Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Front Immunol ; 15: 1390468, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38726006

RESUMO

Introduction: Relapsing fever (RF) remains a neglected human disease that is caused by a number of diverse pathogenic Borrelia (B.) species. Characterized by high cell densities in human blood, relapsing fever spirochetes have developed plentiful strategies to avoid recognition by the host defense mechanisms. In this scenario, spirochetal lipoproteins exhibiting multifunctional binding properties in the interaction with host-derived molecules are known to play a key role in adhesion, fibrinolysis and complement activation. Methods: Binding of CihC/FbpC orthologs to different human proteins and conversion of protein-bound plasminogen to proteolytic active plasmin were examined by ELISA. To analyze the inhibitory capacity of CihC/FbpC orthologs on complement activation, a microtiter-based approach was performed. Finally, AlphaFold predictions were utilized to identified the complement-interacting residues. Results and discussion: Here, we elucidate the binding properties of CihC/FbpC-orthologs from distinct RF spirochetes including B. parkeri, B. hermsii, B. turicatae, and B. recurrentis to human fibronectin, plasminogen, and complement component C1r. All CihC/FbpC-orthologs displayed similar binding properties to fibronectin, plasminogen, and C1r, respectively. Functional studies revealed a dose dependent binding of plasminogen to all borrelial proteins and conversion to active plasmin. The proteolytic activity of plasmin was almost completely abrogated by tranexamic acid, indicating that lysine residues are involved in the interaction with this serine protease. In addition, a strong inactivation capacity toward the classical pathway could be demonstrated for the wild-type CihC/FbpC-orthologs as well as for the C-terminal CihC fragment of B. recurrentis. Pre-incubation of human serum with borrelial molecules except CihC/FbpC variants lacking the C-terminal region protected serum-susceptible Borrelia cells from complement-mediated lysis. Utilizing AlphaFold2 predictions and existing crystal structures, we mapped the putative key residues involved in C1r binding on the CihC/FbpC orthologs attempting to explain the relatively small differences in C1r binding affinity despite the substitutions of key residues. Collectively, our data advance the understanding of the multiple binding properties of structural and functional highly similar molecules of relapsing fever spirochetes proposed to be involved in pathogenesis and virulence.


Assuntos
Proteínas de Bactérias , Borrelia , Fibrinólise , Plasminogênio , Ligação Proteica , Febre Recorrente , Humanos , Borrelia/imunologia , Borrelia/metabolismo , Febre Recorrente/microbiologia , Febre Recorrente/imunologia , Febre Recorrente/metabolismo , Plasminogênio/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Ativação do Complemento , Evasão da Resposta Imune , Aderência Bacteriana , Interações Hospedeiro-Patógeno/imunologia , Fibronectinas/metabolismo , Fibrinolisina/metabolismo , Proteínas do Sistema Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo
2.
J Biol Chem ; 299(8): 104972, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37380082

RESUMO

Borrelial pathogens are vector-borne etiological agents known to cause Lyme disease, relapsing fever, and Borrelia miyamotoi disease. These spirochetes each encode several surface-localized lipoproteins that bind components of the human complement system to evade host immunity. One borrelial lipoprotein, BBK32, protects the Lyme disease spirochete from complement-mediated attack via an alpha helical C-terminal domain that interacts directly with the initiating protease of the classical complement pathway, C1r. In addition, the B. miyamotoi BBK32 orthologs FbpA and FbpB also inhibit C1r, albeit via distinct recognition mechanisms. The C1r-inhibitory activities of a third ortholog termed FbpC, which is found exclusively in relapsing fever-causing spirochetes, remains unknown. Here, we report the crystal structure of the C-terminal domain of Borrelia hermsii FbpC to a limiting resolution of 1.5 Å. We used surface plasmon resonance and assays of complement function to demonstrate that FbpC retains potent BBK32-like anticomplement activities. Based on the structure of FbpC, we hypothesized that conformational dynamics of the complement inhibitory domains of borrelial C1r inhibitors may differ. To test this, we utilized the crystal structures of the C-terminal domains of BBK32, FbpA, FbpB, and FbpC to carry out molecular dynamics simulations, which revealed borrelial C1r inhibitors adopt energetically favored open and closed states defined by two functionally critical regions. Taken together, these results advance our understanding of how protein dynamics contribute to the function of bacterial immune evasion proteins and reveal a surprising plasticity in the structures of borrelial C1r inhibitors.


Assuntos
Proteínas de Bactérias , Borrelia , Proteínas Inativadoras do Complemento 1 , Doença de Lyme , Febre Recorrente , Humanos , Proteínas de Bactérias/química , Doença de Lyme/imunologia , Doença de Lyme/microbiologia , Febre Recorrente/imunologia , Febre Recorrente/microbiologia , Proteínas Inativadoras do Complemento 1/química , Domínios Proteicos , Cristalografia por Raios X
3.
Sci Rep ; 11(1): 5462, 2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33750855

RESUMO

Borrelia miyamotoi, a member of the tick-borne relapsing fever spirochetes, shows a serum-resistant phenotype in vitro. This ability of B. miyamotoi may contribute to bacterial evasion of the host innate immune system. To investigate the molecular mechanism of serum-resistance, we constructed a membrane protein-encoding gene library of B. miyamotoi using Borrelia garinii strain HT59G, which shows a transformable and serum-susceptible phenotype. By screening the library, we found that bom1093 and bom1515 of B. miyamotoi provided a serum-resistant phenotype to the recipient B. garinii. These B. miyamotoi genes are predicted to encode P35-like antigen genes and are conserved among relapsing fever borreliae. Functional analysis revealed that BOM1093 bound to serum vitronectin and that the C-terminal region of BOM1093 was involved in the vitronectin-binding property. Importantly, the B. garinii transformant was not serum-resistant when the C terminus-truncated BOM1093 was expressed. We also observed that the depletion of vitronectin from human serum enhances the bactericidal activity of BOM1093 expressing B. garinii, and the survival rate of BOM1093 expressing B. garinii in vitronectin-depleted serum is enhanced by the addition of purified vitronectin. Our data suggests that B. miyamotoi utilize BOM1093-mediated binding to vitronectin as a mechanism of serum resistance.


Assuntos
Proteínas de Bactérias/imunologia , Borrelia/imunologia , Febre Recorrente/imunologia , Vitronectina/imunologia , Humanos , Imunidade Inata , Soro/imunologia
4.
Front Immunol ; 11: 1560, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32793216

RESUMO

Relapsing fever (RF) is claimed a neglected arthropod-borne disease caused by a number of diverse human pathogenic Borrelia (B.) species. These RF borreliae are separated into the groups of tick-transmitted species including B. duttonii, B. hermsii, B. parkeri, B. turicatae, B. hispanica, B. persica, B. caucasica, and B. myiamotoi, and the louse-borne Borrelia species B. recurrentis. As typical blood-borne pathogens achieving high cell concentrations in human blood, RF borreliae (RFB) must outwit innate immunity, in particular complement as the first line of defense. One prominent strategy developed by RFB to evade innate immunity involves inactivation of complement by recruiting distinct complement regulatory proteins, e.g., C1 esterase inhibitor (C1-INH), C4b-binding protein (C4BP), factor H (FH), FH-like protein-1 (FHL-1), and factor H-related proteins FHR-1 and FHR-2, or binding of individual complement components and plasminogen, respectively. A number of multi-functional, complement and plasminogen-binding molecules from distinct Borrelia species have previously been identified and characterized, exhibiting considerable heterogeneity in their sequences, structures, gene localization, and their capacity to bind host-derived proteins. In addition, RFB possess a unique system of antigenic variation, allowing them to change the composition of surface-exposed variable major proteins, thus evading the acquired immune response of the human host. This review focuses on the current knowledge of the immune evasion strategies by RFB and highlights the role of complement-interfering and infection-associated molecules for the pathogenesis of RFB.


Assuntos
Borrelia/imunologia , Evasão da Resposta Imune/imunologia , Febre Recorrente/imunologia , Imunidade Adaptativa , Variação Antigênica , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Borrelia/patogenicidade , Proteínas do Sistema Complemento/metabolismo , Humanos , Imunidade Inata , Ligação Proteica , Febre Recorrente/microbiologia
5.
Clin Microbiol Infect ; 26(4): 513.e1-513.e6, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31404672

RESUMO

OBJECTIVES: Borrelia miyamotoi is a relapsing fever Borrelia, transmitted by hard (Ixodes) ticks, which are also the main vector for Borrelia burgdorferi. A widely used test for serodiagnosis of Lyme borreliosis is an enzyme immunoassay (EIA) based on the C6 peptide of the B. burgdorferi sl VlsE protein. We set out to study C6 reactivity upon infection with B. miyamotoi in a large well-characterized set of B. miyamotoi disease (BMD) patient sera and in experimental murine infection. METHODS: We performed in silico analyses, comparing the C6-peptide to immunodominant B. miyamotoi variable large proteins (Vlps). Next, we determined C6 reactivity in sera from mice infected with B. miyamotoi and in a unique longitudinal set of 191 sera from 46 BMD patients. RESULTS: In silico analyses revealed similarity of the C6 peptide to domains within B. miyamotoi Vlps. Cross-reactivity against the C6 peptide was confirmed in 21 out of 24 mice experimentally infected with B. miyamotoi. Moreover, 35 out of 46 BMD patients had a C6 EIA Lyme index higher than 1.1 (positive). Interestingly, 27 out of 37 patients with a C6 EIA Lyme index higher than 0.9 (equivocal) were negative when tested for specific B. burgdorferi sl antibodies using a commercially available immunoblot. CONCLUSIONS: We show that infection with B. miyamotoi leads to cross-reactive antibodies to the C6 peptide. Since BMD and Lyme borreliosis are found in the same geographical locations, caution should be used when relying solely on C6 reactivity testing. We propose that a positive C6 EIA with negative immunoblot, especially in patients with fever several weeks after a tick bite, warrants further testing for B. miyamotoi.


Assuntos
Anticorpos Antibacterianos/sangue , Proteínas de Bactérias/imunologia , Borrelia/imunologia , Reações Cruzadas , Doença de Lyme/imunologia , Febre Recorrente/imunologia , Animais , Simulação por Computador , Feminino , Humanos , Immunoblotting , Ixodes/microbiologia , Estudos Longitudinais , Doença de Lyme/diagnóstico , Camundongos , Camundongos Endogâmicos C3H , Peptídeos/imunologia , Kit de Reagentes para Diagnóstico , Febre Recorrente/diagnóstico , Testes Sorológicos
6.
J Immunol ; 204(2): 386-393, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31818980

RESUMO

The spirochete Borrelia miyamotoi has recently been shown to cause relapsing fever. Like the Lyme disease agent, Borrelia burgdorferi, B. miyamotoi is transmitted through the bite of infected ticks; however, little is known about the response of the immune system upon infection. Dendritic cells (DCs) play a central role in the early immune response against B. burgdorferi We investigated the response of DCs to two different strains of B. miyamotoi using in vitro and ex vivo models and compared this to the response elicited by B. burgdorferi. Our findings show that B. miyamotoi is phagocytosed by monocyte-derived DCs, causing upregulation of activation markers and production of proinflammatory cytokines in a similar manner to B. burgdorferi. Recognition of B. miyamotoi was demonstrated to be partially mediated by TLR2. DCs migrated out of human skin explants upon inoculation of the skin with B. miyamotoi. Finally, we showed that B. miyamotoi-stimulated DCs induced proliferation of naive CD4+ and CD8+ T cells to a larger extent than B. burgdorferi. In conclusion, we show in this study that DCs respond to and mount an immune response against B. miyamotoi that is similar to the response to B. burgdorferi and is able to induce T cell proliferation.


Assuntos
Borrelia/fisiologia , Células Dendríticas/imunologia , Mordeduras e Picadas de Insetos/imunologia , Febre Recorrente/imunologia , Pele/patologia , Linfócitos T/imunologia , Carrapatos/imunologia , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Citocinas/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Ativação Linfocitária , Fagocitose , Carrapatos/microbiologia , Receptor 2 Toll-Like/metabolismo
7.
Infect Immun ; 87(4)2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30642902

RESUMO

The global public health impact of relapsing fever (RF) spirochetosis is significant, since the pathogens exist on five of seven continents. The hallmark sign of infection is episodic fever and the greatest threat is to the unborn. With the goal of better understanding the specificity of B-cell responses and the role of immune responses in pathogenicity, we infected rhesus macaques with Borrelia turicatae (a new world RF spirochete species) by tick bite and monitored the immune responses generated in response to the pathogen. Specifically, we evaluated inflammatory mediator induction by the pathogen, host antibody responses to specific antigens, and peripheral lymphocyte population dynamics. Our results indicate that B. turicatae elicits from peripheral blood cells key inflammatory response mediators (interleukin-1ß and tumor necrosis factor alpha), which are associated with preterm abortion. Moreover, a global decline in peripheral B-cell populations was observed in all animals at 14 days postinfection. Serological responses were also evaluated to assess the antigenicity of three surface proteins: BipA, BrpA, and Bta112. Interestingly, a distinction was observed between antibodies generated in nonhuman primates and mice. Our results provide support for the nonhuman primate model not only in studies of prenatal pathogenesis but also for diagnostic and vaccine antigen identification and testing.


Assuntos
Anticorpos Antibacterianos/imunologia , Borrelia/fisiologia , Borrelia/patogenicidade , Febre Recorrente/imunologia , Febre Recorrente/microbiologia , Animais , Formação de Anticorpos , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Borrelia/genética , Borrelia/imunologia , Modelos Animais de Doenças , Humanos , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Macaca mulatta/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Febre Recorrente/diagnóstico , Febre Recorrente/transmissão , Carrapatos/microbiologia , Carrapatos/fisiologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Virulência
8.
J Infect Chemother ; 24(10): 828-833, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30057339

RESUMO

Since 2011, Borrelia miyamotoi disease (BMD) has been reported in five countries in the northern hemisphere. The causative agent of BMD is transmitted by Ixodes ticks, which are also vectors of Lyme disease borreliae. In this study, we examined 459 cases of clinically suspected Lyme disease (LD group), and found twelve cases that were seropositive for the glycerophosphodiester phosphodiesterase (GlpQ) antigen derived from B. miyamotoi. The retrospective surveillance revealed that the seroprevalence of anti-GlpQ in the LD group was significantly higher than in a healthy cohort. Seropositive cases were observed from spring through autumn when ticks are active, and the cases were geographically widespread, being found in Hokkaido-Tohoku, Kanto, Chubu, Kinki, and Kyushu-Okinawa regions. Seropositive cases for GlpQ were most frequent in the Chubu region (6.3%) where B. miyamotoi has been found in Ixodes ticks. Out of the twelve cases that were found in the LD group, three cases exhibited concomitant seropositivity to Lyme disease borreliae by western blot assay. This is the first report of serological surveillance for BMD in Japan, and we conclude that BMD occurs nationwide.


Assuntos
Borrelia/imunologia , Doença de Lyme/epidemiologia , Doença de Lyme/imunologia , Febre Recorrente/epidemiologia , Febre Recorrente/imunologia , Adolescente , Adulto , Idoso , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/sangue , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Estudos de Casos e Controles , Criança , Estudos de Coortes , DNA Bacteriano/genética , Feminino , Humanos , Japão/epidemiologia , Doença de Lyme/sangue , Doença de Lyme/diagnóstico , Masculino , Pessoa de Meia-Idade , Diester Fosfórico Hidrolases/sangue , Diester Fosfórico Hidrolases/genética , Diester Fosfórico Hidrolases/imunologia , Febre Recorrente/sangue , Febre Recorrente/diagnóstico , Estudos Retrospectivos , Estudos Soroepidemiológicos
9.
Yale J Biol Med ; 90(2): 195-218, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28656008

RESUMO

Relapsing fever agents like Borrelia hermsii undergo multiphasic antigenic variation that is attributable to spontaneous DNA non-reciprocal transpositions at a particular locus in the genome. This genetic switch results in a new protein being expressed on the cell surface, allowing cells with that phenotype to escape prevailing immunity. But the switch occurs in only one of several genomes in these spirochetes, and a newly-switched gene is effectively "recessive" until homozygosity is achieved. The longer that descendants of the switched cell expressed both old and new proteins, the longer this lineage risks neutralization by antibody to the old protein. We investigated the implications for antigenic variation of the phenotypic lag that polyploidy would confer on cells. We first experimentally determined the average genome copy number in daughter cells after division during mouse infection with B. hermsii strain HS1. We then applied discrete deterministic and stochastic simulations to predict outcomes when genomes were equably segregated either linearly, i.e. according to their position in one-dimensional arrays, or randomly partitioned, as for a sphere. Linear segregation replication provided for a lag in achievement of homozygosity that was significantly shorter than could be achieved under the random segregation condition. For cells with 16 genomes, this would be a 4-generation lag. A model incorporating the immune response and evolved matrices of switch rates indicated a greater fitness for polyploid over monoploid bacteria in terms of duration of infection.


Assuntos
Variação Antigênica/fisiologia , Borrelia/fisiologia , Animais , Variação Antigênica/genética , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Borrelia/citologia , Borrelia/genética , Borrelia/imunologia , Feminino , Genoma Bacteriano/genética , Camundongos , Camundongos SCID/microbiologia , Microscopia de Contraste de Fase , Reação em Cadeia da Polimerase , Poliploidia , Febre Recorrente/imunologia , Febre Recorrente/microbiologia
10.
J Immunol ; 196(10): 4185-95, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-27076681

RESUMO

Borrelia miyamotoi is a relapsing fever spirochete in Ixodes ticks that has been recently identified as a human pathogen causing hard tick-borne relapsing fever (HTBRF) across the Northern Hemisphere. No validated serologic test exists, and current serologic assays have low sensitivity in early HTBRF. To examine the humoral immune response against B. miyamotoi, we infected C3H/HeN mice with B. miyamotoi strain LB-2001 expressing variable small protein 1 (Vsp1) and demonstrated that spirochetemia was cleared after 3 d, coinciding with anti-Vsp1 IgM production. Clearance was also observed after passive transfer of immune sera to infected SCID mice. Next, we showed that anti-Vsp1 IgG eliminates Vsp1-expressing B. miyamotoi, selecting for spirochetes expressing a variable large protein (VlpC2) resistant to anti-Vsp1. The viability of Asian isolate B. miyamotoi HT31, expressing Vlp15/16 and Vlp18, was also unaffected by anti-Vsp1. Finally, in nine HTBRF patients, we demonstrated IgM reactivity to Vsp1 in two and against Vlp15/16 in four ∼1 wk after these patients tested positive for B. miyamotoi by PCR. Our data show that B. miyamotoi is able to express various variable major proteins (VMPs) to evade humoral immunity and that VMPs are antigenic in humans. We propose that serologic tests based on VMPs are of additional value in diagnosing HTBRF.


Assuntos
Anticorpos Antibacterianos/imunologia , Formação de Anticorpos , Proteínas da Membrana Bacteriana Externa/imunologia , Lipoproteínas/imunologia , Febre Recorrente/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antibacterianos/sangue , Sequência de Bases , Borrelia/imunologia , Feminino , Humanos , Imunização Passiva , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Imunoglobulina M/sangue , Imunoglobulina M/imunologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos SCID , Estrutura Terciária de Proteína
11.
PLoS Negl Trop Dis ; 10(2): e0004404, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26890814

RESUMO

Borrelia persica, a bacterium transmitted by the soft tick Ornithodoros tholozani, causes tick-borne relapsing fever in humans in the Middle East, Central Asia and the Indian peninsula. Immunocompetent C3H/HeOuJ mice were infected intradermally with B. persica at varying doses: 1 x 10(6), 1 x 10(4), 1 x 10(2) and 4 x 10(0) spirochetes/mouse. Subsequently, blood samples were collected and screened for the presence of B. persica DNA. Spirochetes were detected in all mice infected with 1 x 10(6), 1 x 10(4) and 1 x 10(2) borrelia by real-time PCR targeting the flaB gene of the bacterium. Spirochetemia developed with a one- to two-day delay when 1 x 10(4) and 1 x 10(2) borrelia were inoculated. Mice injected with only four organisms were negative in all tests. No clinical signs were observed when infected mice were compared to negative control animals. Organs (heart, spleen, urinary bladder, tarsal joint, skin and brain) were tested for B. persica-specific DNA and cultured for the detection of viable spirochetes. Compiled data show that the target organs of B. persica infections are the brain and the skin. A newly developed serological two-tiered test system (ELISA and western blot) for the detection of murine IgM, IgG and IgA antibody titers against B. persica showed a vigorous antibody response of the mice during infection. In conclusion, the infection model described here for B. persica is a platform for in vivo studies to decipher the so far unexplored survival strategies of this Borrelia species.


Assuntos
Borrelia/fisiologia , Modelos Animais de Doenças , Camundongos , Febre Recorrente/imunologia , Febre Recorrente/microbiologia , Animais , Anticorpos Antibacterianos/imunologia , Borrelia/química , Borrelia/crescimento & desenvolvimento , Borrelia/patogenicidade , Humanos , Hospedeiro Imunocomprometido , Cinética , Camundongos Endogâmicos C3H , Virulência
12.
Ticks Tick Borne Dis ; 5(6): 898-901, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25104575

RESUMO

Borrelia miyamotoi, a relapsing fever spirochete transmitted by ixodid ticks, is able to cause infections associated with systemic complaints, including malaise and fever, as well as meningoencephalitis in immunocompromised patients. In order to elucidate immune evasion of previously difficult to cultivate B. miyamotoi, we have examined the ability of this newly emerging human pathogen to escape the complement system. Growth inhibition assays revealed that B. miyamotoi is strongly resistant to complement-mediated bacteriolysis. Investigating complement activation, we found that B. miyamotoi showed reduced deposition of components C3, C5, C7, C8, C9 as well as the membrane attack complex (MAC) on the borrelial surface. In addition, no aberrations in cell morphology were observed after incubation of B. miyamotoi in active human serum, confirming the findings of the growth inhibition assay. The data presented here provide strong evidence that B. miyamotoi overcome human complement by affecting the central complement component C3, thereby inhibiting formation of the C3 convertase and downstream activation of the complement cascade.


Assuntos
Borrelia/imunologia , Complemento C3/imunologia , Febre Recorrente/imunologia , Animais , Borrelia/crescimento & desenvolvimento , Humanos , Febre Recorrente/virologia
13.
Infect Immun ; 82(8): 3324-32, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24866803

RESUMO

The primary causative agent of tick-borne relapsing fever in North America is Borrelia hermsii. It has been hypothesized that B. hermsii evades complement-mediated destruction by binding factor H (FH), a host-derived negative regulator of complement. In vitro, B. hermsii produces a single FH binding protein designated FhbA (FH binding protein A). The properties and ligand binding activity of FhbA suggest that it plays multiple roles in pathogenesis. It binds plasminogen and has been identified as a significant target of a B1b B cell-mediated IgM response in mice. FhbA has also been explored as a potential diagnostic antigen for B. hermsii infection in humans. The ability to test the hypothesis that FhbA is a critical virulence factor in vivo has been hampered by the lack of well-developed systems for the genetic manipulation of the relapsing fever spirochetes. In this report, we have successfully generated a B. hermsii fhbA deletion mutant (the B. hermsii YORΔfhbA strain) through allelic exchange mutagenesis. Deletion of fhbA abolished FH binding by the YORΔfhbA strain and eliminated cleavage of C3b on the cell surface. However, the YORΔfhbA strain remained infectious in mice and retained resistance to killing in vitro by human complement. Collectively, these results indicate that B. hermsii employs an FhbA/FH-independent mechanism of complement evasion that allows for resistance to killing by human complement and persistence in mice.


Assuntos
Atividade Bactericida do Sangue , Borrelia/imunologia , Proteínas de Transporte/metabolismo , Proteínas do Sistema Complemento/imunologia , Febre Recorrente/imunologia , Febre Recorrente/microbiologia , Fatores de Virulência/metabolismo , Animais , Borrelia/genética , Proteínas de Transporte/genética , Modelos Animais de Doenças , Deleção de Genes , Humanos , Camundongos , Fatores de Virulência/genética
14.
Infect Immun ; 82(6): 2520-31, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24686059

RESUMO

To identify and characterize surface proteins expressed by the relapsing fever (RF) agent Borrelia hermsii in the blood of infected mice, we used a cell-free filtrate of their blood to immunize congenic naive mice. The resultant antiserum was used for Western blotting of cell lysates, and gel slices corresponding to reactive bands were subjected to liquid chromatography-tandem mass spectrometry, followed by a search of the proteome database with the peptides. One of the immunogens was identified as the BHA007 protein, which is encoded by a 174-kb linear plasmid. BHA007 had sequence features of lipoproteins, was surface exposed by the criteria of in situ protease susceptibility and agglutination of Vtp(-) cells by anti-BHA007 antibodies, and was not essential for in vitro growth. BHA007 elicited antibodies during experimental infection of mice, but immunization with recombinant protein did not confer protection against needle-delivered infection. Open reading frames (ORFs) orthologous to BHA007 were found on large plasmids of other RF species, including the coding sequences for the CihC proteins of Borrelia duttonii and B. recurrentis, but not in Lyme disease Borrelia species. Recombinant BHA007 bound both human and bovine fibronectin with Kd (dissociation constant) values of 22 and 33 nM, respectively, and bound to C4-binding protein with less affinity. The distant homology of BHA007 and its orthologs to BBK32 proteins of Lyme disease species, as well as to previously described BBK32-like proteins in relapsing fever species, indicates that BHA007 is a member of a large family of multifunctional proteins in Borrelia species that bind to fibronectin as well as other host proteins.


Assuntos
Adesinas Bacterianas/metabolismo , Infecções por Borrelia/metabolismo , Febre Recorrente/metabolismo , Adesinas Bacterianas/genética , Adesinas Bacterianas/imunologia , Animais , Antígenos de Bactérias/análise , Borrelia , Infecções por Borrelia/imunologia , Modelos Animais de Doenças , Soros Imunes , Camundongos , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Febre Recorrente/imunologia , Análise de Sequência de DNA , Spirochaetales/metabolismo
15.
PLoS Pathog ; 10(4): e1004056, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24699793

RESUMO

Borrelia hermsii, a causative agent of relapsing fever of humans in western North America, is maintained in enzootic cycles that include small mammals and the tick vector Ornithodoros hermsi. In mammals, the spirochetes repeatedly evade the host's acquired immune response by undergoing antigenic variation of the variable major proteins (Vmps) produced on their outer surface. This mechanism prolongs spirochete circulation in blood, which increases the potential for acquisition by fast-feeding ticks and therefore perpetuation of the spirochete in nature. Antigenic variation also underlies the relapsing disease observed when humans are infected. However, most spirochetes switch off the bloodstream Vmp and produce a different outer surface protein, the variable tick protein (Vtp), during persistent infection in the tick salivary glands. Thus the production of Vmps in mammalian blood versus Vtp in ticks is a dominant feature of the spirochete's alternating life cycle. We constructed two mutants, one which was unable to produce a Vmp and the other was unable to produce Vtp. The mutant lacking a Vmp constitutively produced Vtp, was attenuated in mice, produced lower cell densities in blood, and was unable to relapse in animals after its initial spirochetemia. This mutant also colonized ticks and was infectious by tick-bite, but remained attenuated compared to wild-type and reconstituted spirochetes. The mutant lacking Vtp also colonized ticks but produced neither Vtp nor a Vmp in tick salivary glands, which rendered the spirochete noninfectious by tick bite. Thus the ability of B. hermsii to produce Vmps prolonged its survival in blood, while the synthesis of Vtp was essential for mammalian infection by the bite of its tick vector.


Assuntos
Variação Antigênica/imunologia , Borrelia/imunologia , Mutação , Ornithodoros/microbiologia , Febre Recorrente/imunologia , Febre Recorrente/transmissão , Animais , Variação Antigênica/genética , Borrelia/genética , Borrelia/patogenicidade , Camundongos , Camundongos SCID , Febre Recorrente/genética
16.
Yakugaku Zasshi ; 133(12): 1417-23, 2013.
Artigo em Japonês | MEDLINE | ID: mdl-24292191

RESUMO

Borrelia duttonii, a causative agent of relapsing fever, is transmitted between the distinct microenvironments of the vector tick, Ornithodoros moubata, and a mammalian host. We identified the total and membrane fraction proteins of B. duttonii strain Ly isolated from a patient in Tanzania by using two-dimensional gel electrophoresis with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS). The analyses of the total and membrane fractions from bacterial cultures incubated at 37°C identified 68 and 15 proteins, respectively. Since spirochaete clearance in mice is associated with an immunoglobulin M (IgM) and immunoglobulin G3 (IgG3)-mediated response, immunoblot analyses were used to identify the proteins reactive with IgM and IgG3 of gerbil serum against B. duttonii strain Ly. The outcome showed that six proteins (antigen p83/100, membrane-associated protein P66 (P66), flagellar filament outer layer protein, hypothetical protein BDU_412, vlp protein gamma subfamily (γ-Vlp) and flagellin (FlaB)) were identified against IgM, and four (antigen p83/100, P66, γ-Vlp and FlaB) of the six proteins also reacted with IgG3. It is believed that these proteins are immunodominant antigens for the host immune response. Some of these immunogenic proteins might be used as molecular diagnostic tools in the study of relapsing fever in Tanzania.


Assuntos
Proteínas da Membrana Bacteriana Externa/imunologia , Borrelia/imunologia , Febre Recorrente/microbiologia , Anticorpos Antibacterianos/imunologia , Borrelia/isolamento & purificação , Pré-Escolar , Feminino , Humanos , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Febre Recorrente/imunologia
17.
Vector Borne Zoonotic Dis ; 13(6): 376-81, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23488454

RESUMO

Tick-borne relapsing fever (TBRF) is endemic in mountainous regions of the western United States. In California, the principal agent is the spirochete Borrelia hermsii, which is transmitted by the argasid tick Ornithodoros hermsi. Humans are at risk of TBRF when infected ticks leave an abandoned rodent nest in quest of a blood meal. Rodents are the primary vertebrate hosts for B. hermsii. Sciurid rodents were collected from 23 sites in California between August, 2006, and September, 2008, and tested for serum antibodies to B. hermsii by immunoblot using a whole-cell sonicate and a specific antigen, glycerophosphodiester phosphodiesterase (GlpQ). Antibodies were detected in 20% of rodents; seroprevalence was highest (36%) in chipmunks (Tamias spp). Seroprevalence in chipmunks was highest in the Sierra Nevada (41%) and Mono (43%) ecoregions and between 1900 and 2300 meters elevation (43%). The serological studies described here are effective in implicating the primary vertebrate hosts involved in the maintenance of the ticks and spirochetes in regions endemic for TBRF.


Assuntos
Anticorpos Antibacterianos/sangue , Vetores Artrópodes/microbiologia , Borrelia/imunologia , Ornithodoros/microbiologia , Febre Recorrente/epidemiologia , Doenças dos Roedores/epidemiologia , Sciuridae/microbiologia , Animais , Proteínas de Bactérias/imunologia , Borrelia/isolamento & purificação , California/epidemiologia , Humanos , Diester Fosfórico Hidrolases/imunologia , Febre Recorrente/imunologia , Febre Recorrente/microbiologia , Doenças dos Roedores/microbiologia , Roedores , Estudos Soroepidemiológicos , Testes Sorológicos
18.
Front Biosci (Schol Ed) ; 4(2): 699-712, 2012 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-22202086

RESUMO

The bacteria of the genus Borrelia are arthropod-borne spirochetes that cause relapsing fever and Lyme disease in humans. Like most arthropod-borne pathogens, Borreliae must survive in the periphery of their vertebrate hosts to allow for transmission to another arthropod vector. These spatial and temporal restrictions require that Borreliae evade the adaptive immune response. Borreliae have evolved genetic mechanisms that alter their surface protein expression, thereby altering the antigenic target presented to the host. To control Borreliae infection, the host relies on a rapid humoral response. While it is clear that B cell antigen receptor signaling is a critical requirement for the specific antibody responses, growing evidence suggests that additional signaling by innate immune receptors such as Toll-like receptors is necessary for optimal T cell-dependent and T cell-independent antibody responses. This review is focused on the role of Toll-like receptors in B cell responses to relapsing fever and Lyme disease Borreliae.


Assuntos
Infecções por Borrelia/imunologia , Borrelia/imunologia , Doença de Lyme/imunologia , Febre Recorrente/imunologia , Receptores Toll-Like/imunologia , Animais , Linfócitos B/imunologia , Infecções por Borrelia/microbiologia , Humanos , Doença de Lyme/microbiologia , Receptores de Antígenos de Linfócitos B/imunologia , Febre Recorrente/microbiologia
19.
Vet Microbiol ; 148(2-4): 341-7, 2011 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-20970929

RESUMO

Binding of complement factor H is crucial for the resistance of Borrelia to complement-mediated lysis. This study was aimed to assess the correlation between the expression of fH binding proteins (FHBPs) during the early phase of infection (48 h after the entry of Borrelia into the blood circulation) and complement resistance of the Borrelia genus. As expected, B. afzelii, B. burgdorferi sensu stricto and B. garinii (Serotype 4, PBi) showed resistance to complement mediated lysis when incubated with human and dog complement, which coincided with the significantly higher expression (P<0.05) of the FHBPs. Similarly, B. coriaceae showed resistance to cattle complement. In non-reservoir hosts borreliae failed to induce expression of FHBPs within 48 h of complement challenge, and did not survive. It is important to note that not only the expression of FHBP but also their binding to fH is required for borrelial resistance to the complement. fH binding may depend on the coiled-coil (CC) motifs observed in the FHBPs, especially at the C terminus. A loss of the C-terminal CC motif in BgCRASP-1 of SKT-1 strain was found in in-silico CC prediction, and may be coupled with SKT-1's inability to bind factor H and evade complement-mediated attack. In contrast, the C-terminal CC motif was observed (P - 1.0) in BgCRASP-1 of PBi that may contributed to its factor H binding and human complement resistance.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Grupo Borrelia Burgdorferi/metabolismo , Fator H do Complemento/metabolismo , Sequência de Aminoácidos , Animais , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Grupo Borrelia Burgdorferi/imunologia , Bovinos , Fator H do Complemento/imunologia , Cães , Humanos , Doença de Lyme/imunologia , Doença de Lyme/microbiologia , Dados de Sequência Molecular , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Febre Recorrente/imunologia , Febre Recorrente/microbiologia , Alinhamento de Sequência
20.
Infect Immun ; 78(11): 4579-88, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20696824

RESUMO

Overwhelming bacteremia is a leading cause of death. To understand the mechanisms involved in protective antibody and pathological inflammatory responses during bacteremia, we have been studying the murine model of Borrelia hermsii infection. Toll-like receptor (TLR) signaling plays an important role in generating the rapid anti-B. hermsii antibody responses required for the resolution of bacteremia. Using NF-κB reporter assays, we found that B. hermsii activates TLR2 and TLR9. However, TLR2(-/-) TLR9(-/-) mice exhibited an impairment in anti-B. hermsii antibody responses similar to that of TLR2(-/-) mice. Moreover, the impairment in the antibody responses of TLR2(-/-) mice or TLR2(-/-) TLR9(-/-) mice coincides with an order-of-magnitude-higher bacteremia, and death results from septic shock, as evidenced by a dysregulated systemic cytokine response and characteristic organ pathology. Since TLR2 appears to be the major extracellular sensor stimulated by B. hermsii, we hypothesized that during elevated bacteremia the activation of intracellular sensors of bacteria triggers dysregulated inflammation in TLR2(-/-) mice. Indeed, blocking the internalization of B. hermsii prevented the induction of inflammatory cytokine responses in TLR2-deficient cells. Furthermore, we found that B. hermsii activates the cytoplasmic sensor nucleotide-binding oligomerization domain 2 (NOD2). Macrophages deficient in both TLR2 and NOD2 have impaired cytokine responses to B. hermsii compared to cells lacking TLR2 alone, and B. hermsii-infected TLR2(-/-) NOD2(-/-) mice exhibited improved survival compared to TLR2(-/-) mice. These data demonstrate that TLR2 is critical for protective immunity and suggest that, during heightened bacteremia, recognition of bacterial components by intracellular sensors can lead to pathological inflammatory responses.


Assuntos
Formação de Anticorpos/imunologia , Borrelia/patogenicidade , Febre Recorrente/imunologia , Choque Séptico/imunologia , Receptor 2 Toll-Like/deficiência , Animais , Anticorpos Antibacterianos/sangue , Bacteriemia/imunologia , Bacteriemia/microbiologia , Bacteriemia/patologia , Borrelia/classificação , Borrelia/imunologia , Linhagem Celular , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Adaptadora de Sinalização NOD2/genética , Proteína Adaptadora de Sinalização NOD2/metabolismo , Febre Recorrente/microbiologia , Febre Recorrente/patologia , Choque Séptico/microbiologia , Transdução de Sinais , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...