Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Inflammation ; 47(1): 346-362, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37831367

RESUMO

Infectious diseases are a significant burden in global healthcare. Pathogens engage with different host defense mechanisms. However, it is currently unknown if there are disease-specific immune signatures and/or if different pathogens elicit common immune-associated molecular entities to common therapeutic interventions. We studied patients enrolled through the Human Immunology Project Consortium (HIPC), which focuses on immune responses to various infections. Blood samples were collected and analyzed from patients during infection and follow-up time points at the convalescent stage. The study included samples from patients with Lyme disease (LD), tuberculosis (TB), malaria (MLA), dengue virus (DENV), and West Nile virus (WNV), as well as kidney transplant patients with cytomegalovirus (CMV) and polyomavirus (BKV) infections. Using an antibody-based assay, we quantified ~ 350 cell surface markers, cytokines, and chemokines involved in inflammation and immunity. Unique protein signatures were identified specific to the acute phase of infection irrespective of the pathogen type, with significant changes during convalescence. In addition, tumor necrosis factor receptor superfamily member 6 (TNR6), C-C Motif Chemokine Receptor 7 (CCR7), and C-C motif chemokine ligand-1 (CCL1) were increased in the acute and convalescent phases across all viral, bacterial, and protozoan compared to blood from healthy donors. Furthermore, despite the differences between pathogens, proteins were enriched in common biological pathways such as cell surface receptor signaling pathway and response to external stimulus. In conclusion, we demonstrated that irrespective of the pathogen type, there are common immunoregulatory and proinflammatory signals.


Assuntos
Proteoma , Vírus do Nilo Ocidental , Humanos , Inflamação , Citocinas , Transdução de Sinais/fisiologia
2.
Front Immunol ; 13: 1012824, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36569838

RESUMO

Advancement in proteomics methods for interrogating biological samples has helped identify disease biomarkers for early diagnostics and unravel underlying molecular mechanisms of disease. Herein, we examined the serum proteomes of 23 study participants presenting with one of two common arthropod-borne infections: Lyme disease (LD), an extracellular bacterial infection or West Nile virus infection (WNV), an intracellular viral infection. The LC/MS based serum proteomes of samples collected at the time of diagnosis and during convalescence were assessed using a depletion-based high-throughput shotgun proteomics (dHSP) pipeline as well as a non-depleting blotting-based low-throughput platform (MStern). The LC/MS integrated analyses identified host proteome responses in the acute and recovery phases shared by LD and WNV infections, as well as differentially abundant proteins that were unique to each infection. Notably, we also detected proteins that distinguished localized from disseminated LD and asymptomatic from symptomatic WNV infection. The proteins detected in both diseases with the dHSP pipeline identified unique and overlapping proteins detected with the non-depleting MStern platform, supporting the utility of both detection methods. Machine learning confirmed the use of the serum proteome to distinguish the infection from healthy control sera but could not develop discriminatory models between LD and WNV at current sample numbers. Our study is the first to compare the serum proteomes in two arthropod-borne infections and highlights the similarities in host responses even though the pathogens and the vectors themselves are different.


Assuntos
Doença de Lyme , Febre do Nilo Ocidental , Vírus do Nilo Ocidental , Humanos , Febre do Nilo Ocidental/diagnóstico , Vírus do Nilo Ocidental/fisiologia , Proteoma , Proteômica , Doença de Lyme/diagnóstico
3.
JCI Insight ; 6(12)2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34061047

RESUMO

The skin lesion erythema migrans (EM) is an initial sign of the Ixodes tick-transmitted Borreliella spirochetal infection known as Lyme disease. T cells and innate immune cells have previously been shown to predominate the EM lesion and promote the reaction. Despite the established importance of B cells and antibodies in preventing infection, the role of B cells in the skin immune response to Borreliella is unknown. Here, we used single-cell RNA-Seq in conjunction with B cell receptor (BCR) sequencing to immunophenotype EM lesions and their associated B cells and BCR repertoires. We found that B cells were more abundant in EM in comparison with autologous uninvolved skin; many were clonally expanded and had circulating relatives. EM-associated B cells upregulated the expression of MHC class II genes and exhibited preferential IgM isotype usage. A subset also exhibited low levels of somatic hypermutation despite a gene expression profile consistent with memory B cells. Our study demonstrates that single-cell gene expression with paired BCR sequencing can be used to interrogate the sparse B cell populations in human skin and reveals that B cells in the skin infection site in early Lyme disease expressed a phenotype consistent with local antigen presentation and antibody production.


Assuntos
Linfócitos B , Eritema Migrans Crônico , Imunofenotipagem/métodos , Análise de Célula Única/métodos , Adulto , Idoso , Linfócitos B/citologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Eritema Migrans Crônico/imunologia , Eritema Migrans Crônico/patologia , Feminino , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Imunoglobulina M/genética , Imunoglobulina M/metabolismo , Doença de Lyme , Masculino , Pessoa de Meia-Idade , RNA-Seq , Pele/citologia , Transcriptoma/genética , Transcriptoma/imunologia
4.
Clin Infect Dis ; 72(1): e1-e48, 2021 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-33417672

RESUMO

This evidence-based clinical practice guideline for the prevention, diagnosis, and treatment of Lyme disease was developed by a multidisciplinary panel representing the Infectious Diseases Society of America (IDSA), the American Academy of Neurology (AAN), and the American College of Rheumatology (ACR). The scope of this guideline includes prevention of Lyme disease, and the diagnosis and treatment of Lyme disease presenting as erythema migrans, Lyme disease complicated by neurologic, cardiac, and rheumatologic manifestations, Eurasian manifestations of Lyme disease, and Lyme disease complicated by coinfection with other tick-borne pathogens. This guideline does not include comprehensive recommendations for babesiosis and tick-borne rickettsial infections, which are published in separate guidelines. The target audience for this guideline includes primary care physicians and specialists caring for this condition such as infectious diseases specialists, emergency physicians, internists, pediatricians, family physicians, neurologists, rheumatologists, cardiologists and dermatologists in North America.


Assuntos
Doenças Transmissíveis , Doença de Lyme , Neurologia , Reumatologia , Animais , Humanos , Doença de Lyme/diagnóstico , Doença de Lyme/tratamento farmacológico , Doença de Lyme/prevenção & controle , América do Norte , Estados Unidos
5.
Clin Infect Dis ; 72(1): 1-8, 2021 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-33483734

RESUMO

This evidence-based clinical practice guideline for the prevention, diagnosis, and treatment of Lyme disease was developed by a multidisciplinary panel representing the Infectious Diseases Society of America (IDSA), the American Academy of Neurology (AAN), and the American College of Rheumatology (ACR). The scope of this guideline includes prevention of Lyme disease, and the diagnosis and treatment of Lyme disease presenting as erythema migrans, Lyme disease complicated by neurologic, cardiac, and rheumatologic manifestations, Eurasian manifestations of Lyme disease, and Lyme disease complicated by coinfection with other tick-borne pathogens. This guideline does not include comprehensive recommendations for babesiosis and tick-borne rickettsial infections, which are published in separate guidelines. The target audience for this guideline includes primary care physicians and specialists caring for this condition such as infectious diseases specialists, emergency physicians, internists, pediatricians, family physicians, neurologists, rheumatologists, cardiologists and dermatologists in North America.


Assuntos
Doenças Transmissíveis , Doença de Lyme , Neurologia , Reumatologia , Animais , Humanos , Doença de Lyme/diagnóstico , Doença de Lyme/tratamento farmacológico , Doença de Lyme/prevenção & controle , América do Norte , Estados Unidos
6.
Neurology ; 96(6): 262-273, 2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33257476

RESUMO

This evidence-based clinical practice guideline for the prevention, diagnosis, and treatment of Lyme disease was developed by a multidisciplinary panel representing the Infectious Diseases Society of America (IDSA), the American Academy of Neurology (AAN), and the American College of Rheumatology (ACR). The scope of this guideline includes prevention of Lyme disease, and the diagnosis and treatment of Lyme disease presenting as erythema migrans, Lyme disease complicated by neurologic, cardiac, and rheumatologic manifestations, Eurasian manifestations of Lyme disease, and Lyme disease complicated by coinfection with other tick-borne pathogens. This guideline does not include comprehensive recommendations for babesiosis and tick-borne rickettsial infections, which are published in separate guidelines. The target audience for this guideline includes primary care physicians and specialists caring for this condition such as infectious diseases specialists, emergency physicians, internists, pediatricians, family physicians, neurologists, rheumatologists, cardiologists and dermatologists in North America.


Assuntos
Doença de Lyme/diagnóstico , Doença de Lyme/terapia , Guias de Prática Clínica como Assunto/normas , Sociedades Médicas/normas , Humanos , Doença de Lyme/prevenção & controle , Estados Unidos
8.
Arthritis Rheumatol ; 73(1): 12-20, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33251716
9.
Curr Issues Mol Biol ; 42: 145-190, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33289684

RESUMO

The mammalian host responds to infection with Borrelia spirochetes through a highly orchestrated immune defense involving innate and adaptive effector functions aimed toward limiting pathogen burdens, minimizing tissue injury, and preventing subsequent reinfection. The evolutionary adaptation of Borrelia spirochetes to their reservoir mammalian hosts may allow for its persistence despite this immune defense. This review summarizes our current understanding of the host immune response to B. burgdorferi sensu lato, the most widely studied Borrelia spp. and etiologic agent of Lyme borreliosis. Pertinent literature will be reviewed with emphasis on in vitro, ex vivo and animal studies that influenced our understanding of both the earliest responses to B. burgdorferi as it enters the mammalian host and those that evolve as spirochetes disseminate and establish infection in multiple tissues. Our focus is on the immune response of inbred mice, the most commonly studied animal model of B. burgdorferi infection and surrogate for one of this pathogen's principle natural reservoir hosts, the white-footed deer mouse. Comparison will be made to the immune responses of humans with Lyme borreliosis. Our goal is to provide an understanding of the dynamics of the mammalian immune response during infection with B. burgdorferi and its relation to the outcomes in reservoir (mouse) and non-reservoir (human) hosts.


Assuntos
Infecções por Borrelia/imunologia , Infecções por Borrelia/microbiologia , Borrelia/imunologia , Interações Hospedeiro-Patógeno/imunologia , Animais , Evolução Biológica , Infecções por Borrelia/transmissão , Reservatórios de Doenças , Humanos , Doença de Lyme/imunologia , Doença de Lyme/microbiologia , Doença de Lyme/transmissão , Especificidade de Órgãos/imunologia
10.
Front Microbiol ; 10: 1923, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31507550

RESUMO

Maintenance of Borrelia burgdorferi within its enzootic cycle requires a complex regulatory pathway involving the alternative σ factors RpoN and RpoS and two ancillary trans-acting factors, BosR and Rrp2. Activation of this pathway occurs within ticks during the nymphal blood meal when RpoS, the effector σ factor, transcribes genes required for tick transmission and mammalian infection. RpoS also exerts a 'gatekeeper' function by repressing σ70-dependent tick phase genes (e.g., ospA, lp6.6). Herein, we undertook a broad examination of RpoS functionality throughout the enzootic cycle, beginning with modeling to confirm that this alternative σ factor is a 'genuine' RpoS homolog. Using a novel dual color reporter system, we established at the single spirochete level that ospA is expressed in nymphal midguts throughout transmission and is not downregulated until spirochetes have been transmitted to a naïve host. Although it is well established that rpoS/RpoS is expressed throughout infection, its requirement for persistent infection has not been demonstrated. Plasmid retention studies using a trans-complemented ΔrpoS mutant demonstrated that (i) RpoS is required for maximal fitness throughout the mammalian phase and (ii) RpoS represses tick phase genes until spirochetes are acquired by a naïve vector. By transposon mutant screening, we established that bba34/oppA5, the only OppA oligopeptide-binding protein controlled by RpoS, is a bona fide persistence gene. Lastly, comparison of the strain 297 and B31 RpoS DMC regulons identified two cohorts of RpoS-regulated genes. The first consists of highly conserved syntenic genes that are similarly regulated by RpoS in both strains and likely required for maintenance of B. burgdorferi sensu stricto strains in the wild. The second includes RpoS-regulated plasmid-encoded variable surface lipoproteins ospC, dbpA and members of the ospE/ospF/elp, mlp, revA, and Pfam54 paralogous gene families, all of which have evolved via inter- and intra-strain recombination. Thus, while the RpoN/RpoS pathway regulates a 'core' group of orthologous genes, diversity within RpoS regulons of different strains could be an important determinant of reservoir host range as well as spirochete virulence.

11.
Proc Natl Acad Sci U S A ; 116(27): 13498-13507, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31209025

RESUMO

Lyme disease is a multisystem disorder caused by the spirochete Borrelia burgdorferi A common late-stage complication of this disease is oligoarticular arthritis, often involving the knee. In ∼10% of cases, arthritis persists after appropriate antibiotic treatment, leading to a proliferative synovitis typical of chronic inflammatory arthritides. Here, we provide evidence that peptidoglycan (PG), a major component of the B. burgdorferi cell envelope, may contribute to the development and persistence of Lyme arthritis (LA). We show that B. burgdorferi has a chemically atypical PG (PGBb) that is not recycled during cell-wall turnover. Instead, this pathogen sheds PGBb fragments into its environment during growth. Patients with LA mount a specific immunoglobulin G response against PGBb, which is significantly higher in the synovial fluid than in the serum of the same patient. We also detect PGBb in 94% of synovial fluid samples (32 of 34) from patients with LA, many of whom had undergone oral and intravenous antibiotic treatment. These same synovial fluid samples contain proinflammatory cytokines, similar to those produced by human peripheral blood mononuclear cells stimulated with PGBb In addition, systemic administration of PGBb in BALB/c mice elicits acute arthritis. Altogether, our study identifies PGBb as a likely contributor to inflammatory responses in LA. Persistence of this antigen in the joint may contribute to synovitis after antibiotics eradicate the pathogen. Furthermore, our finding that B. burgdorferi sheds immunogenic PGBb fragments during growth suggests a potential role for PGBb in the immunopathogenesis of other Lyme disease manifestations.


Assuntos
Antígenos de Bactérias/imunologia , Borrelia burgdorferi/imunologia , Doença de Lyme/imunologia , Peptidoglicano/imunologia , Imunidade Adaptativa/imunologia , Animais , Citocinas/metabolismo , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Peptidoglicano/análise , Peptidoglicano/química , Líquido Sinovial/química , Líquido Sinovial/imunologia
13.
Ticks Tick Borne Dis ; 10(3): 682-689, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30846418

RESUMO

Borrelia miyamotoi is a relapsing fever spirochete transmitted by ticks in the Ixodes ricinus complex. In the eastern United States, B. miyamotoi is transmitted by I. scapularis, which also vectors several other pathogens including B. burgdorferi sensu stricto. In contrast to Lyme borreliae, B. miyamotoi can be transmitted vertically from infected female ticks to their progeny. Therefore, in addition to nymphs and adults, larvae can vector B. miyamotoi to wildlife and human hosts. Two widely varying filial infection prevalence (FIP) estimates - 6% and 73% - have been reported previously from two vertically infected larval clutches; to our knowledge, no other estimates of FIP or transovarial transmission (TOT) rates for B. miyamotoi have been described in the literature. Thus, we investigated TOT and FIP of larval clutches derived from engorged females collected from hunter-harvested white-tailed deer in 2015 (n = 664) and 2016 (n = 599) from Maine, New Hampshire, Tennessee, and Wisconsin. After engorged females oviposited in the lab, they (n = 492) were tested for B. miyamotoi infection by PCR. Subsequently, from each clutch produced by an infected female, larval pools, as well as 100 individual eggs or larvae, were tested. The TOT rate of the 11 infected females was 90.9% (95% CI; 57.1-99.5%) and the mean FIP of the resulting larval clutches was 84.4% (95% CI; 68.1-100%). Even though the overall observed vertical transmission rate (the product of TOT and FIP; 76.7%, 95% CI; 44.6-93.3%) was high, additional horizontal transmission may be required for enzootic maintenance of B. miyamotoi based on the results of a previously published deterministic model. Further investigation of TOT and FIP variability and the underlying mechanisms, both in nature and the laboratory, will be needed to resolve this question. Meanwhile, studies quantifying the acarological risk of Borrelia miyamotoi disease need to consider not only nymphs and adults, but larval I. scapularis as well.


Assuntos
Infecções por Borrelia/veterinária , Borrelia/isolamento & purificação , Cervos/parasitologia , Transmissão Vertical de Doenças Infecciosas , Ixodes/microbiologia , Animais , Animais Selvagens/parasitologia , Borrelia/genética , Infecções por Borrelia/epidemiologia , Infecções por Borrelia/transmissão , Feminino , Larva/microbiologia , Maine/epidemiologia , New Hampshire/epidemiologia , Ninfa/microbiologia , Reação em Cadeia da Polimerase , Tennessee/epidemiologia
14.
Methods Mol Biol ; 1690: 279-290, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29032551

RESUMO

Two-photon intravital microscopy is a powerful tool that allows visualization of cells in intact tissues in a live animal in real time. In recent years, this advanced technology has been applied to understand pathogen-host interactions using fluorescently labeled bacteria. In particular, infectious fluorescent transformants of the Lyme disease spirochete Borrelia burgdorferi, an Ixodes tick-transmitted pathogen, have been imaged by two-photon intravital microscopy to study bacterial motility and interactions of the pathogen with feeding ticks and host tissues. Here, we describe the techniques and equipment used to image mammalian-adapted spirochetes in the skin of living mice in vivo and in joints ex vivo using two-photon intravital microscopy.


Assuntos
Borrelia burgdorferi/fisiologia , Interações Hospedeiro-Patógeno , Microscopia Intravital/métodos , Doença de Lyme/microbiologia , Doença de Lyme/patologia , Pele/microbiologia , Pele/patologia , Animais , Vetores Aracnídeos/microbiologia , Vetores Aracnídeos/fisiologia , Borrelia burgdorferi/isolamento & purificação , Modelos Animais de Doenças , Ixodes/microbiologia , Ixodes/fisiologia , Camundongos , Camundongos Endogâmicos C57BL
15.
Biophys J ; 112(4): 746-754, 2017 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-28256234

RESUMO

Borrelia burgdorferi, the spirochete that causes Lyme disease, is a tick-transmitted pathogen that requires motility to invade and colonize mammalian and tick hosts. These bacteria use a unique undulating flat-wave shape to penetrate and propel themselves through host tissues. Previous mathematical modeling has suggested that the morphology and motility of these spirochetes depends crucially on the flagellar/cell wall stiffness ratio. Here, we test this prediction using the antibiotic vancomycin to weaken the cell wall. We found that low to moderate doses of vancomycin (≤2.0 µg/mL for 24 h) produced small alterations in cell shape and that as the dose was increased, cell speed decreased. Vancomycin concentrations >1.0 µg/mL also inhibited cell growth and led to bleb formation on a fraction of the cells. To quantitatively assess how vancomycin affects cell stiffness, we used optical traps to bend unflagellated mutants of B. burgdorferi. We found that in the presence of vancomycin, cell wall stiffness gradually decreased over time, with a 40% reduction in the bending stiffness after 36 h. Under the same conditions, the swimming speed of wild-type B. burgdorferi slowed by ∼15%, with only marginal changes to cell morphology. Interestingly, our biophysical model for the swimming dynamics of B. burgdorferi suggested that cell speed should increase with decreasing cell stiffness. We show that this discrepancy can be resolved if the periplasmic volume decreases as the cell wall becomes softer. These results provide a testable hypothesis for how alterations of cell wall stiffness affect periplasmic volume regulation. Furthermore, since motility is crucial to the virulence of B. burgdorferi, the results suggest that sublethal doses of antibiotics could negatively impact spirochete survival by impeding their swim speed, thereby enabling their capture and elimination by phagocytes.


Assuntos
Antibacterianos/farmacologia , Borrelia burgdorferi/efeitos dos fármacos , Parede Celular/efeitos dos fármacos , Doença de Lyme/microbiologia , Fenômenos Mecânicos/efeitos dos fármacos , Movimento/efeitos dos fármacos , Vancomicina/farmacologia , Fenômenos Biomecânicos/efeitos dos fármacos , Borrelia burgdorferi/citologia , Borrelia burgdorferi/metabolismo , Borrelia burgdorferi/fisiologia
16.
J Exp Med ; 213(7): 1307-18, 2016 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-27270894

RESUMO

Human babesiosis is a tick-borne multisystem disease caused by Babesia species of the apicomplexan phylum. Most clinical cases and fatalities of babesiosis are caused by Babesia microti Current treatment for human babesiosis consists of two drug combinations, atovaquone + azithromycin or quinine + clindamycin. These treatments are associated with adverse side effects and a significant rate of drug failure. Here, we provide evidence for radical cure of experimental babesiosis in immunodeficient mice using a combination of an endochin-like quinolone (ELQ) prodrug and atovaquone. In vivo efficacy studies in mice using ELQ-271, ELQ-316, and the ELQ-316 prodrug, ELQ-334, demonstrated excellent growth inhibitory activity against the parasite, with potency equal to that of orally administered atovaquone at 10 mg/kg. Analysis of recrudescent parasites after ELQ or atovaquone monotherapy identified genetic substitutions in the Qi or Qo sites, respectively, of the cytochrome bc1 complex. Impressively, a combination of ELQ-334 and atovaquone, at doses as low as 5.0 mg/kg each, resulted in complete clearance of the parasite with no recrudescence up to 122 d after discontinuation of therapy. These results will set the stage for future clinical evaluation of ELQ and atovaquone combination therapy for treatment of human babesiosis.


Assuntos
Atovaquona/farmacologia , Babesia microti/imunologia , Babesiose/tratamento farmacológico , Síndromes de Imunodeficiência/parasitologia , Pró-Fármacos/farmacologia , Quinolonas/farmacologia , Animais , Babesiose/genética , Babesiose/imunologia , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , Camundongos , Camundongos SCID
17.
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
18.
PLoS One ; 9(12): e115494, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25545393

RESUMO

Babesia microti and Borrelia burgdorferi, the respective causative agents of human babesiosis and Lyme disease, are maintained in their enzootic cycles by the blacklegged tick (Ixodes scapularis) and use the white-footed mouse (Peromyscus leucopus) as primary reservoir host. The geographic range of both pathogens has expanded in the United States, but the spread of babesiosis has lagged behind that of Lyme disease. Several studies have estimated the basic reproduction number (R0) for B. microti to be below the threshold for persistence (<1), a finding that is inconsistent with the persistence and geographic expansion of this pathogen. We tested the hypothesis that host coinfection with B. burgdorferi increases the likelihood of B. microti transmission and establishment in new areas. We fed I. scapularis larva on P. leucopus mice that had been infected in the laboratory with B. microti and/or B. burgdorferi. We observed that coinfection in mice increases the frequency of B. microti infected ticks. To identify the ecological variables that would increase the probability of B. microti establishment in the field, we integrated our laboratory data with field data on tick burden and feeding activity in an R0 model. Our model predicts that high prevalence of B. burgdorferi infected mice lowers the ecological threshold for B. microti establishment, especially at sites where larval burden on P. leucopus is lower and where larvae feed simultaneously or soon after nymphs infect mice, when most of the transmission enhancement due to coinfection occurs. Our studies suggest that B. burgdorferi contributes to the emergence and expansion of B. microti and provides a model to predict the ecological factors that are sufficient for emergence of B. microti in the wild.


Assuntos
Babesia microti/patogenicidade , Babesiose/transmissão , Borrelia burgdorferi/patogenicidade , Animais , Coinfecção/microbiologia , Coinfecção/parasitologia , Ixodes/microbiologia , Ixodes/parasitologia , New England , Peromyscus/microbiologia , Peromyscus/parasitologia
20.
Artigo em Inglês | MEDLINE | ID: mdl-24967215

RESUMO

Arthritis in mice infected with the Lyme disease spirochete, Borrelia burgdorferi, results from the influx of innate immune cells responding to the pathogen in the joint and is influenced in part by mouse genetics. Production of inflammatory cytokines by innate immune cells in vitro is largely mediated by Toll-like receptor (TLR) interaction with Borrelia lipoproteins, yet surprisingly mice deficient in TLR2 or the TLR signaling molecule MyD88 still develop arthritis comparable to that seen in wild type mice after B. burgdorferi infection. These findings suggest that other, MyD88-independent inflammatory pathways can contribute to arthritis expression. Clearance of B. burgdorferi is dependent on the production of specific antibody and phagocytosis of the organism. As Fc receptors (FcγR) are important for IgG-mediated clearance of immune complexes and opsonized particles by phagocytes, we examined the role that FcγR play in host defense and disease in B. burgdorferi-infected mice. B. burgdorferi-infected mice deficient in the Fc receptor common gamma chain (FcεRγ(-/-) mice) harbored ~10 fold more spirochetes than similarly infected wild type mice, and this was associated with a transient increase in arthritis severity. While the elevated pathogen burdens seen in B. burgdorferi-infected MyD88(-/-) mice were not affected by concomitant deficiency in FcγR, arthritis was reduced in FcεRγ(-/-) MyD88(-/-) mice in comparison to wild type or single knockout mice. Gene expression analysis from infected joints demonstrated that absence of both MyD88 and FcγR lowers mRNA levels of proteins involved in inflammation, including Cxcl1 (KC), Xcr1 (Gpr5), IL-1beta, and C reactive protein. Taken together, our results demonstrate a role for FcγR-mediated immunity in limiting pathogen burden and arthritis in mice during the acute phase of B. burgdorferi infection, and further suggest that this pathway contributes to the arthritis that develops in B. burgdorferi-infected MyD88(-/-) mice.


Assuntos
Borrelia burgdorferi/imunologia , Doença de Lyme/imunologia , Doença de Lyme/patologia , Receptores de IgG/imunologia , Animais , Carga Bacteriana , Modelos Animais de Doenças , Doença de Lyme/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de IgG/deficiência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...