Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Nature ; 629(8013): 878-885, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38720086

RESUMEN

The COVID-19 pandemic underscored the promise of monoclonal antibody-based prophylactic and therapeutic drugs1-3 and revealed how quickly viral escape can curtail effective options4,5. When the SARS-CoV-2 Omicron variant emerged in 2021, many antibody drug products lost potency, including Evusheld and its constituent, cilgavimab4-6. Cilgavimab, like its progenitor COV2-2130, is a class 3 antibody that is compatible with other antibodies in combination4 and is challenging to replace with existing approaches. Rapidly modifying such high-value antibodies to restore efficacy against emerging variants is a compelling mitigation strategy. We sought to redesign and renew the efficacy of COV2-2130 against Omicron BA.1 and BA.1.1 strains while maintaining efficacy against the dominant Delta variant. Here we show that our computationally redesigned antibody, 2130-1-0114-112, achieves this objective, simultaneously increases neutralization potency against Delta and subsequent variants of concern, and provides protection in vivo against the strains tested: WA1/2020, BA.1.1 and BA.5. Deep mutational scanning of tens of thousands of pseudovirus variants reveals that 2130-1-0114-112 improves broad potency without increasing escape liabilities. Our results suggest that computational approaches can optimize an antibody to target multiple escape variants, while simultaneously enriching potency. Our computational approach does not require experimental iterations or pre-existing binding data, thus enabling rapid response strategies to address escape variants or lessen escape vulnerabilities.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Simulación por Computador , Diseño de Fármacos , SARS-CoV-2 , Animales , Femenino , Humanos , Ratones , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/química , Anticuerpos Antivirales/inmunología , COVID-19/inmunología , COVID-19/virología , Mutación , Pruebas de Neutralización , SARS-CoV-2/clasificación , SARS-CoV-2/genética , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología , Análisis Mutacional de ADN , Deriva y Cambio Antigénico/genética , Deriva y Cambio Antigénico/inmunología , Diseño de Fármacos/métodos
2.
Infect Immun ; 82(8): 3324-32, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24866803

RESUMEN

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


Asunto(s)
Actividad Bactericida de la Sangre , Borrelia/inmunología , Proteínas Portadoras/metabolismo , Proteínas del Sistema Complemento/inmunología , Fiebre Recurrente/inmunología , Fiebre Recurrente/microbiología , Factores de Virulencia/metabolismo , Animales , Borrelia/genética , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Eliminación de Gen , Humanos , Ratones , Factores de Virulencia/genética
3.
bioRxiv ; 2023 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-36324800

RESUMEN

The COVID-19 pandemic underscored the promise of monoclonal antibody-based prophylactic and therapeutic drugs1-3, but also revealed how quickly viral escape can curtail effective options4,5. With the emergence of the SARS-CoV-2 Omicron variant in late 2021, many clinically used antibody drug products lost potency, including Evusheld™ and its constituent, cilgavimab4,6. Cilgavimab, like its progenitor COV2-2130, is a class 3 antibody that is compatible with other antibodies in combination4 and is challenging to replace with existing approaches. Rapidly modifying such high-value antibodies with a known clinical profile to restore efficacy against emerging variants is a compelling mitigation strategy. We sought to redesign COV2-2130 to rescue in vivo efficacy against Omicron BA.1 and BA.1.1 strains while maintaining efficacy against the contemporaneously dominant Delta variant. Here we show that our computationally redesigned antibody, 2130-1-0114-112, achieves this objective, simultaneously increases neutralization potency against Delta and many variants of concern that subsequently emerged, and provides protection in vivo against the strains tested, WA1/2020, BA.1.1, and BA.5. Deep mutational scanning of tens of thousands pseudovirus variants reveals 2130-1-0114-112 improves broad potency without incurring additional escape liabilities. Our results suggest that computational approaches can optimize an antibody to target multiple escape variants, while simultaneously enriching potency. Because our approach is computationally driven, not requiring experimental iterations or pre-existing binding data, it could enable rapid response strategies to address escape variants or pre-emptively mitigate escape vulnerabilities.

4.
PLoS One ; 17(6): e0267796, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35657812

RESUMEN

The current global COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in a public health crisis with more than 168 million cases reported globally and more than 4.5 million deaths at the time of writing. In addition to the direct impact of the disease, the economic impact has been significant as public health measures to contain or reduce the spread have led to country wide lockdowns resulting in near closure of many sectors of the economy. Antibodies are a principal determinant of the humoral immune response to COVID-19 infections and may have the potential to reduce disease and spread of the virus. The development of monoclonal antibodies (mAbs) represents a therapeutic option that can be produced at large quantity and high quality. In the present study, a mAb combination mixture therapy was investigated for its capability to specifically neutralize SARS-CoV-2. We demonstrate that each of the antibodies bind the spike protein and neutralize the virus, preventing it from infecting cells in an in vitro cell-based assay, including multiple viral variants that are currently circulating in the human population. In addition, we investigated the effects of two different mutations in the Fc portion (YTE and LALA) of the antibody on Fc effector function and the ability to alleviate potential antibody-dependent enhancement of disease. These data demonstrate the potential of a combination of two mAbs that target two different epitopes on the SARS-CoV2 spike protein to provide protection against SARS-CoV-2 infection in humans while extending serum half-life and preventing antibody-dependent enhancement of disease.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , SARS-CoV-2 , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes , Anticuerpos Antivirales/uso terapéutico , Control de Enfermedades Transmisibles , Humanos , Pandemias , ARN Viral , Glicoproteína de la Espiga del Coronavirus
5.
Med ; 3(3): 188-203.e4, 2022 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-35132398

RESUMEN

BACKGROUND: Human monoclonal antibody (mAb) treatments are promising for COVID-19 prevention or therapy. The pre-exposure prophylactic efficacy of neutralizing antibodies that are engineered with mutations to extend their persistence in human serum and the neutralizing antibody titer in serum required for protection against SARS-CoV-2 infection remain poorly characterized. METHODS: The Fc region of two neutralizing mAbs (COV2-2130 and COV2-2381) targeting non-overlapping epitopes on the receptor binding domain of SARS-CoV-2 spike protein was engineered to extend their persistence in humans and reduce interactions with Fc gamma receptors. We assessed protection by individual antibodies or a combination of the two antibodies (designated ADM03820) given prophylactically by an intravenous or intramuscular route in a non-human primate (NHP) model of SARS-CoV-2 infection. FINDINGS: Passive transfer of individual mAbs or ADM03820 conferred virological protection in the NHP respiratory tract in a dose-dependent manner, and ADM03820 potently neutralized SARS-CoV-2 variants of concern in vitro. We defined a protective serum-neutralizing antibody titer and concentration in NHPs for passively transferred human antibodies that acted by direct viral neutralization. CONCLUSIONS: In summary, we demonstrate that neutralizing antibodies with extended half-life and lacking Fc-mediated effector functions are efficient for pre-exposure prophylaxis of SARS-CoV-2 infection in NHPs. These results support clinical development of ADM03820 for COVID-19 prevention. FUNDING: This research was supported by a contract from the JPEO-CBRND (W911QY-20-9-003, 20-05); the Joint Sciences and Technology Office and Joint Program Executive Office (MCDC-16-01-002 JSTO, JPEO); a DARPA grant (HR0011-18-2-0001); an NIH grant (R01 AI157155); and the 2019 Future Insight Prize from Merck KGaA.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Anticuerpos Monoclonales , Anticuerpos Neutralizantes/uso terapéutico , COVID-19/prevención & control , Humanos , Macaca , Glicoproteína de la Espiga del Coronavirus
6.
J Bacteriol ; 193(13): 3241-5, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21551306

RESUMEN

Tick-borne relapsing fever (TBRF) is a spirochetal disease caused by at least 15 different Borrelia species. It is a serious human health concern in regions of endemicity throughout the world. Transmission to humans occurs through the bites of infected Ornithodoros ticks. In North America, the primary Borrelia species associated with human disease are B. hermsii and B. turicatae. Direct demonstration of the role of putative TBRF spirochete virulence factors in the disease process has been hindered by the lack of a genetic manipulation system and complete genome sequences. Expanding on recent developments in these areas, here we demonstrate the successful generation of a clone of B. hermsii YOR that constitutively produces green fluorescent protein (GFP) (B. hermsii YOR::kan gfp). This strain was generated through introduction of a kan-gfp cassette into a noncoding region of the 200-kb B. hermsii linear plasmid lp200. Genetic manipulation did not affect the growth rate or trigger the loss of native plasmids. B. hermsii YOR::kan gfp retained infectivity and elicited host seroconversion. Stable production of GFP was demonstrated both in vitro and in vivo. This study represents a significant step forward in the development of tools that can be employed to study the virulence mechanisms of TBRF spirochetes.


Asunto(s)
Borrelia/genética , Expresión Génica , Proteínas Fluorescentes Verdes/biosíntesis , Biología Molecular/métodos , Plásmidos/genética , Transformación Bacteriana , Animales , Anticuerpos Antibacterianos/sangre , Borrelia/crecimiento & desarrollo , Borrelia/inmunología , Borrelia/patogenicidad , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Inestabilidad Genómica , Ratones , Mutagénesis Insercional , Recombinación Genética , Fiebre Recurrente/microbiología , Virulencia
7.
Mol Microbiol ; 76(2): 393-408, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20199597

RESUMEN

Borrelia burgdorferi outer surface protein C (ospC) is required for the establishment of infection in mammals. However, its precise function remains controversial. The biologically active form of OspC appears to be a homodimer. Alpha helix 1 and 1' of the apposing monomers form a solvent-accessible pocket at the dimeric interface that presents a putative ligand-binding domain (LBD1). Here we employ site-directed and allelic-exchange mutagenesis to test the hypothesis that LBD1 is a determinant of OspC function in the mammalian environment. Substitution of residues K60, E61 and E63 which line LBD1 resulted in the loss of infectivity or influenced dissemination. Analyses of the corresponding recombinant proteins demonstrated that the loss of function was not due to structural perturbation, impaired dimer formation or the loss of plasminogen binding. This study is the first to assess the involvement of individual residues and domains of OspC in its in vivo function. The data support the hypothesis that OspC interacts with a mammalian derived ligand that is critical for survival during early infection. These results shed new light on the structure-functions relationships of OspC and challenge existing hypotheses regarding OspC function in mammals.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Borrelia burgdorferi/patogenicidad , Factores de Virulencia/metabolismo , Sustitución de Aminoácidos/genética , Animales , Antígenos Bacterianos/genética , Proteínas de la Membrana Bacteriana Externa/genética , Sitios de Unión , Borrelia burgdorferi/genética , Dimerización , Técnicas de Inactivación de Genes , Ratones , Ratones Endogámicos C3H , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Plasminógeno/metabolismo , Unión Proteica , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Factores de Virulencia/genética
8.
Microbiol Resour Announc ; 9(50)2020 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-33303672

RESUMEN

The genome of Francisella tularensis live vaccine strain NR-28537 was sequenced by a hybrid approach utilizing an Oxford Nanopore Technologies R9 flow cell and an Illumina MiSeq platform. De novo assembly of the resulting long and short reads produced a single-contig whole-genome sequence.

9.
Vaccine ; 38(8): 1915-1924, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-31959423

RESUMEN

Experimental Outer surface protein (Osp) C based subunit chimeritope vaccinogens for Lyme disease (LD) were assessed for immunogenicity, structure, ability to elicit antibody (Ab) responses to divergent OspC proteins, and bactericidal activity. Chimeritopes are chimeric epitope based proteins that consist of linear epitopes derived from multiple proteins or multiple variants of a protein. An inherent advantage to chimeritope vaccinogens is that they can be constructed to trigger broadly protective Ab responses. Three OspC chimeritope proteins were comparatively assessed: Chv1, Chv2 and Chv3. The Chv proteins possess the same set of 18 linear epitopes derived from 9 OspC type proteins but differ in the physical ordering of epitopes or by the presence or absence of linkers. All Chv proteins were immunogenic in mice and rats eliciting high titer Ab. Immunoblot and enzyme linked immunosorbent assays demonstrated that the Chv proteins elicit IgG that recognizes a diverse array of OspC type proteins. The panel included OspC proteins produced by N. American and European strains of the LD spirochetes. Rat anti-Chv antisera uniformly labeled intact, non-permeabilized Borreliella burgdorferi demonstrating that vaccinal Ab can bind to targets that are naturally presented on the spirochete cell surface. Vaccinal Ab also displayed potent complement dependent-Ab mediated killing activity. This study highlights the ability of OspC chimeritopes to serve as vaccinogens that trigger potentially broadly protective Ab responses. In addition to the current use of an OspC chimeritope in a canine LD vaccine, chimeritopes can serve as key components of human LD subunit vaccines.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Borrelia burgdorferi , Lipoproteínas/inmunología , Vacunas contra Enfermedad de Lyme/inmunología , Enfermedad de Lyme/prevención & control , Animales , Anticuerpos Antibacterianos/inmunología , Borrelia burgdorferi/inmunología , Epítopos/inmunología , Ratones , Ratas
10.
Vaccine ; 37(17): 2401-2407, 2019 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-30922701

RESUMEN

As Ixodes ticks spread to new regions, the incidence of Lyme disease (LD) in companion animals and humans will increase. Preventive strategies for LD in canines center on vaccination and tick control (acaricides). Both subunit and bacterin based LD veterinary vaccines are available. Outer surface protein C (OspC), a potent immunogen and dominant early antigen, has been demonstrated to elicit protective antibody (Ab) responses. However, a single OspC protein elicits a relatively narrow range of protection. There are conflicting reports as to whether the immunodominant epitopes of OspC reside within variable or conserved domains. A detailed understanding of the antigenic determinants of OspC is essential for understanding immune responses to this essential virulence factor and vaccinogen. Here, we investigate the contribution of the conserved C-terminal C10 motif in OspC triggered Ab responses. Using a panel of diverse recombinant full length OspC proteins and their corresponding C10 deletion variants (OspCΔC10), we demonstrate that the C10 motif does not significantly contribute to immunization or infection induced Ab responses in rabbits, rats, canines, horses and non-human primates. Furthermore, the C10 motif is not required to trigger potent bactericidal Ab responses. This study provides insight into the antigenic structure of OspC. The results enhance our understanding of immune responses that develop during infection or upon vaccination and have implications for interpretation of LD diagnostic assays that employ OspC.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Borrelia burgdorferi/inmunología , Epítopos/inmunología , Enfermedad de Lyme/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Formación de Anticuerpos , Perros , Ensayo de Inmunoadsorción Enzimática , Caballos , Humanos , Enfermedad de Lyme/prevención & control , Primates , Conejos , Proteínas Recombinantes/inmunología , Vacunación
11.
Front Microbiol ; 10: 1923, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31507550

RESUMEN

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

12.
Toxins (Basel) ; 11(4)2019 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-30959899

RESUMEN

Botulinum neurotoxins (BoNT) are some of the most toxic proteins known, with a human LD50 of ~1 ng/kg. Equine antitoxin has a half-life in circulation of less than 1 day and is limited to a treatment rather than a prevention indication. The development of monoclonal antibodies (mAbs) may represent an alternative therapeutic option that can be produced at high quantities and of high quality and with half-lives of >10 days. Two different three mAb combinations are being developed that specifically neutralize BoNT serotypes A (BoNT/A) and B (BoNT/B). We investigated the pharmacokinetics of the anti-BoNT/A and anti-BoNT/B antibodies in guinea pigs (Cavia porcellus) and their ability to protect guinea pigs against an aerosol challenge of BoNT/A1 or BoNT/B1. Each antibody exhibited dose-dependent exposure and reached maximum circulating concentrations within 48 h post intraperitoneal or intramuscular injection. A single intramuscular dose of the three mAb combination protected guinea pigs against an aerosol challenge dose of 93 LD50 of BoNT/A1 and 116 LD50 of BoNT/B1 at 48 h post antibody administration. These mAbs are effective in preventing botulism after an aerosol challenge of BoNT/A1 and BoNT/B1 and may represent an alternative to vaccination to prevent type A or B botulism in those at risk of BoNT exposure.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Toxinas Botulínicas Tipo A/inmunología , Botulismo/prevención & control , Aerosoles , Animales , Anticuerpos Monoclonales/farmacocinética , Toxinas Botulínicas Tipo A/administración & dosificación , Sinergismo Farmacológico , Quimioterapia Combinada , Cobayas , Dosificación Letal Mediana , Masculino , Ratones Endogámicos ICR , Serogrupo
13.
Vaccine ; 35(24): 3178-3185, 2017 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-28479174

RESUMEN

The lipoprotein OspA is produced by the Lyme disease spirochetes primarily in unfed ticks. OspA production is down-regulated by the blood meal and it is not produced in mammals except for possible transient production during late stage infection in patients with Lyme arthritis. Vaccination with OspA elicits antibody (Ab) that can target spirochetes in the tick midgut during feeding and inhibit transmission to mammals. OspA was the primary component of the human LYMErix™ vaccine. LYMErix™ was available from 1998 to 2002 but then pulled from the market due to declining sales as a result of unsubstantiated concerns about vaccination induced adverse events and poor efficacy. It was postulated that a segment of OspA that shares sequence similarity with a region in human LFA-1 and may trigger putative autoimmune events. While evidence supporting such a link has not been demonstrated, most efforts to move forward with OspA as a vaccine component have sought to eliminate this region of concern. Here we identify an OspA linear epitope localized within OspA amino acid residues 221-240 (OspA221-240) that lacks the OspA region suggested to elicit autoimmunity. A peptide consisting of residues 221-240 was immunogenic in mice. Ab raised against OspA221-240 peptide surface labeled B. burgdorferi in IFAs and displayed potent Ab mediated-complement dependent bactericidal activity. BLAST analyses identified several variants of OspA221-240 and a closely related sequence in OspB. It is our hypothesis that integration of the OspA221-240 epitope into a multivalent-OspC based chimeric epitope based vaccine antigen (chimeritope) could result in a subunit vaccine that protects against Lyme disease through synergistic mechanisms.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Antígenos de Superficie/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Vacunas Bacterianas/inmunología , Borrelia burgdorferi/inmunología , Epítopos/inmunología , Lipoproteínas/inmunología , Vacunas contra Enfermedad de Lyme/inmunología , Péptidos/inmunología , Animales , Antígenos Bacterianos/inmunología , Antígenos de Superficie/química , Antígenos de Superficie/genética , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/genética , Vacunas Bacterianas/química , Vacunas Bacterianas/genética , Borrelia burgdorferi/química , Borrelia burgdorferi/genética , Borrelia burgdorferi/crecimiento & desarrollo , Descubrimiento de Drogas , Epítopos/química , Inmunoglobulina G/sangre , Ixodes/microbiología , Lipoproteínas/química , Lipoproteínas/genética , Enfermedad de Lyme/prevención & control , Ratones , Péptidos/administración & dosificación , Péptidos/química , Reacción en Cadena de la Polimerasa , Determinación de Anticuerpos Séricos Bactericidas , Vacunación , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología
14.
Vet J ; 218: 27-33, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27938705

RESUMEN

OspC performs essential functions during the enzootic cycle of the Lyme disease (LD) spirochetes. In this study, the specificity of antibody (Ab) responses to OspC was profiled to define the antigenic determinants during infection and after vaccination. Several OspC variants or 'types' were screened with serum from SNAP4Dx C6 positive dogs and with serum from rabbits hyperimmunized with OspC proteins. The OspC type-specific nature of the Ab response revealed that variable domains of OspC are immunodominant during infection and upon vaccination. To assess the potential of OspC to elicit Ab in the context of a bacterin vaccine, OspC production in strains cultivated in vitro was assessed. Immunoblot and indirect immunofluorescent antibody analyses demonstrated that production is low and that only a subset of cells actively produces OspC in vitro, raising questions about the potential of bacterin vaccines to stimulate significant anti-OspC Ab responses. The specificity of the OspC Ab response in experimentally infected mice over time was assessed to determine if domains shielded in the OspC homodimer become accessible and stimulate Ab production as infection progresses. The results demonstrate that the OspC Ab response remains focused on surface exposed variable regions of the protein throughout infection. In contrast to some earlier studies, it is concluded that conserved domains of OspC, including the C7 or C10 domain, do not elicit significant Ab responses during infection or upon vaccination. Collectively, the results indicate that OspC diversity must be considered in vaccine design and in the interpretation of diagnostic assays that employ OspC as a diagnostic antigen.


Asunto(s)
Antígenos Bacterianos/genética , Proteínas de la Membrana Bacteriana Externa/genética , Borrelia burgdorferi/inmunología , Enfermedades de los Perros/prevención & control , Inmunización/veterinaria , Vacunas contra Enfermedad de Lyme/inmunología , Enfermedad de Lyme/prevención & control , Enfermedades de los Roedores/prevención & control , Animales , Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Enfermedades de los Perros/microbiología , Perros , Epítopos/sangre , Inmunoglobulina G/inmunología , Enfermedad de Lyme/microbiología , Ratones , Conejos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Enfermedades de los Roedores/microbiología
15.
Int J Parasitol ; 35(2): 171-84, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15710438

RESUMEN

The protozoan oyster parasite Perkinsus marinus releases a complex set of extracellular products (ECP) during in vitro culture. These products have been previously implicated in parasite virulence, and their expression can be altered by medium supplementation with oyster tissue homogenate. Little is known regarding ECP function, regulation, or mechanism of storage and release. Perkinsus marinus ECP were purified from a protein-free medium and used to produce a panel of five monoclonal antibodies. Several of the antibodies recognised series of proteins implying that the ECP may originate from comparatively few parental molecules. The ECP are secreted by several pathways, including the release of one product from an external cell layer, and two other products from two morphologically distinct intracellular compartments. Antibodies against separate epitopes on one protein provided information about possible protein structure. A sandwich ELISA format allowed sensitive quantification of that protein and showed significantly reduced protein expression in oyster tissue homogenate supplemented cultures. Immunopurification allowed tandem mass spectroscopic amino acid sequencing of that protein. Another antibody was used to characterise the P. marinus cell wall. This antibody specifically bound to trophozoite and tomont walls, and was used to investigate the morphological and antigenic changes in these walls during Ray's fluid thioglycollate medium-induced formation of hypnospores. It was also used to confirm that oyster tissue homogenate supplementation could induce formation of hypnospores. This antibody labeled P. marinus cells in fixed oyster tissue in a species-specific manner.


Asunto(s)
Anticuerpos Monoclonales/análisis , Eucariontes/inmunología , Ostreidae/parasitología , Secuencia de Aminoácidos , Animales , Ensayo de Inmunoadsorción Enzimática/métodos , Eucariontes/ultraestructura , Espacio Extracelular/inmunología , Técnica del Anticuerpo Fluorescente/métodos , Inmunohistoquímica/métodos , Microscopía Electrónica , Proteínas Nucleares/análisis , Proteínas Protozoarias/análisis , Proteínas Protozoarias/inmunología , Proteína p107 Similar a la del Retinoblastoma , Virulencia
16.
J Parasitol ; 91(3): 689-91, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16108568

RESUMEN

The extracellular products (ECPs) of the oyster parasite Perkinsus marinus have been posed to contain virulence factors, including serine proteases. Supplementation of P. marinus cultures with oyster homogenates enhances infectivity and changes the ECP composition. Therefore, subtractive immunization was used to attempt creation of antibodies to proteins unique to ECPs produced following parasite exposure to oyster homogenates. While control mice remained competent to respond to an unrelated antigen, no serum titers against novel ECP epitopes were detected in experimental mice. Attempts to create discriminatory hybridomas resulted in no clones with anti-ECP specificity. These findings suggest that, because no unique epitopes can be found within ECPs generated following exposure of P. marinus to host homogenates, the changes to ECPs are greatly constrained.


Asunto(s)
Epítopos/inmunología , Eucariontes/inmunología , Ostreidae/química , Infecciones Protozoarias en Animales/inmunología , Factores de Virulencia/inmunología , Animales , Medios de Cultivo , Ciclofosfamida/farmacología , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Eucariontes/enzimología , Eucariontes/patogenicidad , Femenino , Hibridomas , Inmunización/métodos , Inmunosupresores/farmacología , Ratones , Ratones Endogámicos BALB C , Ostreidae/inmunología , Serina Endopeptidasas/inmunología , Serina Endopeptidasas/metabolismo , Tinción con Nitrato de Plata , Factores de Virulencia/metabolismo
17.
J Immunol Methods ; 278(1-2): 67-78, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12957397

RESUMEN

Concentrated culture supernatants containing the extracellular products (ECP) of the protozoan oyster parasite Perkinsus marinus were used to immunize mice. This preparation, produced by ultrafiltration, was found to be both poorly immunogenic and toxic to experimental animals. The possibility that these effects were due to toxic parasite products and/or medium constituents was examined. Co-administration of this material with highly immunogenic oyster hemolymph caused a substantive suppression of the specific antibody response to hemolymph, as well as a decrease in the number of epitopes recognized. Potential protein/protease toxin-mediated causes of the immunosuppression were addressed by heat denaturation and proteolytic inhibition of the concentrate; neither substantially enhanced immunogenicity. Analysis of media constituents revealed that the known immunomodulatory surfactant, Pluronic F-68 (PF68), used in the defined lipid concentrate supplement, was capable of eliciting significant immunosuppression. Although isolated protein antigens from P. marinus ECP remain highly immunosuppressive, separation of the protein antigens from the PF68 has enabled production of polyclonal antisera with a broader recognition of antigens.


Asunto(s)
Medios de Cultivo Condicionados/química , Medios de Cultivo Condicionados/farmacología , Inmunoglobulina G/sangre , Proteínas Protozoarias/inmunología , Vacunación/métodos , Animales , Formación de Anticuerpos , Western Blotting , Cromatografía por Intercambio Iónico , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática , Femenino , Hemolinfa/inmunología , Terapia de Inmunosupresión , Ratones , Ratones Endogámicos BALB C , Ostreidae , Fluoruro de Fenilmetilsulfonilo/farmacología , Poloxámero/química , Poloxámero/farmacología , Desnaturalización Proteica
18.
Pathog Dis ; 70(2): 176-84, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24376161

RESUMEN

OspC is produced by all species of the Borrelia burgdorferi sensu lato complex and is required for infectivity in mammals. To test the hypothesis that the conserved C-terminal motif (C10) of OspC is required for function in vivo, a mutant B. burgdorferi strain (B31::ospCΔC10) was created in which ospC was replaced with an ospC gene lacking the C10 motif. The ability of the mutant to infect mice was investigated using tick transmission and needle inoculation. Infectivity was assessed by cultivation, qRT-PCR, and measurement of IgG antibody responses. B31::ospCΔC10 retained the ability to infect mice by both needle and tick challenge and was competent to survive in ticks after exposure to the blood meal. To determine whether recombinant OspC protein lacking the C-terminal 10 amino acid residues (rOspCΔC10) can bind plasminogen, the only known mammalian-derived ligand for OspC, binding analyses were performed. Deletion of the C10 motif resulted in a statistically significant decrease in plasminogen binding. Although deletion of the C10 motif influenced plasminogen binding, it can be concluded that the C10 motif is not required for OspC to carry out its critical in vivo functions in tick to mouse transmission.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Grupo Borrelia Burgdorferi/fisiología , Enfermedad de Lyme/transmisión , Factores de Virulencia/metabolismo , Secuencias de Aminoácidos , Animales , Antígenos Bacterianos/genética , Adhesión Bacteriana , Proteínas de la Membrana Bacteriana Externa/genética , Grupo Borrelia Burgdorferi/genética , Análisis Mutacional de ADN , Ratones Endogámicos C3H , Plasminógeno/metabolismo , Eliminación de Secuencia , Garrapatas
19.
Clin Vaccine Immunol ; 18(6): 901-6, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21525304

RESUMEN

Borrelia burgdorferi OspC is an outer membrane lipoprotein required for the establishment of infection in mammals. Due to its universal distribution among B. burgdorferi sensu lato strains and high antigenicity, it is being explored for the development of a next-generation Lyme disease vaccine. An understanding of the surface presentation of OspC will facilitate efforts to maximize its potential as a vaccine candidate. OspC forms homodimers at the cell surface, and it has been hypothesized that it may also form oligomeric arrays. Here, we employ site-directed mutagenesis to test the hypothesis that interdimeric disulfide bonds at cysteine 130 (C130) mediate oligomerization. B. burgdorferi B31 ospC was replaced with a C130A substitution mutant to yield strain B31::ospC(C130A). Recombinant protein was also generated. Disulfide-bond-dependent oligomer formation was demonstrated and determined to be dependent on C130. Oligomerization was not required for in vivo function, as B31::ospC(C130A) retained infectivity and disseminated normally. The total IgG response and the induced isotype pattern were similar between mice infected with untransformed B31 and those infected with the B31::ospC(C130A) strain. These data indicate that the immune response to OspC is not significantly altered by formation of OspC oligomers, a finding that has significant implications in Lyme disease vaccine design.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Disulfuros , Vacunas contra Enfermedad de Lyme/metabolismo , Multimerización de Proteína , Factores de Virulencia/metabolismo , Sustitución de Aminoácidos/genética , Animales , Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/química , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/inmunología , Grupo Borrelia Burgdorferi/genética , Grupo Borrelia Burgdorferi/inmunología , Cisteína/genética , Inmunoglobulina G/sangre , Vacunas contra Enfermedad de Lyme/química , Vacunas contra Enfermedad de Lyme/genética , Vacunas contra Enfermedad de Lyme/inmunología , Ratones , Ratones Endogámicos C3H , Mutagénesis Sitio-Dirigida , Factores de Virulencia/química , Factores de Virulencia/genética , Factores de Virulencia/inmunología
20.
Hum Vaccin ; 3(6): 281-9, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17921702

RESUMEN

Lyme disease is the most common vector-borne disease in North America and Europe and, if untreated, has significant arthritic, cardiac, dermatological and neurological sequelae. There is no currently available human Lyme disease vaccine. Outer surface protein C, because of its antigenicity, protective ability, and expression characteristics has emerged as a promising second generation vaccine candidate; however, significant sequence heterogeneity has impeded its development. Analyses of OspC sequences have revealed the existence of stable phylogenetic clusters or types, and that the type-defining sequence variation occurs within defined domains of the protein. Recent data indicating that immunodominant, and potentially protective OspC epitopes are located in these hypervariable regions has allowed development of a tetravalent, epitope-based, chimeric vaccine. In this report, we have extended that previously described tetravalent construct to include four additional OspC types. We demonstrate that the construct is highly immunogenic, and elicits type-specific antibodies that recognize each of the eight incorporated OspC type-specific epitopes. Antibody raised to the octavalent construct readily binds to the surface of strains expressing each component OspC type, indicating that the incorporated epitopes are presented on the surface of intact cells. In addition, the construct elicits antibody isotypes associated with complement-dependent bactericidal activity. These results represent an important step forward in the design of a broadly protective polyvalent OspC-based Lyme disease vaccine.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Borrelia burgdorferi/inmunología , Vacunas contra Enfermedad de Lyme/química , Vacunas contra Enfermedad de Lyme/inmunología , Enfermedad de Lyme/prevención & control , Secuencia de Aminoácidos , Animales , Anticuerpos Antibacterianos , Especificidad de Anticuerpos , Antígenos Bacterianos/química , Proteínas de la Membrana Bacteriana Externa/química , Vacunas Bacterianas/química , Borrelia burgdorferi/aislamiento & purificación , Quimera/inmunología , Epítopos/inmunología , Enfermedad de Lyme/inmunología , Vacunas contra Enfermedad de Lyme/genética , Ratones , Ratones Endogámicos C3H , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA