Infect Immun ; 70(11): 6005-12, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12379676

RESUMO

The antigenically variant major surface protein 2 (MSP2) of Anaplasma marginale is expressed from a 3.5-kb operon that contains, in a 5'-to-3' direction, four open reading frames, opag3, opag2, opag1, and msp2. This operon structure was shown to be conserved among genotypically and phenotypically distinct A. marginale, A. ovis, and A. centrale strains. The individual OpAG amino acid sequences are highly conserved among A. marginale strains, with identities ranging from 95 to 99%. OpAG2 and OpAG3 were expressed by all examined A. marginale strains during the acute rickettsemia in the mammalian host and, like MSP2, localize to the bacterial surface. OpAG2 and OpAG3 were also expressed in an infected Ixodes scapularis tick cell line. In contrast, the same A. marginale strains expressed only OpAG2 in two different Dermacentor spp. during transmission feeding. OpAG1 expression was not detected in the infected mammalian host, the infected tick cell line, or within infected Dermacentor ticks. The differential expression of outer membrane proteins from within an operon is a novel finding in tick-transmitted bacteria, and the regulation of expression may be broadly applicable to understanding how the pathogen adapts to the mammalian host-tick vector transition.


Assuntos
Anaplasma/genética , Anaplasmose/metabolismo , Antígenos de Bactérias , Proteínas da Membrana Bacteriana Externa/genética , Óperon , Carrapatos/microbiologia , Sequência de Aminoácidos , Animais , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/imunologia , Células Cultivadas , Sequência Conservada , Regulação Bacteriana da Expressão Gênica , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular
12.
J Infect Dis ; 186(2): 274-80, 2002 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12134266

RESUMO

Anaplasma phagocytophila persists within neutrophils and prevents the respiratory burst by inhibiting gp91(phox). Mutations in gp91(phox) result in chronic granulomatous disease (CGD), which is diagnosed by use of the nitroblue tetrazolium (NBT) and Fc-Oxyburst assays that examine whether cells produce O2-. This study assessed whether the NBT and Fc-Oxyburst assays could detect a respiratory burst during A. phagocytophila infection. O2- production was inhibited in HL-60 cells and neutrophils infected with A. phagocytophila. In a mouse model of A. phagocytophila infection, 15%+/-4% (mean+/-SD) of polymorphonuclear leukocytes from infected mice had an ineffective respiratory burst compared with 1%+/-1% (mean+/-SD) of the neutrophils from uninfected animals. A population of neutrophils that did not produce O2- was also detected in 2 patients with A. phagocytophila infection. These data demonstrate respiratory burst inhibition by A. phagocytophila in vivo and on an individual cell basis by use of assays designed to evaluate CGD.


Assuntos
Anaplasma/metabolismo , Anaplasmose/metabolismo , Neutrófilos/metabolismo , Explosão Respiratória , Superóxidos/metabolismo , Anaplasmose/sangue , Animais , Células Cultivadas , Ehrlichiose/sangue , Ehrlichiose/diagnóstico , Ehrlichiose/metabolismo , Citometria de Fluxo , Células HL-60 , Humanos , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Camundongos SCID , Nitroazul de Tetrazólio , Superóxidos/sangue
13.
Infect Immun ; 66(3): 1200-7, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9488414

RESUMO

Anaplasma marginale is an intraerythrocytic rickettsial pathogen of cattle in which infection persists for the life of the animal. Persistent A. marginale infection is characterized by repetitive rickettsemic cycles which we hypothesize reflect emergence of A. marginale antigenic variants. In this study, we determined whether variants of major surface protein 2 (MSP-2), a target of protective immunity encoded by a polymorphic multigene family, arise during persistent rickettsemia. By using a quantitative competitive PCR to identify rickettsemic cycles, msp-2 transcripts expressed in vivo were isolated from peak rickettsemia of sequential cycles. Cloning and sequencing of msp-2 cDNA revealed that genetic variants of MSP-2 emerge representing a minimum of four genetic variant types in each cycle during persistent infection. Two-color immunofluorescence using variant-specific antibody showed that emergence of MSP-2 variants resulted in expression of a minimum of three antigenic types of MSP-2 within one rickettsemic cycle. Therefore immune control of each cycle would require responses to an antigenically diverse A. marginale population. These findings demonstrate that polymorphic MSP-2 variants emerge during cyclic rickettsemia in persistent A. marginale infection and suggest that emergent variants play an important role in persistence.


Assuntos
Anaplasmose/metabolismo , Bacteriemia/metabolismo , Proteínas de Bactérias/biossíntese , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/genética , Bovinos , Dados de Sequência Molecular , Polimorfismo Genético , RNA Mensageiro/análise
16.
J Bacteriol ; 93(3): 806-10, 1967 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-6025428

RESUMO

Extracts from preparations of partially purified Anaplasma marginale revealed low levels of lactate dehydrogenase (LDH). Enzyme inhibition by immune sera further indicated that A. marginale possesses a protein moiety the same as that of the normal red blood cell (RBC), although data suggested an alteration of LDH(1) from that observed in normal RBC. Bimodal isozyme distribution was detected after electrophoresis of the extracts. One isozyme approached the cathode and the other the anode, and both appeared to be nicotinamide adenine dinucleotide-dependent. Heterogeneity of parasite and host cell isozymes was established on the basis of zone electrophoresis on cellulose acetate strips.


Assuntos
Anaplasma/enzimologia , L-Lactato Desidrogenase/análise , Anaplasmose/metabolismo , Animais , Bovinos , Eletroforese , Soros Imunes/farmacologia , Isoenzimas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA