Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Front Immunol ; 14: 1212167, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38022511

RESUMO

Hepatocytes play a crucial role in host response to infection. Ehrlichia is an obligate intracellular bacterium that causes potentially life-threatening human monocytic ehrlichiosis (HME) characterized by an initial liver injury followed by sepsis and multi-organ failure. We previously showed that infection with highly virulent Ehrlichia japonica (E. japonica) induces liver damage and fatal ehrlichiosis in mice via deleterious MyD88-dependent activation of CASP11 and inhibition of autophagy in macrophage. While macrophages are major target cells for Ehrlichia, the role of hepatocytes (HCs) in ehrlichiosis remains unclear. We investigated here the role of MyD88 signaling in HCs during infection with E. japonica using primary cells from wild-type (WT) and MyD88-/- mice, along with pharmacologic inhibitors of MyD88 in a murine HC cell line. Similar to macrophages, MyD88 signaling in infected HCs led to deleterious CASP11 activation, cleavage of Gasdermin D, secretion of high mobility group box 1, IL-6 production, and inflammatory cell death, while controlling bacterial replication. Unlike macrophages, MyD88 signaling in Ehrlichia-infected HCs attenuated CASP1 activation but activated CASP3. Mechanistically, active CASP1/canonical inflammasome pathway negatively regulated the activation of CASP3 in infected MyD88-/- HCs. Further, MyD88 promoted autophagy induction in HCs, which was surprisingly associated with the activation of the mammalian target of rapamycin complex 1 (mTORC1), a known negative regulator of autophagy. Pharmacologic blocking mTORC1 activation in E. japonica-infected WT, but not infected MyD88-/- HCs, resulted in significant induction of autophagy, suggesting that MyD88 promotes autophagy during Ehrlichia infection not only in an mTORC1-indpenedent manner, but also abrogates mTORC1-mediated inhibition of autophagy in HCs. In conclusion, this study demonstrates that hepatocyte-specific regulation of autophagy and inflammasome pathway via MyD88 is distinct than MyD88 signaling in macrophages during fatal ehrlichiosis. Understanding hepatocyte-specific signaling is critical for the development of new therapeutics against liver-targeting pathogens such as Ehrlichia.


Assuntos
Ehrlichiose , Inflamassomos , Animais , Humanos , Camundongos , Autofagia , Caspase 3/metabolismo , Ehrlichia , Ehrlichiose/microbiologia , Hepatócitos/metabolismo , Inflamassomos/metabolismo , Mamíferos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo
2.
NPJ Vaccines ; 8(1): 3, 2023 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-36693884

RESUMO

Staphylococcus aureus infections are a major public health issue, and a vaccine is urgently needed. Despite a considerable promise in preclinical models, all vaccines tested thus far have failed to protect humans against S. aureus. Unlike laboratory mice, humans are exposed to S. aureus throughout life. In the current study, we hypothesized that prior exposure to S. aureus "imprints" the immune response to inhibit vaccine-mediated protection. We established a mouse model in which S. aureus skin and soft tissue infection (SSTI) is followed by vaccination and secondary SSTI. Unlike naïve mice, S. aureus-sensitized mice were incompletely protected against secondary SSTI by vaccination with the inactivated α-hemolysin (Hla) mutant HlaH35L. Inhibition of protection was specific for the HlaH35L vaccine and required hla expression during primary SSTI. Surprisingly, inhibition occurred at the level of vaccine-elicited effector T cells; hla expression during primary infection limited the expansion of T cells and dendritic cells and impaired vaccine-specific T cell responses. Importantly, the T cell-stimulating adjuvant CAF01 rescued inhibition and restored vaccine-mediated protection. Together, these findings identify a potential mechanism for the failure of translation of promising S. aureus vaccines from mouse models to clinical practice and suggest a path forward to prevent these devastating infections.

3.
Can Vet J ; 62(6): 622-628, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34219771

RESUMO

Clinical findings, geographic locations, laboratory diagnoses, and culture isolation of Neorickettsia spp. in Potomac horse fever (PHF) cases diagnosed in Ontario between 2015 and 2019 are described. Forty-six confirmed PHF cases occurred from late June to early September. Of 41 horses admitted to the Ontario Veterinary College, 28 (68%) survived and 13 (32%) were euthanized due to poor prognosis or financial constraints. Most cases were in southern Ontario along the Canada-USA border. Blood and fecal samples from 43 suspect PHF cases were submitted to 2 laboratories for polymerase chain reaction (PCR) testing for Neorickettsia risticii. Agreement between both laboratories for detection of N. risticii DNA was excellent for feces [κ = 0.932, 95% confidence interval (CI): 0.80 to 1], and fair for blood samples (κ = 0.494, 95% CI: 0.13 to 0.85). Neorickettia spp. were isolated from 16 of 41 (39%) blood samples. DNA analysis confirmed 14 isolates were N. risticii and 2 were N. findlayensis, a novel species of Neorickettsia recently demonstrated to cause PHF.


La fièvre équine du Potomac en Ontario : aspects cliniques, géographiques et diagnostiques. Les résultats cliniques, emplacements géographiques, diagnostics de laboratoire et isolement par culture de Neorickettsia spp. dans les cas de fièvre équine du Potomac (PHF) diagnostiqués en Ontario entre 2015 et 2019 sont décrits. Quarante-six cas confirmés de PHF sont survenus de la fin juin au début septembre. Sur 41 chevaux admis au Ontario Veterinary College, 28 (68%) ont survécu et 13 (32%) ont été euthanasiés en raison d'un mauvais pronostic ou de contraintes financières. La plupart des cas se trouvaient dans le sud de l'Ontario, le long de la frontière canado-américaine. Des échantillons de sang et de matières fécales provenant de 43 cas suspects de PHF ont été soumis à deux laboratoires pour des tests de réaction d'amplification en chaîne par la polymérase (PCR) pour Neorickettsia risticii. La concordance entre les deux laboratoires pour la détection de l'ADN de N. risticii était excellente pour les selles [κ = 0,932, intervalle de confiance (IC) à 95% : 0,80 à 1] et passable pour les échantillons sanguins (κ = 0,494, IC à 95% : 0,13 à 0,85). Neorickettia spp. ont été isolés à partir de 16 des 41 échantillons de sang (39%). L'analyse de l'ADN a confirmé que 14 isolats étaient N. risticii et deux étaient N. findlayensis, une nouvelle espèce de Neorickettsia récemment démontrée comme causant le PHF.(Traduit par Dr Serge Messier).


Assuntos
Infecções por Anaplasmataceae , Doenças dos Cavalos , Neorickettsia risticii , Infecções por Anaplasmataceae/diagnóstico , Infecções por Anaplasmataceae/epidemiologia , Infecções por Anaplasmataceae/veterinária , Animais , Eutanásia Animal , Doenças dos Cavalos/diagnóstico , Doenças dos Cavalos/epidemiologia , Cavalos , Ontário/epidemiologia
4.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-34074773

RESUMO

Iron is essential for survival and proliferation of Ehrlichia chaffeensis, an obligatory intracellular bacterium that causes an emerging zoonosis, human monocytic ehrlichiosis. However, how Ehrlichia acquires iron in the host cells is poorly understood. Here, we found that native and recombinant (cloned into the Ehrlichia genome) Ehrlichia translocated factor-3 (Etf-3), a previously predicted effector of the Ehrlichia type IV secretion system (T4SS), is secreted into the host cell cytoplasm. Secreted Etf-3 directly bound ferritin light chain with high affinity and induced ferritinophagy by recruiting NCOA4, a cargo receptor that mediates ferritinophagy, a selective form of autophagy, and LC3, an autophagosome biogenesis protein. Etf-3-induced ferritinophagy caused ferritin degradation and significantly increased the labile cellular iron pool, which feeds Ehrlichia Indeed, an increase in cellular ferritin by ferric ammonium citrate or overexpression of Etf-3 or NCOA4 enhanced Ehrlichia proliferation, whereas knockdown of Etf-3 in Ehrlichia via transfection with a plasmid encoding an Etf-3 antisense peptide nucleic acid inhibited Ehrlichia proliferation. Excessive ferritinophagy induces the generation of toxic reactive oxygen species (ROS), which could presumably kill both Ehrlichia and host cells. However, during Ehrlichia proliferation, we observed concomitant up-regulation of Ehrlichia Fe-superoxide dismutase, which is an integral component of Ehrlichia T4SS operon, and increased mitochondrial Mn-superoxide dismutase by cosecreted T4SS effector Etf-1. Consequently, despite enhanced ferritinophagy, cellular ROS levels were reduced in Ehrlichia-infected cells compared with uninfected cells. Thus, Ehrlichia safely robs host cell iron sequestered in ferritin. Etf-3 is a unique example of a bacterial protein that induces ferritinophagy to facilitate pathogen iron capture.


Assuntos
Autofagia/fisiologia , Bactérias/metabolismo , Ehrlichia chaffeensis/metabolismo , Ferritinas/metabolismo , Ferro/metabolismo , Autofagossomos/metabolismo , Bactérias/genética , Proteínas de Bactérias/metabolismo , Ehrlichia chaffeensis/genética , Ehrlichiose/microbiologia , Regulação Bacteriana da Expressão Gênica , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Mitocôndrias/metabolismo , Monócitos/metabolismo , Coativadores de Receptor Nuclear , RNA Ribossômico 16S , Espécies Reativas de Oxigênio/metabolismo , Sistemas de Secreção Tipo IV/metabolismo
5.
Front Immunol ; 12: 633672, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33692805

RESUMO

Recurrent S. aureus infections are common, suggesting that natural immune responses are not protective. All candidate vaccines tested thus far have failed to protect against S. aureus infections, highlighting an urgent need to better understand the mechanisms by which the bacterium interacts with the host immune system to evade or prevent protective immunity. Although there is evidence in murine models that both cellular and humoral immune responses are important for protection against S. aureus, human studies suggest that T cells are critical in determining susceptibility to infection. This review will use an "anatomic" approach to systematically outline the steps necessary in generating a T cell-mediated immune response against S. aureus. Through the processes of bacterial uptake by antigen presenting cells, processing and presentation of antigens to T cells, and differentiation and proliferation of memory and effector T cell subsets, the ability of S. aureus to evade or inhibit each step of the T-cell mediated response will be reviewed. We hypothesize that these interactions result in the redirection of immune responses away from protective antigens, thereby precluding the establishment of "natural" memory and potentially inhibiting the efficacy of vaccination. It is anticipated that this approach will reveal important implications for future design of vaccines to prevent these infections.


Assuntos
Desenho de Fármacos , Evasão da Resposta Imune , Memória Imunológica , Reinfecção/prevenção & controle , Infecções Estafilocócicas/prevenção & controle , Vacinas Antiestafilocócicas/uso terapêutico , Staphylococcus aureus/imunologia , Linfócitos T/imunologia , Imunidade Adaptativa , Animais , Antígenos de Bactérias/imunologia , Epitopos , Humanos , Imunogenicidade da Vacina , Ativação Linfocitária , Reinfecção/imunologia , Reinfecção/microbiologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Vacinas Antiestafilocócicas/efeitos adversos , Vacinas Antiestafilocócicas/imunologia , Staphylococcus aureus/patogenicidade , Linfócitos T/microbiologia
6.
mBio ; 11(4)2020 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-32723916

RESUMO

Ehrlichia chaffeensis is an obligatory intracellular bacterium that causes human monocytic ehrlichiosis, an emerging disease transmitted by the Lone Star tick, Amblyomma americanum. E. chaffeensis outer membrane protein entry triggering protein of Ehrlichia (EtpE) is necessary for bacterial entry into human cells. We investigated the role of EtpE in transmission of the bacteria from tick to human cells and whether or not vaccination with EtpE can prevent transmission of ehrlichiae from ticks to mammals. An antiserum against the recombinant C terminus of EtpE (rEtpE-C), which binds a mammalian cell-surface receptor and triggers bacterial entry, significantly inhibited E. chaffeensis transmission from infected tick cells to human monocytes in culture. Each of five specific-pathogen-free dogs were vaccinated with rEtpE-C along with an immunostimulating complex or were sham vaccinated with the complex alone. Dogs vaccinated with rEtpE-C developed high antibody titers against rEtpE-C and produced interferon-γ-secreting cells, as assessed with the ELISpot assay. All 10 dogs were challenged with A. americanum adult ticks infected as nymphs by syringe inoculation with E. chaffeensis Upon challenge, both the vaccinated and control dogs became infected by day 1 post-tick attachment, but the majority of rEtpE-C-vaccinated dogs rapidly cleared the infection from the bloodstream as soon as day 7, whereas most of sham-vaccinated dogs remained infected at day 35. Peripheral blood leukocytes from vaccinated dogs had significantly elevated interferon-γ mRNA levels and secreted significantly elevated interferon-γ soon after tick attachment. Thus, the EtpE-C vaccine represents the first ehrlichial protein vaccine demonstrated to reduce bacterial infection in mammals upon challenge with infected ticks.IMPORTANCE The incidence of tick-borne diseases has risen dramatically in the past two decades and continues to rise. Discovered in 1986 and designated a nationally notifiable disease in 1998 by the Centers for Disease Control and Prevention, human monocytic ehrlichiosis, which is caused by the bacterium Ehrlichia chaffeensis, is one of the most prevalent, life-threatening, emerging tick-borne zoonoses in the United States. We investigated the role of the E. chaffeensis protein EtpE in transmission of the bacterium from tick to human cells and in vaccinated dogs with EtpE to assess the efficacy of vaccination against E. chaffeensis-infected tick challenge. Our results help fill gaps in our understanding of E. chaffeensis-derived protective antigens that could be used in a candidate vaccine for immunization of humans to counter tick-transmitted ehrlichiosis.


Assuntos
Vacinas Bacterianas/imunologia , Ehrlichia chaffeensis/imunologia , Ehrlichiose/prevenção & controle , Ehrlichiose/transmissão , Carrapatos/microbiologia , Animais , Proteínas de Bactérias/imunologia , Linhagem Celular , Cães , Ehrlichia chaffeensis/genética , Ehrlichiose/imunologia , Feminino , Humanos , Interferon gama/imunologia , Masculino , Monócitos/imunologia , Monócitos/microbiologia , Organismos Livres de Patógenos Específicos , Vacinação
7.
mBio ; 11(2)2020 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-32317318

RESUMO

The obligatory intracellular pathogen Ehrlichia chaffeensis lacks most factors that could respond to oxidative stress (a host cell defense mechanism). We previously found that the C terminus of Ehrlichia surface invasin, entry-triggering protein of Ehrlichia (EtpE; EtpE-C) directly binds mammalian DNase X, a glycosylphosphatidylinositol-anchored cell surface receptor and that binding is required to induce bacterial entry and simultaneously to block the generation of reactive oxygen species (ROS) by host monocytes and macrophages. However, how the EtpE-C-DNase X complex mediates the ROS blockade was unknown. A mammalian transmembrane glycoprotein CD147 (basigin) binds to the EtpE-DNase X complex and is required for Ehrlichia entry and infection of host cells. Here, we found that bone marrow-derived macrophages (BMDM) from myeloid cell lineage-selective CD147-null mice had significantly reduced Ehrlichia-induced or EtpE-C-induced blockade of ROS generation in response to phorbol myristate acetate. In BMDM from CD147-null mice, nucleofection with CD147 partially restored the Ehrlichia-mediated inhibition of ROS generation. Indeed, CD147-null mice as well as their BMDM were resistant to Ehrlichia infection. Moreover, in human monocytes, anti-CD147 partially abrogated EtpE-C-induced blockade of ROS generation. Both Ehrlichia and EtpE-C could block activation of the small GTPase Rac1 (which in turn activates phagocyte NADPH oxidase) and suppress activation of Vav1, a hematopoietic-specific Rho/Rac guanine nucleotide exchange factor by phorbol myristate acetate. Vav1 suppression by Ehrlichia was CD147 dependent. E. chaffeensis is the first example of pathogens that block Rac1 activation to colonize macrophages. Furthermore, Ehrlichia uses EtpE to hijack the unique host DNase X-CD147-Vav1 signaling to block Rac1 activation.IMPORTANCEEhrlichia chaffeensis is an obligatory intracellular bacterium with the capability of causing an emerging infectious disease called human monocytic ehrlichiosis. E. chaffeensis preferentially infects monocytes and macrophages, professional phagocytes, equipped with an arsenal of antimicrobial mechanisms, including rapid reactive oxygen species (ROS) generation upon encountering bacteria. As Ehrlichia isolated from host cells are readily killed upon exposure to ROS, Ehrlichia must have evolved a unique mechanism to safely enter phagocytes. We discovered that binding of the Ehrlichia surface invasin to the host cell surface receptor not only triggers Ehrlichia entry but also blocks ROS generation by the host cells by mobilizing a novel intracellular signaling pathway. Knowledge of the mechanisms by which ROS production is inhibited may lead to the development of therapeutics for ehrlichiosis as well as other ROS-related pathologies.


Assuntos
Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Basigina/metabolismo , Ehrlichia chaffeensis/fisiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Proteínas Proto-Oncogênicas c-vav/antagonistas & inibidores , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores , Animais , Basigina/genética , Humanos , Macrófagos/microbiologia , Camundongos , Camundongos Knockout , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/microbiologia , Espécies Reativas de Oxigênio/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
9.
Artigo em Inglês | MEDLINE | ID: mdl-32117791

RESUMO

Ehrlichia spp. are emerging tick-borne obligatory intracellular bacteria that cause febrile and sometimes fatal diseases with abnormal blood cell counts and signs of hepatitis. Ehrlichia HF strain provides an excellent mouse disease model of fatal human ehrlichiosis. We recently obtained and established stable culture of Ehrlichia HF strain in DH82 canine macrophage cell line, and obtained its whole genome sequence and annotation. To identify genes required for in vivo virulence of Ehrlichia, we constructed random insertional HF strain mutants by using Himar1 transposon-based mutagenesis procedure. Of total 158 insertional mutants isolated via antibiotic selection in DH82 cells, 74 insertions were in the coding regions of 55 distinct protein-coding genes, including TRP120 and multi-copy genes, such as p28/omp-1, virB2, and virB6. Among 84 insertions mapped within the non-coding regions, seven are located in the putative promoter region since they were within 50 bp upstream of the seven distinct genes. Using limited dilution methods, nine stable clonal mutants that had no apparent defect for multiplication in DH82 cells, were obtained. Mouse virulence of seven mutant clones was similar to that of wild-type HF strain, whereas two mutant clones showed significantly retarded growth in blood, livers, and spleens, and the mice inoculated with them lived longer than mice inoculated with wild-type. The two clones contained mutations in genes encoding a conserved hypothetical protein and a staphylococcal superantigen-like domain protein, respectively, and both genes are conserved among Ehrlichia spp., but lack homology to other bacterial genes. Inflammatory cytokine mRNA levels in the liver of mice infected with the two mutants were significantly diminished than those infected with HF strain wild-type, except IL-1ß and IL-12 p40 in one clone. Thus, we identified two Ehrlichia virulence genes responsible for in vivo infection, but not for infection and growth in macrophages.


Assuntos
Ehrlichia/genética , Ehrlichia/patogenicidade , Ehrlichiose/microbiologia , Genes Bacterianos , Animais , Carga Bacteriana , Linhagem Celular , Clonagem Molecular , Citocinas/genética , Citocinas/metabolismo , Elementos de DNA Transponíveis , Cães , Ehrlichia/crescimento & desenvolvimento , Expressão Gênica , Humanos , Ixodes , Dose Letal Mediana , Macrófagos/microbiologia , Camundongos , Mutagênese Insercional , Virulência/genética
10.
mBio ; 11(1)2020 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-32098825

RESUMO

Potomac horse fever (PHF), a severe and frequently fatal febrile diarrheal disease, has been known to be caused only by Neorickettsia risticii, an endosymbiont of digenean trematodes. Here, we report the cell culture isolation of a new Neorickettsia species found in two locations in eastern Ontario, Canada, in 2016 and 2017 (in addition to 10 variable strains of N. risticii) from N. risticii PCR-negative horses with clinical signs of PHF. Gene sequences of 16S rRNA and the major surface antigen P51 of this new Neorickettsia species were distinct from those of all previously characterized N. risticii strains and Neorickettsia species, except for those from an uncharacterized Neorickettsia species culture isolate from a horse with PHF in northern Ohio in 1991. The new Neorickettsia species nonetheless had the characteristic intramolecular repeats within strain-specific antigen 3 (Ssa3), which were found in all sequenced Ssa3s of N. risticii strains. Experimental inoculation of two naive ponies with the new Neorickettsia species produced severe and subclinical PHF, respectively, and the bacteria were reisolated from both of them, fulfilling Koch's postulates. Serological assay titers against the new Neorickettsia species were higher than those against N. risticii Whole-genome sequence analysis of the new Neorickettsia species revealed unique features of this bacterium compared with N. risticii We propose to classify this new bacterium as Neorickettsia finleia sp. nov. This finding will improve the laboratory diagnosis of and vaccine for PHF, environmental risk assessment of PHF, and understanding of PHF pathogenesis and Neorickettsia biology in general.IMPORTANCE Despite the detection of Neorickettsia species DNA sequences in various trematode species and their hosts, only three Neorickettsia species have been cell culture isolated and whole-genome sequenced and are known to infect mammals and/or cause disease. The molecular mechanisms that enable the obligatory intracellular bacterium Neorickettsia to colonize trematodes and to horizontally transmit from trematodes to mammals, as well as the virulence factors associated with specific mammalian hosts, are unknown. Potomac horse fever (PHF) is a severe and acute systemic infectious disease of horses, with clinical signs that include diarrhea. Neorickettsia risticii is the only known bacterial species that causes PHF. Ingestion of insects harboring N. risticii-infected trematodes by horses leads to PHF. Our discovery of a new Neorickettsia species that causes PHF and whole-genome sequence analysis of this bacterium will improve laboratory diagnosis and vaccine development for PHF and will contribute to our understanding of Neorickettsia ecology, pathogenesis, and biology.


Assuntos
Infecções por Anaplasmataceae/microbiologia , Doenças dos Cavalos/microbiologia , Neorickettsia/classificação , Neorickettsia/genética , Neorickettsia/isolamento & purificação , Filogenia , Infecções por Anaplasmataceae/diagnóstico , Animais , Antígenos de Bactérias/genética , Canadá , DNA Bacteriano/análise , Modelos Animais de Doenças , Feminino , Doenças dos Cavalos/diagnóstico , Cavalos , Masculino , Neorickettsia/patogenicidade , Neorickettsia risticii/genética , Neorickettsia risticii/isolamento & purificação , Reação em Cadeia da Polimerase , RNA Ribossômico 16S/genética , Análise de Sequência , Trematódeos/microbiologia , Sequenciamento Completo do Genoma
11.
Microb Pathog ; 139: 103907, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31811888

RESUMO

Enterococcus faecalis is one of the important causes of nosocomial infections. Nowadays, increasing prevalence of antibiotic-resistant bacteria and slow progress in recognizing new antimicrobial agents has limited the efficiency of conventional antibiotics, which cause to find novel strategies to overcome bacteria. Therefore, in this study, we aimed to assess the role of efaA gene in the biofilm formation and the role of ftsZ gene in the controlling of bacterial growth by the anti-sense PNAs(Peptide Nucleic Acid).E. faecalis ATCC® 29212™was used for the study of PNAs designed to targeting the start codon section of the ftsZ andefaA genes. PNA attachment to RNA was confirmed by blotting. Electroporation technique was used for the intracellular transfer of anti-ftsZ PNAs. The spot-plating method was used to the assessment of alteration in bacterial growth. Biofilm formation assay and real-time PCR were used for detection of biofilm inhibitory effect of cell penetrating peptide (CPP) conjugated to anti-efaA PNAs.ByftsZ PNAs treatment, no growth was seen from the strain in agar by a spot plating method and the inhibition zone of anti-ftsZ PNAs was not seen. PNAs against the efaA gene decreased by 95% the expression of the efaA gene and biofilm formation. In addition, the(3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide) MTT assay showed no toxicity on MCF7 cells for both of anti-ftsZand anti-efaA PNAs.This study used new genetic and molecular tools to inhibit pathogenicity and infection by E. faecalis. In this study, we suggested that efaA gene plays a critical role in the biofilm formation and anti-efaA PNAs could decrease the formation of biofilm, as well as, anti-ftsZ PNAs could eliminate bacterial growth.


Assuntos
Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Biofilmes , Proteínas do Citoesqueleto/genética , Enterococcus faecalis/genética , Ácidos Nucleicos Peptídicos/genética , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas do Citoesqueleto/metabolismo , Enterococcus faecalis/crescimento & desenvolvimento , Enterococcus faecalis/fisiologia , Regulação Bacteriana da Expressão Gênica
12.
Proc Natl Acad Sci U S A ; 115(38): E8977-E8986, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30181274

RESUMO

Ehrlichia chaffeensis, an obligatory intracellular bacterium, infects monocytes/macrophages by sequestering a regulator of endosomal traffic, the small GTPase RAB5, on its membrane-bound inclusions to avoid routing to host-cell phagolysosomes. How RAB5 is sequestered on ehrlichial inclusions is poorly understood, however. We found that native Ehrlichia translocated factor-2 (Etf-2), a previously predicted effector of the Ehrlichia type IV secretion system, and recombinant Etf-2 (cloned into the Ehrlichia genome) are secreted into the host-cell cytoplasm and localize to ehrlichial inclusions. Ectopically expressed Etf-2-GFP also localized to inclusions and membranes of early endosomes marked with RAB5 and interacted with GTP-bound RAB5 but not with a GDP-bound RAB5. Etf-2, although lacking a RAB GTPase-activating protein (GAP) Tre2-Bub2-Cdc16 (TBC) domain, contains two conserved TBC domain motifs, namely an Arg finger and a Gln finger, and site-directed mutagenesis revealed that both Arg188 and Gln245 are required for Etf-2 localization to early endosomes. The yeast two-hybrid assay and microscale thermophoresis revealed that Etf-2 binds tightly to GTP-bound RAB5 but not to GDP-bound RAB5. However, Etf-2 lacks RAB5-specific GAP activity. Etf-2 localized to bead-containing phagosomes as well as endosomes containing beads coated with the C-terminal fragment of EtpE (entry-triggering protein of Ehrlichia), an Ehrlichia outer-membrane invasin, and significantly delayed RAB5 dissociation from and RAB7 localization to phagosomes/endosomes and RABGAP5 localization to endosomes. Thus, binding of Etf-2 to RAB5-GTP appears to delay RAB5 inactivation by impeding RABGAP5 localization to endosomes. This suggests a unique mechanism by which RAB5 is sequestered on ehrlichial inclusions to benefit bacterial survival and replication.


Assuntos
Proteínas de Bactérias/metabolismo , Ehrlichia chaffeensis/fisiologia , Endossomos/imunologia , Fagossomos/imunologia , Sistemas de Secreção Tipo IV/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Linhagem Celular , Endossomos/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Técnicas de Silenciamento de Genes , Interações Hospedeiro-Patógeno/imunologia , Humanos , Macaca mulatta , Fagossomos/metabolismo , Ligação Proteica , Alinhamento de Sequência
13.
APMIS ; 126(1): 76-84, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29135054

RESUMO

Toll-like receptor-4 (TLR4) polymorphisms may influence host immune response against Helicobacter pylori (H. pylori). This study aimed to investigate whether TLR4 polymorphisms are associated with H. pylori susceptibility and risk of peptic ulcer development or not. The TLR4 + 3725 G/C polymorphism was studied using polymerase chain reaction with confronting two-pair primers (PCR-CTPP). In addition, TLR4 Asp299Gly and Thr399Ile polymorphisms were evaluated by PCR-restriction fragment length polymorphism (RFLP). There was no significant difference in TLR4 + 3725 G/C and Asp299Gly genotype frequencies between non-peptic ulcer (NPUD) and peptic ulcer (PUD) individuals in the context of peptic ulcer development and susceptibility to infection with H. pylori. Nevertheless, a significant association with increased risk for PUD development was observed for polymorphism TLR4 Thr399Ile [odds ratio (OR) = 4.2; 95% confidence interval (CI) = 1.35-13.26; p = 0.01]. Correspondingly, TLR4 Thr399Ile polymorphism was associated with H. pylori susceptibility (OR = 0.27; 95% CI = 0.08-0.88; p = 0.04). In addition, TLR4 Thr399Ile polymorphism increased 4.2-fold, the risk of peptic ulcer development in individuals infected by H. pylori carrying CT + TT genotype. Our results showed that TLR4 Thr399Ile polymorphism along with H. pylori infection may play critical roles in peptic ulcer development in North of Iran.


Assuntos
Predisposição Genética para Doença , Infecções por Helicobacter/genética , Helicobacter pylori , Úlcera Péptica/etiologia , Polimorfismo Genético , Receptor 4 Toll-Like/genética , Adulto , Idoso , Estudos de Casos e Controles , Feminino , Haplótipos , Humanos , Masculino , Pessoa de Meia-Idade , Risco
14.
mBio ; 8(6)2017 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-29162709

RESUMO

The obligatory intracellular pathogen Ehrlichia chaffeensis lacks most genes that confer resistance to oxidative stress but can block reactive oxygen species (ROS) generation by host monocytes-macrophages. Bacterial and host molecules responsible for this inhibition have not been identified. To infect host cells, Ehrlichia uses the C terminus of its surface invasin, entry-triggering protein of Ehrlichia (EtpE; EtpE-C), which directly binds the mammalian cell surface receptor glycosylphosphatidylinositol-anchored protein DNase X. We investigated whether EtpE-C binding to DNase X blocks ROS production by mouse bone marrow-derived macrophages (BMDMs). On the basis of a luminol-dependent chemiluminescence assay, E. chaffeensis inhibited phorbol myristate acetate (PMA)-induced ROS generation by BMDMs from wild-type, but not DNase X-/-, mice. EtpE-C is critical for inhibition, as recombinant EtpE-C (rEtpE-C)-coated latex beads, but not recombinant N-terminal EtpE-coated or uncoated beads, inhibited PMA-induced ROS generation by BMDMs from wild-type mice. DNase X is required for this inhibition, as none of these beads inhibited PMA-induced ROS generation by BMDMs from DNase X-/- mice. Previous studies showed that E. chaffeensis does not block ROS generation in neutrophils, a cell type that is a potent ROS generator but is not infected by E. chaffeensis Human and mouse peripheral blood neutrophils did not express DNase X. Our findings point to a unique survival mechanism of ROS-sensitive obligate intramonocytic bacteria that involves invasin EtpE binding to DNase X on the host cell surface. This is the first report of bacterial invasin having such a subversive activity on ROS generation.IMPORTANCEEhrlichia chaffeensis preferentially infects monocytes-macrophages and causes a life-threatening emerging tick-transmitted infectious disease called human monocytic ehrlichiosis. Ehrlichial infection, and hence the disease, depends on the ability of this bacterium to avoid or overcome powerful microbicidal mechanisms of host monocytes-macrophages, one of which is the generation of ROS. Our findings reveal that an ehrlichial surface invasin, EtpE, not only triggers bacterial entry but also blocks ROS generation by host macrophages through its host cell receptor, DNase X. As ROS sensitivity is an Achilles' heel of this group of pathogens, understanding the mechanism by which E. chaffeensis rapidly blocks ROS generation suggests a new approach for developing effective anti-infective measures. The discovery of a ROS-blocking pathway is also important, as modulation of ROS generation is important in a variety of ailments and biological processes.


Assuntos
Adesinas Bacterianas/imunologia , Desoxirribonucleases/metabolismo , Ehrlichia chaffeensis/imunologia , Macrófagos/imunologia , Espécies Reativas de Oxigênio/imunologia , Animais , Desoxirribonucleases/deficiência , Desoxirribonucleases/genética , Ehrlichia chaffeensis/genética , Ehrlichia chaffeensis/patogenicidade , Humanos , Macrófagos/microbiologia , Camundongos , Monócitos/imunologia , Monócitos/microbiologia , Neutrófilos/imunologia , Neutrófilos/microbiologia , Transdução de Sinais
15.
Sci Rep ; 7(1): 8036, 2017 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-28808292

RESUMO

The H. pylori outer inflammatory protein A (OipA) is an outer membrane protein that contributes to gastric inflammation. OipA is believed to affect intra-cellular signalling and modulate the host signalling pathways. The aim of the current study was to clarify the role of OipA in H. pylori pathogenesis and its effect on host cell signalling pathways. To this end, the oipA gene was isolated and inserted into cloning and expression vectors. The recombinant plasmid was transferred into an expression host to produce OipA, which was subsequently purified by affinity chromatography and used for antibody production. A confluent monolayer of gastric cell lines was treated with various concentrations of OipA and investigated for attachment, toxicity, and apoptosis and alterations in signalling pathways. OipA bound to gastric cell lines confirming its role in the attachment of H. pylori to host cells. The ratio of Bax/Bcl-2 and caspase3, 8, FasL in the host cells were assessed and the results showed that the Bax/Bcl-2 ratio as well as the level of cleaved-caspase 3 was elevated in OipA-treated cells. These findings suggest that OipA can bind and induce toxic events as well as triggering apoptotic cascade in host gastric cells through intrinsic pathway.


Assuntos
Apoptose/fisiologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/microbiologia , Helicobacter pylori/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo , Animais , Antígenos de Bactérias/metabolismo , Caspase 3/metabolismo , Caspase 8/metabolismo , Linhagem Celular , Proteína Ligante Fas/metabolismo , Feminino , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiologia , Gastrite/metabolismo , Gastrite/microbiologia , Humanos , Coelhos , Transdução de Sinais/fisiologia , Estômago/microbiologia
16.
Artigo em Inglês | MEDLINE | ID: mdl-28638803

RESUMO

Survival of Ehrlichia chaffeensis depends on obligatory intracellular infection. One of the barriers to E. chaffeensis research progress has been the inability, using conventional techniques, to generate knock-out mutants for genes essential for intracellular infection. This study examined the use of Peptide Nucleic Acids (PNAs) technology to interrupt type IV secretion system (T4SS) effector protein expression in E. chaffeensis followed by intracellular complementation of the effector to determine its requirement for infection. Successful E. chaffeensis infection depends on the E. chaffeensis-specific T4SS protein effector, ehrlichial translocated factor-1 (Etf-1), which induces Rab5-regulated autophagy to provide host cytosolic nutrients required for E. chaffeensis proliferation. Etf-1 is also imported by host cell mitochondria where it inhibits host cell apoptosis to prolong its infection. We designed a PNA specific to Etf-1 and showed that the PNA bound to the target region of single-stranded Etf-1 RNA using a competitive binding assay. Electroporation of E. chaffeensis with this PNA significantly reduced Etf-1 mRNA and protein, and the bacteria's ability to induce host cell autophagy and infect host cells. Etf-1 PNA-mediated inhibition of ehrlichial Etf-1 expression and E. chaffeensis infection could be intracellularly trans-complemented by ectopic expression of Etf-1-GFP in host cells. These data affirmed the critical role of bacterial T4SS effector in host cell autophagy and E. chaffeensis infection, and demonstrated the use of PNA to analyze the gene functions of obligate intracellular bacteria.


Assuntos
Proteínas do Sistema Complemento/metabolismo , Ehrlichia chaffeensis/genética , Ehrlichia chaffeensis/metabolismo , Ehrlichiose/imunologia , Técnicas de Silenciamento de Genes , Interações Hospedeiro-Patógeno/imunologia , Ácidos Nucleicos Peptídicos/genética , Sistemas de Secreção Tipo IV/metabolismo , Apoptose , Autofagia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Ehrlichia chaffeensis/imunologia , Ehrlichia chaffeensis/patogenicidade , Ehrlichiose/microbiologia , Regulação Bacteriana da Expressão Gênica , Células HEK293 , Humanos , Mitocôndrias/metabolismo , Fatores de Terminação de Peptídeos/genética , Fatores de Terminação de Peptídeos/metabolismo , RNA Mensageiro/metabolismo , Células THP-1 , Proteínas rab5 de Ligação ao GTP/genética , Proteínas rab5 de Ligação ao GTP/metabolismo
17.
Microb Pathog ; 99: 1-4, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27457974

RESUMO

Pseudomonas aeruginosa possesses various virulence factors which contribute to the bacterial invasion and toxicity. Moreover, children suffered from Cystic Fibrosis (CF) and burn wounds are at a high risk of various bacterial infections. The aim of this study was to determine the prevalence of virulent genes in P. aeruginosa isolated from children with CF and burn wounds and comparing their virulence genes to figure out the role of every virulent factor in the infections. P. aeruginosa were isolated from sputum, oropharyngeal swabs, and broncho-alveolar lavage (BAL) specimens from CF and burn wounds between June 2013 and June 2014 in Tehran's hospitals. Bacterial genomic DNAs were extracted and uniplex, duplex and multiplex PCR were performed for detection of toxA, algD and plcN, exoS, lasB, plcH genes, respectively. The prevalence rate of virulence genes in P. aeruginosa isolated from CF was; toxA (63.1%), algD (64.6%), plcH (87.7%), plcN (60%), lasB (95.4%) and exoS (70.8%) and virulence genes in P. aeruginosa from burn patients were: toxA (36.9%), algD (70.1%), plcH (79%), plcN (63.1%), lasB (82%) and exoS (21.1%). The prevalence of three virulent genes in P. aeruginosa was higher in CF comparing to burn wound infections. We found that the number of toxA, lasB and exoS were significantly higher in the bacteria which were isolated from children with CF. This finding shows that these virulence factors play an important role in CF infections by P. aeroginosa.


Assuntos
Queimaduras/complicações , Fibrose Cística/complicações , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidade , Fatores de Virulência/genética , Infecção dos Ferimentos/microbiologia , Criança , DNA Bacteriano/genética , DNA Bacteriano/isolamento & purificação , Genes Bacterianos , Genótipo , Humanos , Irã (Geográfico) , Reação em Cadeia da Polimerase Multiplex , Prevalência , Pseudomonas aeruginosa/isolamento & purificação , Fatores de Virulência/análise
18.
Infect Immun ; 83(4): 1610-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25644008

RESUMO

Brachyspira hyodysenteriae colonizes the pig colon, resulting in mucohemorrhagic diarrhea and growth retardation. Fecal mucus is a characteristic feature of swine dysentery; therefore, we investigated how the mucin environment changes in the colon during infection with B. hyodysenteriae and how these changes affect this bacterium's interaction with mucins. We isolated and characterized mucins, the main component of mucus, from the colon of experimentally inoculated and control pigs and investigated B. hyodysenteriae binding to these mucins. Fluorescence microscopy revealed a massive mucus induction and disorganized mucus structure in the colon of pigs with swine dysentery. Quantitative PCR (qPCR) and antibody detection demonstrated that the mucus composition of pigs with swine dysentery was characterized by de novo expression of MUC5AC and increased expression of MUC2 in the colon. Mucins from the colon of inoculated and control pigs were isolated by two steps of isopycnic density gradient centrifugation. The mucin densities of control and inoculated pigs were similar, whereas the mucin quantity was 5-fold higher during infection. The level of B. hyodysenteriae binding to mucins differed between pigs, and there was increased binding to soluble mucins isolated from pigs with swine dysentery. The ability of B. hyodysenteriae to bind, measured in relation to the total mucin contents of mucus in sick versus healthy pigs, increased 7-fold during infection. Together, the results indicate that B. hyodysenteriae binds to carbohydrate structures on the mucins as these differ between individuals. Furthermore, B. hyodysenteriae infection induces changes to the mucus niche which substantially increase the amount of B. hyodysenteriae binding sites in the mucus.


Assuntos
Aderência Bacteriana/fisiologia , Brachyspira hyodysenteriae/patogenicidade , Mucinas Gástricas/metabolismo , Infecções por Bactérias Gram-Negativas/microbiologia , Mucina-5AC/biossíntese , Mucina-2/biossíntese , Doenças dos Suínos/microbiologia , Animais , Colo/microbiologia , Muco/metabolismo , Ligação Proteica , Suínos
19.
J Pathog ; 2014: 871601, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25120932

RESUMO

Frequently reported H. pylori antimicrobial therapy failures suggest that there might be a different niche where the bacteria can stay safe. Current study aims to examine potential role of oral colonization of H. pylori to feed reinfection after primary therapy. However, patients who were admitted to the gastroscopy section were chosen and gastric biopsy and dental plaque specimens were collected. Molecular and biochemical tests were applied to confirm H. pylori identity in different colonization niches. Results showed that 88.8% of dyspeptic patients had epigastric pains with nocturnal awakening when they were hungry (P = 0.023). All patients who received therapy already were again H. pylori positive while they are still carrying H. pylori in dental plaque (P = 0.001). Moreover, H. pylori infection was sought in 100% of gastric biopsy's dyspeptic patients who had ulcerated esophagitis and erosive duodenitis and who were H. pylori positive, and 75% of dyspeptic patients with duodenum deformity had this bacterium in gastric biopsies (P = 0.004). Present study showed that only successful eradication of gastric H. pylori cannot guarantee prevention of reinfection. Conclusively, a new strategy which indicates concomitant eradication in oral and gastric colonization can result in clearance of H. pylori infection.

20.
Helicobacter ; 19(2): 136-43, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24495278

RESUMO

BACKGROUND: Outer inflammatory protein A (OipA) has an important role in Helicobacter pylori pathogenesis. In this study, we purified the outer membrane protein and evaluated the effects of this protein on maturation and cytokine production by dendritic cells (DCs). MATERIALS AND METHODS: The oipA gene was inserted into pET28a, and this construct was transformed into Escherichia coli BL21 (DE3). Purification of the recombinant protein was performed by Ni-NTA affinity chromatography. Immature DCs were purified from spleen of C57BL/6 mice with more than 90% purity and were treated with several concentrations of OipA (1-20 µg/mL) overnight. Expression of maturation markers (CD86, CD40, and MHC-II) on the surface of DCs and production of IL-10 and IL-12 were assessed by flow cytometry and ELISA, respectively. RESULTS: The expression of DC maturation markers CD40, CD86, and MHC-II was downregulated on the surface of OipA-treated DCs at concentrations of 10 and 20 µg/mL compared with negative control. Production of IL-10 decreases with increasing OipA concentration at a concentration of 5 µg/mL, but we detected no change in IL-12 production. CONCLUSION: Inability to eliminate H. pylori from stomach is partly due to the evasion of the bacteria from the immune response. DCs are central mediators between innate and adaptive immunity, and DC cytokines direct the types of adaptive immune response. This study indicated that OipA of H. pylori is a DC maturation suppression factor. Previous studies have shown that H. pylori manage tolerogenic programming in DCs leading to long-time gastric colonization. In conclusion, H. pylori OipA helps the establishment of chronic infection with reduction in IL-10 and suppression of DC maturation.


Assuntos
Proteínas da Membrana Bacteriana Externa/imunologia , Células Dendríticas/imunologia , Helicobacter pylori/imunologia , Evasão da Resposta Imune/imunologia , Proteínas Recombinantes/farmacologia , Animais , Antígeno B7-2/biossíntese , Proteínas da Membrana Bacteriana Externa/genética , Antígenos CD40/biossíntese , Células Cultivadas , Regulação para Baixo , Feminino , Expressão Gênica/efeitos dos fármacos , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Helicobacter pylori/patogenicidade , Antígenos de Histocompatibilidade Classe II/biossíntese , Interleucina-10/biossíntese , Interleucina-12/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA