Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 330
Filtrar
1.
J Biol Chem ; 299(9): 104927, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37330175

RESUMO

Methicillin-resistant Staphylococcus aureus, or MRSA, is one of the major causative agents of hospital-acquired infections worldwide. Novel antimicrobial strategies efficient against antibiotic-resistant strains are necessary and not only against S. aureus. Among those, strategies that aim at blocking or dismantling proteins involved in the acquisition of essential nutrients, helping the bacteria to colonize the host, are intensively studied. A major route for S. aureus to acquire iron from the host organism is the Isd (iron surface determinant) system. In particular, the hemoglobin receptors IsdH and IsdB located on the surface of the bacterium are necessary to acquire the heme moiety containing iron, making them a plausible antibacterial target. Herein, we obtained an antibody of camelid origin that blocked heme acquisition. We determined that the antibody recognized the heme-binding pocket of both IsdH and IsdB with nanomolar order affinity through its second and third complementary-determining regions. The mechanism explaining the inhibition of acquisition of heme in vitro could be described as a competitive process in which the complementary-determining region 3 from the antibody blocked the acquisition of heme by the bacterial receptor. Moreover, this antibody markedly reduced the growth of three different pathogenic strains of MRSA. Collectively, our results highlight a mechanism for inhibiting nutrient uptake as an antibacterial strategy against MRSA.


Assuntos
Anticorpos Antibacterianos , Staphylococcus aureus Resistente à Meticilina , Receptores de Superfície Celular , Anticorpos de Domínio Único , Humanos , Antibacterianos/farmacologia , Heme/metabolismo , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/uso terapêutico , Anticorpos de Domínio Único/biossíntese , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/metabolismo , Anticorpos de Domínio Único/farmacologia , Infecções Estafilocócicas/tratamento farmacológico , Antígenos de Bactérias/imunologia , Anticorpos Antibacterianos/genética , Anticorpos Antibacterianos/imunologia , Camelídeos Americanos , Animais , Ligação Proteica/efeitos dos fármacos , Modelos Moleculares , Simulação de Dinâmica Molecular
2.
Front Immunol ; 13: 931052, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35898510

RESUMO

Salmonella enterica is a leading cause of food-borne diseases in humans worldwide, resulting in severe morbidity and mortality. They are carried asymptomatically in the intestine or gallbladder of livestock, and are transmitted predominantly from animals to humans via the fecal-oral route. Thus, the best preventive strategy is to preemptively prevent transmission to humans by vaccinating livestock. Live attenuated vaccines have been mostly favored because they elicit both cellular and humoral immunity and provide long-term protective immunity. However, developing these vaccines is a laborious and time-consuming process. Therefore, most live attenuated vaccines have been mainly used for phenotypic screening using the auxotrophic replica plate method, and new types of vaccines have not been sufficiently explored. In this study, we used Radiation-Mutation Enhancement Technology (R-MET) to introduce a wide variety of mutations and attenuate the virulence of Salmonella spp. to develop live vaccine strains. The Salmonella Typhimurium, ST454 strain (ST WT) was irradiated with Cobalt60 gamma-irradiator at 1.5 kGy for 1 h to maximize the mutation rate, and attenuated daughter colonies were screened using in vitro macrophage replication capacity and in vivo mouse infection assays. Among 30 candidates, ATOMSal-L6, with 9,961-fold lower virulence than the parent strain (ST454) in the mouse LD50 model, was chosen. This vaccine candidate was mutated at 71 sites, and in particular, lost one bacteriophage. As a vaccine, ATOMSal-L6 induced a Salmonella-specific IgG response to provide effective protective immunity upon intramuscular vaccination of mice. Furthermore, when mice and sows were orally immunized with ATOMSal-L6, we found a strong protective immune response, including multifunctional cellular immunity. These results indicate that ATOMSal-L6 is the first live vaccine candidate to be developed using R-MET, to the best of our knowledge. R-MET can be used as a fast and effective live vaccine development technology that can be used to develop vaccine strains against emerging or serotype-shifting pathogens.


Assuntos
Melhoramento Biomédico , Vacinas contra Salmonella , Animais , Anticorpos Antibacterianos/genética , Feminino , Humanos , Imunoglobulina G/genética , Camundongos , Mutação , Vacinas contra Salmonella/genética , Salmonella typhimurium , Suínos , Vacinas Atenuadas
3.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35058363

RESUMO

Gram-positive organisms with their thick envelope cannot be lysed by complement alone. Nonetheless, antibody-binding on the surface can recruit complement and mark these invaders for uptake and killing by phagocytes, a process known as opsonophagocytosis. The crystallizable fragment of immunoglobulins (Fcγ) is key for complement recruitment. The cell surface of S. aureus is coated with Staphylococcal protein A (SpA). SpA captures the Fcγ domain of IgG and interferes with opsonization by anti-S. aureus antibodies. In principle, the Fcγ domain of therapeutic antibodies could be engineered to avoid the inhibitory activity of SpA. However, the SpA-binding site on Fcγ overlaps with that of the neonatal Fc receptor (FcRn), an interaction that is critical for prolonging the half-life of serum IgG. This evolutionary adaptation poses a challenge for the exploration of Fcγ mutants that can both weaken SpA-IgG interactions and retain stability. Here, we use both wild-type and transgenic human FcRn mice to identify antibodies with enhanced half-life and increased opsonophagocytic killing in models of S. aureus infection and demonstrate that antibody-based immunotherapy can be improved by modifying Fcγ. Our experiments also show that by competing for FcRn-binding, staphylococci effectively reduce the half-life of antibodies during infection. These observations may have profound impact in treating cancer, autoimmune, and asthma patients colonized or infected with S. aureus and undergoing monoclonal antibody treatment.


Assuntos
Anticorpos Antibacterianos/genética , Anticorpos Antibacterianos/imunologia , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Opsonização/imunologia , Engenharia de Proteínas , Sequência de Aminoácidos , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Ativação do Complemento , Relação Dose-Resposta a Droga , Relação Dose-Resposta Imunológica , Humanos , Fagocitose/imunologia , Ligação Proteica , Engenharia de Proteínas/métodos , Domínios e Motivos de Interação entre Proteínas/genética , Domínios e Motivos de Interação entre Proteínas/imunologia , Receptores Fc/genética , Proteína Estafilocócica A/imunologia , Staphylococcus aureus/imunologia
4.
Front Immunol ; 12: 732938, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34887850

RESUMO

Staphylococcal enterotoxin B (SEB) simultaneously crosslinks MHC class II antigen and TCR, promoting proliferation of T cells and releasing a large number of toxic cytokines. In this report, we computationally examined the possibility of using a single-chain biparatopic bispecific antibody to target SEB and prevent TCR binding. The design was inspired by the observation that mixing two anti-SEB antibodies 14G8 and 6D3 can block SEB-TCR activation, and we used 14G8-6D3-SEB tertiary crystal structure as a template. Twelve simulation systems were constructed to systematically examine the effects of the designed bispecific scFV MB102a, including isolated SEB, MB102a with different linkers, MB102a-SEB complex, MB102a-SEB-TCRß complex, MB102a-SEB-TCR-MHC II complex, and MB102a-SEB-MHC II. Our all atom molecular dynamics simulations (total 18,900 ns) confirmed that the designed single-chain bispecific antibody may allosterically prevent SEB-TCRß chain binding and inhibit SEB-TCR-MHC II formation. Subsequent analysis indicated that the binding of scFV to SEB correlates with SEB-TCR binding site motion and weakens SEB-TCR interactions.


Assuntos
Anticorpos Antibacterianos/imunologia , Anticorpos Biespecíficos/química , Anticorpos Biespecíficos/imunologia , Enterotoxinas/imunologia , Animais , Anticorpos Antibacterianos/química , Anticorpos Antibacterianos/genética , Anticorpos Biespecíficos/genética , Sítios de Ligação de Anticorpos , Simulação por Computador , Cristalografia por Raios X , Desenho de Fármacos , Enterotoxinas/química , Humanos , Camundongos , Simulação de Dinâmica Molecular , Conformação Proteica , Engenharia de Proteínas , Receptores de Antígenos de Linfócitos T/química , Receptores de Antígenos de Linfócitos T/imunologia , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia , Staphylococcus aureus/imunologia , Linfócitos T/imunologia , Linfócitos T/microbiologia
5.
mBio ; 12(2)2021 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-33879588

RESUMO

Vibrio cholerae causes the severe diarrheal disease cholera. Clinical disease and current oral cholera vaccines generate antibody responses associated with protection. Immunity is thought to be largely mediated by lipopolysaccharide (LPS)-specific antibodies, primarily targeting the O-antigen. However, the properties and protective mechanism of functionally relevant antibodies have not been well defined. We previously reported on the early B cell response to cholera in a cohort of Bangladeshi patients, from which we characterized a panel of human monoclonal antibodies (MAbs) isolated from acutely induced plasmablasts. All antibodies in that previous study were expressed in an IgG1 backbone irrespective of their original isotype. To clearly determine the impact of affinity, immunoglobulin isotype and subclass on the functional properties of these MAbs, we re-engineered a subset of low- and high-affinity antibodies in different isotype and subclass immunoglobulin backbones and characterized the impact of these changes on binding, vibriocidal, agglutination, and motility inhibition activity. While the high-affinity antibodies bound similarly to O-antigen, irrespective of isotype, the low-affinity antibodies displayed significant avidity differences. Interestingly, despite exhibiting lower binding properties, variants derived from the low-affinity MAbs had comparable agglutination and motility inhibition properties to the potently binding antibodies, suggesting that how the MAb binds to the O-antigen may be critical to function. In addition, not only pentameric IgM and dimeric IgA, but also monomeric IgA, was remarkably more potent than their IgG counterparts at inhibiting motility. Finally, analyzing highly purified F(ab) versions of these antibodies, we show that LPS cross-linking is essential for motility inhibition.IMPORTANCE Immunity to the severe diarrheal disease cholera is largely mediated by lipopolysaccharide (LPS)-specific antibodies. However, the properties and protective mechanisms of functionally relevant antibodies have not been well defined. Here, we have engineered low and high-affinity LPS-specific antibodies in different immunoglobulin backbones in order to assess the impact of affinity, immunoglobulin isotype, and subclass on binding, vibriocidal, agglutination, and motility inhibition functional properties. Importantly, we found that affinity did not directly dictate functional potency since variants derived from the low-affinity MAbs had comparable agglutination and motility inhibition properties to the potently binding antibodies. This suggests that how the antibody binds sterically may be critical to function. In addition, not only pentameric IgM and dimeric IgA, but also monomeric IgA, was remarkably more potent than their IgG counterparts at inhibiting motility. Finally, analyzing highly purified F(ab) versions of these antibodies, we show that LPS cross-linking is essential for motility inhibition.


Assuntos
Anticorpos Antibacterianos/imunologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Isotipos de Imunoglobulinas/metabolismo , Antígenos O/imunologia , Vibrio cholerae O1/imunologia , Anticorpos Antibacterianos/genética , Anticorpos Monoclonais/classificação , Anticorpos Monoclonais/genética , Sítios de Ligação de Anticorpos , Imunoglobulina A/genética , Imunoglobulina A/imunologia , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Isotipos de Imunoglobulinas/classificação , Isotipos de Imunoglobulinas/genética , Isotipos de Imunoglobulinas/imunologia , Vibrio cholerae O1/química
6.
J Clin Invest ; 131(11)2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33914704

RESUMO

Disrupting transmission of Borrelia burgdorferi sensu lato complex (B. burgdorferi) from infected ticks to humans is one strategy to prevent the significant morbidity from Lyme disease. We have previously shown that an anti-OspA human mAb, 2217, prevents transmission of B. burgdorferi from infected ticks in animal models. Maintenance of a protective plasma concentration of a human mAb for tick season presents a significant challenge for a preexposure prophylaxis strategy. Here, we describe the optimization of mAb 2217 by amino acid substitutions (2217LS: M428L and N434S) in the Fc domain. The LS mutation led to a 2-fold increase in half-life in cynomolgus monkeys. In a rhesus macaque model, 2217LS protected animals from tick transmission of spirochetes at a dose of 3 mg/kg. Crystallographic analysis of Fab in complex with OspA revealed that 2217 bound an epitope that was highly conserved among the B. burgdorferi, B. garinii, and B. afzelii species. Unlike most vaccines that may require boosters to achieve protection, our work supports the development of 2217LS as an effective preexposure prophylaxis in Lyme-endemic regions, with a single dose at the beginning of tick season offering immediate protection that remains for the duration of exposure risk.


Assuntos
Anticorpos Antibacterianos , Anticorpos Monoclonais/farmacologia , Borrelia burgdorferi , Doença de Lyme , Substituição de Aminoácidos , Animais , Anticorpos Antibacterianos/genética , Anticorpos Antibacterianos/imunologia , Anticorpos Antibacterianos/farmacologia , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Antígenos de Superfície/genética , Antígenos de Superfície/imunologia , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Vacinas Bacterianas/genética , Vacinas Bacterianas/imunologia , Borrelia burgdorferi/genética , Borrelia burgdorferi/imunologia , Modelos Animais de Doenças , Humanos , Lipoproteínas/genética , Lipoproteínas/imunologia , Doença de Lyme/tratamento farmacológico , Doença de Lyme/genética , Doença de Lyme/imunologia , Doença de Lyme/transmissão , Macaca fascicularis , Macaca mulatta , Masculino , Camundongos , Camundongos Transgênicos , Mutação de Sentido Incorreto , Carrapatos/imunologia , Carrapatos/microbiologia
7.
mBio ; 12(2)2021 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-33727348

RESUMO

Monoclonal antibody (MAb) 2C7 recognizes a lipooligosaccharide epitope expressed by most clinical Neisseria gonorrhoeae isolates and mediates complement-dependent bactericidal activity. We recently showed that a recombinant human IgG1 chimeric variant of MAb 2C7 containing an E430G Fc modification (2C7_E430G), which enhances complement activation, outperformed the parental MAb 2C7 (2C7_WT) in vivo Because natural infection with N. gonorrhoeae often does not elicit protective immunity and reinfections are common, approaches that prolong bacterial control in vivo are of great interest. Advances in DNA-based approaches have demonstrated the combined benefit of genetic engineering, formulation optimizations, and facilitated delivery via CELLECTRA-EP technology, which can induce robust in vivo expression of protective DNA-encoded monoclonal antibodies (DMAbs) with durable serum activity relative to traditional recombinant MAb therapies. Here, we created optimized 2C7-derived DMAbs encoding the parental Fc (2C7_WT) or complement-enhancing Fc variants (2C7_E430G and 2C7_E345K). 2C7 DMAbs were rapidly generated and detected throughout the 4-month study. While all complement-engaging 2C7 variants facilitated rapid clearance following primary N. gonorrhoeae challenge (day 8 after DMAb administration), the complement-enhancing 2C7_E430G variant demonstrated significantly higher potency against mice rechallenged 65 days after DMAb administration. Passive intravenous transfer of in vivo-produced, purified 2C7 DMAbs confirmed the increased potency of the complement-enhancing variants. This study highlights the ability of the DMAb platform to launch the in vivo production of antibodies engineered to promote and optimize downstream innate effector mechanisms such as complement-mediated killing, leading to hastened bacterial elimination.IMPORTANCENeisseria gonorrhoeae has become resistant to most antibiotics in clinical use. Currently, there is no safe and effective vaccine against gonorrhea. Measures to prevent the spread of gonorrhea are a global health priority. A monoclonal antibody (MAb) called 2C7, directed against a lipooligosaccharide glycan epitope expressed by most clinical isolates, displays complement-dependent bactericidal activity and hastens clearance of gonococcal vaginal colonization in mice. Fc mutations in a human IgG1 chimeric version of MAb 2C7 further enhance complement activation, and the resulting MAb displays greater activity than wild-type MAb 2C7 in vivo Here, we utilized a DNA-encoded MAb (DMAb) construct designed to launch production and assembly of "complement-enhanced" chimeric MAb 2C7 in vivo The ensuing rapid and sustained MAb 2C7 expression attenuated gonococcal colonization in mice at 8 days as well as 65 days postadministration. The DMAb system may provide an effective, economical platform to deliver MAbs for durable protection against gonorrhea.


Assuntos
Anticorpos Antibacterianos/administração & dosagem , Anticorpos Monoclonais/administração & dosagem , Vacinas Bacterianas/imunologia , Epitopos/imunologia , Gonorreia/prevenção & controle , Imunização Passiva , Imunoglobulina G/administração & dosagem , Neisseria gonorrhoeae/imunologia , Animais , Anticorpos Antibacterianos/genética , Anticorpos Antibacterianos/imunologia , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Antígenos de Bactérias/imunologia , Vacinas Bacterianas/administração & dosagem , Ativação do Complemento , Feminino , Gonorreia/imunologia , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Camundongos , Camundongos Endogâmicos BALB C
8.
Gut Microbes ; 13(1): 1-14, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33439092

RESUMO

Passive immunization with antibodies is a promising approach against enterotoxigenic Escherichia coli diarrhea, a prevalent disease in LMICs. The objective of this study was to investigate expression of a monoclonal anti-ETEC CfaE secretory IgA antibody in N. benthamiana plants, with a view to facilitating access to ETEC passive immunotherapy. SIgA1 and SIgA2 forms of mAb 68-81 were produced by co-expressing the light and engineered heavy chains with J chain and secretory component in N. benthamiana. Antibody expression and assembly were compared with CHO-derived antibodies by SDS-PAGE, western blotting, size-exclusion chromatography and LC-MS peptide mapping. N-linked glycosylation was assessed by rapid fluorescence/mass spectrometry and LC-ESI-MS. Susceptibility to gastric digestion was assessed in an in vitro model. Antibody function was compared for antigen binding, a Caco-2 cell-based ETEC adhesion assay, an ETEC hemagglutination inhibition assay and a murine in vivo challenge study. SIgA1 assembly appeared superior to SIgA2 in plants. Both sub-classes exhibited resistance to degradation by simulated gastric fluid, comparable to CHO-produced 68-61 SIgA1. The plant expressed SIgAs had more homogeneous N-glycosylation than CHO-derived SIgAs, but no alteration of in vitro functional activity was observed, including antibodies expressed in a plant line engineered for mammalian-like N glycosylation. The plant-derived SIgA2 mAb demonstrated protection against diarrhea in a murine infection model. Although antibody yield and purification need to be optimized, anti-ETEC SIgA antibodies produced in a low-cost plant platform are functionally equivalent to CHO antibodies, and provide promise for passive immunotherapy in LMICs.


Assuntos
Anticorpos Monoclonais/imunologia , Escherichia coli Enterotoxigênica/imunologia , Imunoglobulina A Secretora/imunologia , Nicotiana/metabolismo , Animais , Anticorpos Antibacterianos/genética , Anticorpos Antibacterianos/imunologia , Anticorpos Antibacterianos/metabolismo , Anticorpos Antibacterianos/uso terapêutico , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/uso terapêutico , Afinidade de Anticorpos , Aderência Bacteriana/efeitos dos fármacos , Células CACO-2 , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/terapia , Ácido Gástrico/metabolismo , Glicosilação , Humanos , Imunoglobulina A Secretora/genética , Imunoglobulina A Secretora/metabolismo , Imunoglobulina A Secretora/uso terapêutico , Imunoterapia , Camundongos , Plantas Geneticamente Modificadas , Nicotiana/genética
9.
Anticancer Res ; 40(11): 6387-6398, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33109577

RESUMO

BACKGROUND/AIM: Helicobacter pylori (Hp) infection affects a substantial proportion of the world population and is a major risk factor of gastric cancer (GC). The caveats of common Hp-tests can be evaded by a serological biomarker test (GastroPanel®, Biohit Oyj, Helsinki), the most comprehensive Hp-test on the market. The clinical validation of Helicobacter pylori IgG ELISA of the new-generation GastroPanel® test is reported. The aim of the study is to validate the clinical performance of the Helicobacter pylori IgG ELISA test in diagnosis of biopsy-confirmed Hp-infection in gastroscopy referral patients. PATIENTS AND METHODS: A cohort of 101 patients (mean age=50.1 years) referred for gastroscopy at the outpatient Department of Gastroenterology (SM Clinic, St. Petersburg) were examined by two test versions to validate the new-generation GastroPanel®. All patients were examined by gastroscopy and biopsies, which were stained with Giemsa for specific identification of Hp in the antrum (A) and corpus (C). RESULTS: Biopsy-confirmed Hp-infection was found in 64% of patients, most often confined to antrum. The overall agreement between Hp IgG ELISA and gastric biopsies in Hp-detection was 91% (95%CI=84.1-95.8%). Hp IgG ELISA diagnosed biopsy-confirmed Hp (A&C) with sensitivity (SE) of 92.3%, specificity (SP) of 88.6%, positive predictive value (PPV) of 93.8% and negative predictive value (NPV) of 86.1%, with AUC=0.904 (95%CI=0.842-0.967). In ROC analysis for Hp detection (A&C), Hp IgG ELISA shows AUC=0.978 (95%CI=0.956-1.000). CONCLUSION: The Hp IgG ELISA test successfully concludes the clinical validation process of the new-generation GastroPanel® test, which retains the unrivalled diagnostic performance of all its four biomarkers, extensively documented for the first-generation test in different clinical settings.


Assuntos
Anticorpos Antibacterianos/isolamento & purificação , Infecções por Helicobacter/diagnóstico , Helicobacter pylori/isolamento & purificação , Imunoglobulina G/isolamento & purificação , Adolescente , Adulto , Anticorpos Antibacterianos/genética , Anticorpos Antibacterianos/imunologia , Biópsia , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Gastrinas/genética , Gastrinas/isolamento & purificação , Gastrite Atrófica/diagnóstico , Gastrite Atrófica/genética , Gastrite Atrófica/microbiologia , Gastrite Atrófica/patologia , Gastroscopia/métodos , Infecções por Helicobacter/genética , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Helicobacter pylori/genética , Helicobacter pylori/patogenicidade , Humanos , Imunoglobulina G/imunologia , Masculino , Pessoa de Meia-Idade , Pepsinogênio A/genética , Pepsinogênio A/isolamento & purificação , Pepsinogênio C/genética , Pepsinogênio C/isolamento & purificação , Encaminhamento e Consulta , Estômago/microbiologia , Estômago/patologia , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/genética , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/patologia , Adulto Jovem
10.
PLoS One ; 15(1): e0227864, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31990915

RESUMO

The type VI secretion system (T6SS) is a nanomachine capable of killing adjacent microbial cells in a contact-dependent manner. Due to limited studies, relatively little is known about the range of marine bacteria that are susceptible to T6SS attack. Here, 15 diverse marine bacterial isolates from the phyla Bacteroidetes and Ɣ-Proteobacteria were challenged against the marine bacterium and human pathogen, Vibrio cholerae, which has a well described T6SS. V. cholerae killed several of the tested Ɣ-Proteobacteria, including members of the orders Vibrionales, Alteromonadales, Oceanospirillales, and Pseudomonadales. In contrast, V. cholerae co-existed with multiple Bacteroidetes and Ɣ-Proteobacteria isolates, but was killed by Vibrio coralliilyticus. Follow-up experiments revealed that five V. coralliilyticus strains, including known coral and shellfish pathogens survived the T6SS challenge and killed V. cholerae. By using predicted protein comparisons and mutagenesis, we conclude that V. coralliilyticus protected itself in the challenge by using its own T6SS to kill V. cholerae. This study provides valuable insight into the resilience and susceptibility of marine bacteria to the V. cholerae T6SS, and provides the first evidence for a functional T6SS in V. coralliilyticus, both of which have implications for human and ocean health.


Assuntos
Sistemas de Secreção Tipo VI/genética , Vibrio cholerae/patogenicidade , Vibrio/genética , Animais , Antozoários/microbiologia , Anticorpos Antibacterianos/genética , Proteínas de Bactérias/genética , Bacteroidetes/genética , Regulação Bacteriana da Expressão Gênica/genética , Proteobactérias/genética , Frutos do Mar/microbiologia , Vibrio cholerae/genética
11.
J Biol Chem ; 295(1): 55-68, 2020 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-31699895

RESUMO

Single-chain antibodies from camelids have served as powerful tools ranging from diagnostics and therapeutics to crystallization chaperones meant to study protein structure and function. In this study, we isolated a single-chain antibody from an Indian dromedary camel (ICab) immunized against a bacterial 14TM helix transporter, NorC, from Staphylococcus aureus We identified this antibody in a yeast display screen built from mononuclear cells isolated from the immunized camel and purified the antibody from Escherichia coli after refolding it from inclusion bodies. The X-ray structure of the antibody at 2.15 Å resolution revealed a unique feature within its CDR3 loop, which harbors a Zn2+-binding site that substitutes for a loop-stabilizing disulfide bond. We performed mutagenesis to compromise the Zn2+-binding site and observed that this change severely hampered antibody stability and its ability to interact with the antigen. The lack of bound Zn2+ also made the CDR3 loop highly flexible, as observed in all-atom simulations. Using confocal imaging of NorC-expressing E. coli spheroplasts, we found that the ICab interacts with the extracellular surface of NorC. This suggests that the ICab could be a valuable tool for detecting methicillin-resistant S. aureus strains that express efflux transporters such as NorC in hospital and community settings.


Assuntos
Anticorpos Antibacterianos/química , Proteínas de Bactérias/imunologia , Proteínas Associadas à Resistência a Múltiplos Medicamentos/imunologia , Anticorpos de Domínio Único/química , Zinco/metabolismo , Animais , Anticorpos Antibacterianos/genética , Anticorpos Antibacterianos/imunologia , Afinidade de Anticorpos , Sítios de Ligação , Camelus , Staphylococcus aureus Resistente à Meticilina/imunologia , Simulação de Dinâmica Molecular , Mutação , Ligação Proteica , Estabilidade Proteica , Anticorpos de Domínio Único/genética , Anticorpos de Domínio Único/imunologia , Zinco/química
12.
Indian J Med Res ; 149(3): 412-417, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-31249208

RESUMO

Background & objectives: : Shiga toxin (Stx) is produced by Shigella dysenteriae, a Gram-negative, facultative anaerobic bacillus that causes shigellosis, haemolytic uraemic syndrome (HUS) and Reiter's syndrome. The detection methods for shiga toxin needs to be rapid, accurate, reliable and must be extensively evaluated under field conditions. The aim of this study was to develop rapid, sensitive and specific detection method for Stx. Methods: : Mice and rabbits were immunized with purified recombinant Shiga toxin B (rStxB). Using these antibodies dot ELISA, sandwich ELISA and flow through assay were developed. Results: : The high-titre antibodies specifically reacted with purified rStxB. Dot-ELISA, sandwich ELISA and flow-through assay were developed and standardized that could detect StxB with limit of detection (LOD) of 9.75, 9.7 ng/ml and 0.46 µg/cassette, respectively. Interpretation & conclusions: : The rStxB was used to produce antibodies to avoid handling of pathogen. The Flow through assay 'developed was specific, rapid and field amenable.


Assuntos
Disenteria Bacilar/diagnóstico , Síndrome Hemolítico-Urêmica/diagnóstico , Toxina Shiga/isolamento & purificação , Shigella dysenteriae/genética , Animais , Anticorpos Antibacterianos/genética , Anticorpos Antibacterianos/imunologia , Artrite Reativa/diagnóstico , Artrite Reativa/genética , Artrite Reativa/microbiologia , Disenteria Bacilar/genética , Disenteria Bacilar/microbiologia , Ensaio de Imunoadsorção Enzimática , Escherichia coli/genética , Síndrome Hemolítico-Urêmica/genética , Síndrome Hemolítico-Urêmica/microbiologia , Humanos , Camundongos , Toxina Shiga/genética , Shigella dysenteriae/patogenicidade
13.
Genes (Basel) ; 10(6)2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-31142036

RESUMO

Host immunogenetic factors can affect late complications of urogenital infections with Chlamydia trachomatis. These findings are creating new avenues for updating existing risk prediction models for C. trachomatis-associated tubal factor infertility (TFI). Research into host factors and its utilization may therefore have future implications for diagnosing C. trachomatis-induced infertility. We outline the epidemiological situation regarding C. trachomatis and TFI in high-income countries. Thereupon, we review the main characteristics of the population undergoing fertility work-up and identify screening and diagnostic strategies for TFI currently in place. The Netherlands is an exemplary model for the state of the art in high-income countries. Within the framework of existing clinical approaches, we propose a scenario for the translation of relevant genome-based information into triage of infertile women, with the objective of implementing genetic profiling in the routine investigation of TFI. Furthermore, we describe the state of the art in relevant gene- and single nucleotide polymorphism (SNP) based clinical prediction models and place our perspectives in the context of these applications. We conclude that the introduction of a genetic test of proven validity into the assessment of TFI should help reduce patient burden from invasive and costly examinations by achieving a more precise risk stratification.


Assuntos
Infecções por Chlamydia/genética , Chlamydia trachomatis/genética , Testes Genéticos , Infertilidade Feminina/genética , Anticorpos Antibacterianos/genética , Anticorpos Antibacterianos/imunologia , Infecções por Chlamydia/epidemiologia , Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/isolamento & purificação , Chlamydia trachomatis/patogenicidade , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Infertilidade Feminina/diagnóstico , Infertilidade Feminina/epidemiologia , Infertilidade Feminina/microbiologia , Países Baixos/epidemiologia , Polimorfismo de Nucleotídeo Único/genética
14.
Acta Trop ; 197: 105026, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31103700

RESUMO

Brucellosis is caused by the genus Brucella. Brucella is widely distributed in cattle, swine, sheep, goat and other mammals including human. Animal brucellosis causes severe economic losses and affects related international transportation and trade. Human brucellosis causes both acute and chronic symptoms of multi-organ dysfunction. Brucella type IV secretion system (T4SS) VirB5 was required for macrophages infection and essential for virulence in mice. VirB5 is located on the cell surface and serves as a specific adhesin targeting host cell receptors. The aim of this study was to isolate and characterize a specific human domain antibody against Brucella abortus (B. abortus) VirB5 from human single domain antibody (sdAb or VHH) phage display library. Following five rounds of screening, an sdAb named as BaV5VH4 showed the highest affinity by enzyme-linked immunosorbent assay (ELISA). Its interaction with B. abortus VirB5 was verified by binding assay, dot blot and molecular docking. These findings in this paper could greatly help elucidate the molecular mechanisms of Brucella infection, and accelerate the development of sdAbs-based vaccines and neutralizing therapeutics of brucellosis.


Assuntos
Anticorpos Antibacterianos/imunologia , Brucella abortus/imunologia , Brucelose/prevenção & controle , Sequência de Aminoácidos , Animais , Anticorpos Antibacterianos/genética , Anticorpos Antibacterianos/isolamento & purificação , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/isolamento & purificação , Bacteriófagos/imunologia , Sequência de Bases , Brucella abortus/genética , Brucella abortus/isolamento & purificação , Brucelose/economia , Bovinos , Ensaio de Imunoadsorção Enzimática , Cabras , Humanos , Immunoblotting , Camundongos , Simulação de Acoplamento Molecular , Filogenia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Ovinos , Suínos , Virulência , Fatores de Virulência/metabolismo , Zoonoses/prevenção & controle
15.
Cell Rep ; 26(13): 3574-3585.e3, 2019 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-30917313

RESUMO

To understand how the interaction between an intracellular bacterium and the host immune system contributes to outcome at the site of infection, we studied leprosy, a disease that forms a clinical spectrum, in which progressive infection by the intracellular bacterium Mycobacterium leprae is characterized by the production of type I IFNs and antibody production. Dual RNA-seq on patient lesions identifies two independent molecular measures of M. leprae, each of which correlates with distinct aspects of the host immune response. The fraction of bacterial transcripts, reflecting bacterial burden, correlates with a host type I IFN gene signature, known to inhibit antimicrobial responses. Second, the bacterial mRNA:rRNA ratio, reflecting bacterial viability, links bacterial heat shock proteins with the BAFF-BCMA host antibody response pathway. Our findings provide a platform for the interrogation of host and pathogen transcriptomes at the site of infection, allowing insight into mechanisms of inflammation in human disease.


Assuntos
Hanseníase/imunologia , Hanseníase/microbiologia , Mycobacterium leprae/genética , RNA Bacteriano , RNA-Seq , Adulto , Anticorpos Antibacterianos/genética , Anticorpos Antibacterianos/imunologia , Fator Ativador de Células B/imunologia , Feminino , Interações Hospedeiro-Patógeno , Humanos , Imunidade Humoral/genética , Interferon Tipo I/metabolismo , Hanseníase/patologia , Masculino , Mycobacterium leprae/imunologia , Plasmócitos/imunologia , RNA Mensageiro , RNA Ribossômico , Transcriptoma
16.
PLoS One ; 14(1): e0210351, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30633767

RESUMO

Campylobacter jejuni (C. jejuni) is a foodborne intestinal pathogen and major cause of gastroenteritis worldwide. C. jejuni proteins that are immunogenic have been sought for their potential use in the development of biomarkers, diagnostic assays, or subunit vaccines for humans or livestock. To identify new immunogenic C. jejuni proteins, we used a native protein microarray approach. A protein chip, with over 1400 individually purified GST-tagged C. jejuni proteins, representing over 86% of the proteome, was constructed to screen for antibody titers present in test sera raised against whole C. jejuni cells. Dual detection of GST signals was incorporated as a way of normalizing the variation of protein concentrations contributing to the antibody staining intensities. We detected strong signals to 102 C. jejuni antigens. In addition to antigens recognized by antiserum raised against C. jejuni, parallel experiments were conducted to identify antigens cross-reactive to antiserum raised against various serotypes of E. coli or Salmonella or to healthy human sera. This led to the identification of 34 antigens specifically recognized by the C. jejuni antiserum, only four of which were previously known. The chip approach also allowed identification of conformational antigens. We demonstrate in the case of Cj1621 that antigen signals are lost to denaturing conditions commonly used in other approaches to identify immunogens. Antigens identified in this study include those possessing sequence features indicative of cell surface localization, as well as those that do not. Together, our results indicate that the unbiased chip-based screen can help reveal the full repertoire of host antibodies against microbial proteomes.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Campylobacter jejuni/imunologia , Campylobacter jejuni/metabolismo , Proteoma/imunologia , Proteoma/metabolismo , Animais , Anticorpos Antibacterianos/química , Anticorpos Antibacterianos/genética , Antígenos de Bactérias/química , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Infecções por Campylobacter/imunologia , Campylobacter jejuni/genética , Reações Cruzadas , Humanos , Camundongos , Análise Serial de Proteínas/métodos , Conformação Proteica , Proteoma/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Análise de Sequência de Proteína
17.
Hum Antibodies ; 27(1): 31-35, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30103311

RESUMO

BACKGROUND: Congenital toxoplasmosis is an important disease that occurs when pregnant women become infected with Toxoplasma gondii during pregnancy. The aim of this study was to investigate the presence of T. gondii B1 gene in placental tissues of IgM seronegative women. MATERIALS AND METHODS: In this research, chronic toxoplasmosis was identified through examination of blood samples in a group of pregnant women by anti-Toxoplasma IgG and IgM ELISA and nested-PCR techniques. IgG avidity test was used to estimate the onset of infection in some pregnant women with chronic infection. After delivery, some umbilical cord and neonatal blood were tested by anti-Toxoplasma IgM ELISA, and also the B1 gene of T. gondii was investigated in their placental tissue by nested-PCR. Some factors such as blood sampling time and some clinical symptoms experienced during pregnancy were recorded. RESULTS: One hundred and sixty seven out of 653 (25.6%) pregnant women were positive for anti-Toxoplasma IgG. Of them, 165 (98.8%) were negative for anti-T. gondii IgM. Six out of 10 (60%) placental tissues from IgG seropositive, IgM seronegative women were positive for T. gondii B1 gene, while anti-Toxoplasma IgM was negative in the umbilical cord and neonatal blood samples. The results of IgG avidity test showed low avidity in one and high avidity in two women's samples. The B1 gene was not found in the blood samples of any of the six mothers. The most symptoms experienced during pregnancy were headache and nausea. CONCLUSION: The detection of B1 gene in placental tissues of the healthy newborn infants reiterates that presence of T. gondii in the placenta does not always result in congenital toxoplasmosis.


Assuntos
Genes de Protozoários , Técnicas de Diagnóstico Molecular , Placenta/química , Toxoplasma/genética , Toxoplasmose Congênita/diagnóstico , Toxoplasmose Congênita/genética , Adulto , Anticorpos Antibacterianos/análise , Anticorpos Antibacterianos/genética , Afinidade de Anticorpos , DNA Bacteriano/genética , DNA Bacteriano/isolamento & purificação , DNA de Protozoário , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imunoglobulina G/análise , Imunoglobulina G/imunologia , Imunoglobulina M/análise , Imunoglobulina M/imunologia , Recém-Nascido , Reação em Cadeia da Polimerase , Gravidez , Reprodutibilidade dos Testes
18.
Front Immunol ; 9: 2072, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30254643

RESUMO

Tissue factor pathway inhibitor-2 (TFPI-2) has previously been characterized as an endogenous anticoagulant. TFPI-2 is expressed in the vast majority of cells, mainly secreted into the extracellular matrix. Recently we reported that EDC34, a C-terminal peptide derived from TFPI-2, exerts a broad antimicrobial activity. In the present study, we describe a previously unknown antimicrobial mode of action for the human TFPI-2 C-terminal peptide EDC34, mediated via binding to immunoglobulins of the classes IgG, IgA, IgE, and IgM. In particular the interaction of EDC34 with the Fc part of IgG is of importance since this boosts interaction between the immunoglobulin and complement factor C1q. Moreover, we find that the binding increases the C1q engagement of the antigen-antibody interaction, leading to enhanced activation of the classical complement pathway during bacterial infection. In experimental murine models of infection and endotoxin challenge, we show that TFPI-2 is up-regulated in several organs, including the lung. Correspondingly, TFPI-2-/- mice are more susceptible to pulmonary Pseudomonas aeruginosa bacterial infection. No anti-coagulant role of TFPI-2 was observed in these models in vivo. Furthermore, in vivo, the mouse TFPI-2-derived C-terminal peptide VKG24, a homolog to human EDC34 is protective against systemic Escherichia coli bacterial infection. Moreover, in sputum from cystic fibrosis patients TFPI-2 C-terminal fragments are generated and found associated with immunoglobulins. Together our data describe a previously unknown host defense mechanism and therapeutic importance of TFPI-2 against invading Gram-negative bacterial pathogens.


Assuntos
Anticorpos Antibacterianos/imunologia , Infecções por Escherichia coli/imunologia , Escherichia coli/imunologia , Glicoproteínas/imunologia , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Animais , Anticorpos Antibacterianos/genética , Infecções por Escherichia coli/genética , Infecções por Escherichia coli/patologia , Glicoproteínas/genética , Humanos , Camundongos , Camundongos Knockout , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Infecções por Pseudomonas/genética , Infecções por Pseudomonas/patologia , Regulação para Cima/imunologia
19.
Dev Comp Immunol ; 89: 141-151, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30142358

RESUMO

Interleukin 6 (IL-6), a pleiotropic cytokine, plays an important role in humoral immune response, not only inducing the differentiation of B cells into plasma cells, but also promoting antibody-secreting cells (ASCs) to produce antibodies. In this study, Nile tilapia (Oreochromis niloticus) IL-6 (OnIL-6) was identified and characterized at expression level in response to bacterial infection and promotion of antibody production. The open reading frame of OnIL-6 ORF is consisted of 663 bp encoding a polypeptide of 220 amino acids. The deduced OnIL-6 protein contained an IL-6/G-CSF family signature, two conserved cysteine, and four α-helix bundles, which was highly homologous to other species. Spatial mRNA expression analysis revealed that the highest expression of OnIL-6 was observed in the thymus. After in vivo challenges of lipopolysaccharide (LPS) and Streptococcus agalactia (S. agalactiae), OnIL-6 expressions were significantly up-regulated in head kidney and spleen. The similar up-regulation of OnIL-6 was observed in the head kidney and spleen leukocytes in vitro stimulation with LPS and S. agalactiae. In addition, inducement with the recombinant OnIL-6 ((r)OnIL-6) in vitro caused significant increases in expressions of both sIgM and mIgM. Moreover, the (r)OnIL-6 stimulation enhanced the secretion of sIgM (more especially in P50 plasma-like B cells) and the production of mIgM in P60 and P70 B cell subsets (resting B cells, activated B cells and plasmablast-like B cells) in vitro. Taken together, this study indicated that OnIL-6 might be involved in host defense against bacterial infection and promote the production of antibody in Nile tilapia.


Assuntos
Ciclídeos/imunologia , Proteínas de Peixes/imunologia , Interleucina-6/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antibacterianos/biossíntese , Anticorpos Antibacterianos/genética , Subpopulações de Linfócitos B/imunologia , Ciclídeos/genética , Ciclídeos/microbiologia , Doenças dos Peixes/imunologia , Proteínas de Peixes/biossíntese , Proteínas de Peixes/genética , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Imunoglobulina M/biossíntese , Imunoglobulina M/genética , Interleucina-6/química , Interleucina-6/genética , Modelos Moleculares , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Homologia de Sequência de Aminoácidos , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/veterinária , Streptococcus agalactiae/imunologia , Streptococcus agalactiae/patogenicidade
20.
Appl Microbiol Biotechnol ; 102(16): 6899-6913, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29862446

RESUMO

Infection with Helicobacter pylori may result in the emergence of gastric adenocarcinoma. Among various toxins assisting pathogenesis of H. pylori, the vacuolating cytotoxin A (VacA) is one of the most potent toxins known as the major cause of the peptic ulcer and gastric adenocarcinoma. To isolate single-chain variable fragments (scFvs) against two conserved regions of VacA, we capitalized on the phage display technology and a solution-phase biopanning (SPB). Characterization of scFvs was carried out by enzyme-linked immunosorbent assay (ELISA), immunoblotting, and surface plasmon resonance (SPR). Bioinformatics analyses were also performed in order to characterize the structural and functional properties of the isolated scFvs and the interaction(s) between the isolated antibodies (Ab)-antigen (Ag). After four rounds of biopanning, the positive colonies detected by scFv ELISA were harvested to extract the plasmids and perform sequencing. Of several colonies, three colonies showed high affinity to the VacA1 and two colonies for the VacA2. Further complementary examinations (e.g., sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE), western blot, SPR, and flow cytometry) displayed the high affinity and specificity of the isolated scFvs to the VacA. Docking results revealed the interaction of the complementarity-determining regions (CDRs) with the VacA peptide. In conclusion, for the first time, we report on the isolation of several scFvs against conserved residues of VacA toxin with high affinity and specificity, which may be used as novel diagnostic/therapeutic tool in the H. pylori infection.


Assuntos
Anticorpos Antibacterianos/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Anticorpos Antibacterianos/genética , Western Blotting , Técnicas de Visualização da Superfície Celular , Sequência Conservada/genética , Ensaio de Imunoadsorção Enzimática , Infecções por Helicobacter/diagnóstico , Helicobacter pylori/química , Helicobacter pylori/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA