Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 198(10): 3989-3998, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28320831

RESUMO

Staphylococcal enterotoxin B (SEB) is a bacterial superantigen that binds the receptors in the APC/T cell synapse and causes increased proliferation of T cells and a cytokine storm syndrome in vivo. Exposure to the toxin can be lethal and cause significant pathology in humans. The lack of effective therapies for SEB exposure remains an area of concern, particularly in scenarios of acute mass casualties. We hypothesized that blockade of the T cell costimulatory signal by the CTLA4-Ig synthetic protein (abatacept) could prevent SEB-dependent pathology. In this article, we demonstrate mice treated with a single dose of abatacept 8 h post SEB exposure had reduced pathology compared with control SEB-exposed mice. SEB-exposed mice showed significant reductions in body weight between days 4 and 9, whereas mice exposed to SEB and also treated with abatacept showed no weight loss for the duration of the study, suggesting therapeutic mitigation of SEB-induced morbidity. Histopathology and magnetic resonance imaging demonstrated that SEB mediated lung damage and edema, which were absent after treatment with abatacept. Analysis of plasma and lung tissues from SEB-exposed mice treated with abatacept demonstrated significantly lower levels of IL-6 and IFN-γ (p < 0.0001), which is likely to have resulted in less pathology. In addition, exposure of human and mouse PBMCs to SEB in vitro showed a significant reduction in levels of IL-2 (p < 0.0001) after treatment with abatacept, indicating that T cell proliferation is the main target for intervention. Our findings demonstrate that abatacept is a robust and potentially credible drug to prevent toxic effects from SEB exposure.


Assuntos
Abatacepte/uso terapêutico , Células Apresentadoras de Antígenos/imunologia , Enterotoxinas/toxicidade , Infecções Estafilocócicas/tratamento farmacológico , Linfócitos T/imunologia , Abatacepte/administração & dosagem , Abatacepte/farmacologia , Animais , Peso Corporal , Citocinas/imunologia , Enterotoxinas/farmacologia , Humanos , Interferon gama/sangue , Interleucina-2/biossíntese , Interleucina-2/imunologia , Interleucina-6/sangue , Pulmão/patologia , Camundongos , Monócitos/imunologia , Infecções Estafilocócicas/imunologia
2.
Crit Rev Biotechnol ; 37(5): 553-565, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27373876

RESUMO

The purpose of this review is to survey current, emerging and predicted future biotechnologies which are impacting, or are likely to impact in the future on the life sciences, with a projection for the coming 20 years. This review is intended to discuss current and future technical strategies, and to explore areas of potential growth during the foreseeable future. Information technology approaches have been employed to gather and collate data. Twelve broad categories of biotechnology have been identified which are currently impacting the life sciences and will continue to do so. In some cases, technology areas are being pushed forward by the requirement to deal with contemporary questions such as the need to address the emergence of anti-microbial resistance. In other cases, the biotechnology application is made feasible by advances in allied fields in biophysics (e.g. biosensing) and biochemistry (e.g. bio-imaging). In all cases, the biotechnologies are underpinned by the rapidly advancing fields of information systems, electronic communications and the World Wide Web together with developments in computing power and the capacity to handle extensive biological data. A rationale and narrative is given for the identification of each technology as a growth area. These technologies have been categorized by major applications, and are discussed further. This review highlights: Biotechnology has far-reaching applications which impinge on every aspect of human existence. The applications of biotechnology are currently wide ranging and will become even more diverse in the future. Access to supercomputing facilities and the ability to manipulate large, complex biological datasets, will significantly enhance knowledge and biotechnological development.


Assuntos
Biotecnologia
3.
PLoS Pathog ; 10(5): e1004085, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24788397

RESUMO

Bacillus anthracis produces a binary toxin composed of protective antigen (PA) and one of two subunits, lethal factor (LF) or edema factor (EF). Most studies have concentrated on induction of toxin-specific antibodies as the correlate of protective immunity, in contrast to which understanding of cellular immunity to these toxins and its impact on infection is limited. We characterized CD4+ T cell immunity to LF in a panel of humanized HLA-DR and DQ transgenic mice and in naturally exposed patients. As the variation in antigen presentation governed by HLA polymorphism has a major impact on protective immunity to specific epitopes, we examined relative binding affinities of LF peptides to purified HLA class II molecules, identifying those regions likely to be of broad applicability to human immune studies through their ability to bind multiple alleles. Transgenics differing only in their expression of human HLA class II alleles showed a marked hierarchy of immunity to LF. Immunogenicity in HLA transgenics was primarily restricted to epitopes from domains II and IV of LF and promiscuous, dominant epitopes, common to all HLA types, were identified in domain II. The relevance of this model was further demonstrated by the fact that a number of the immunodominant epitopes identified in mice were recognized by T cells from humans previously infected with cutaneous anthrax and from vaccinated individuals. The ability of the identified epitopes to confer protective immunity was demonstrated by lethal anthrax challenge of HLA transgenic mice immunized with a peptide subunit vaccine comprising the immunodominant epitopes that we identified.


Assuntos
Vacinas contra Antraz , Antraz/prevenção & controle , Antígenos de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Linfócitos T CD4-Positivos/imunologia , Antígenos HLA/genética , Imunidade Celular/genética , Polimorfismo Genético , Dermatopatias Bacterianas/prevenção & controle , Adulto , Sequência de Aminoácidos , Animais , Antraz/imunologia , Vacinas contra Antraz/química , Vacinas contra Antraz/uso terapêutico , Antígenos de Bactérias/química , Toxinas Bacterianas/química , Mapeamento de Epitopos , Antígenos HLA/imunologia , Humanos , Epitopos Imunodominantes/química , Epitopos Imunodominantes/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Moleculares , Terapia de Alvo Molecular , Dermatopatias Bacterianas/imunologia , Adulto Jovem
4.
Anal Bioanal Chem ; 407(24): 7349-57, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26187320

RESUMO

A unified approach to affinity screening for Fab and Fc interactions of an antibody for its antigen and FcγR receptor has been developed. An antigen array is used for the Fab affinity and cross-reactivity screening and protein A/G proxy is the FcγR receptor. The affinities are derived using a simple 1:1 binding model with a consistent error analysis. The association and dissociation kinetics are measured over optimised times for accurate determination. The Fab/Fc affinities are derived for ten antibodies: mAb-actin (mouse), pAb-BSA (sheep), pAb-collagen V (rabbit), pAb-CRP (goat), mAb-F1 (mouse), mAbs (mouse) 7.3, 12.3, 29.3, 36.3 and 46.3 raised against LcrV in Yersinia pestis. The rate of the dissociation of antigen-antibody complexes relates directly to their immunological function as does the Fc-FcγR complex and a new half-life plot has been defined with a Fab/Fc half-life range of 17-470 min. The upper half-life value points to surface avidity. Two antibodies that are protective as an immunotherapy define a Fab half-life >250 min and an Fc half-life >50 min as characteristics of ideal interactions which can form the basis of an antibody screen for immunotherapy.


Assuntos
Anticorpos Monoclonais/imunologia , Afinidade de Anticorpos , Fragmentos Fab das Imunoglobulinas/imunologia , Fragmentos Fc das Imunoglobulinas/imunologia , Anticorpos Monoclonais/farmacocinética , Reações Cruzadas , Meia-Vida
5.
Infect Immun ; 82(8): 3206-13, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24866807

RESUMO

Burkholderia pseudomallei, the etiologic agent of melioidosis, is a CDC tier 1 select agent that causes severe disease in both humans and animals. Diagnosis and treatment of melioidosis can be challenging, and in the absence of optimal chemotherapeutic intervention, acute disease is frequently fatal. Melioidosis is an emerging infectious disease for which there are currently no licensed vaccines. Due to the potential malicious use of B. pseudomallei as well as its impact on public health in regions where the disease is endemic, there is significant interest in developing vaccines for immunization against this disease. In the present study, type A O-polysaccharide (OPS) and manno-heptose capsular polysaccharide (CPS) antigens were isolated from nonpathogenic, select-agent-excluded strains of B. pseudomallei and covalently linked to carrier proteins. By using these conjugates (OPS2B1 and CPS2B1, respectively), it was shown that although high-titer IgG responses against the OPS or CPS component of the glycoconjugates could be raised in BALB/c mice, only those animals immunized with CPS2B1 were protected against intraperitoneal challenge with B. pseudomallei. Extending upon these studies, it was also demonstrated that when the mice were immunized with a combination of CPS2B1 and recombinant B. pseudomallei LolC, rather than with CPS2B1 or LolC individually, they exhibited higher survival rates when challenged with a lethal dose of B. pseudomallei. Collectively, these results suggest that CPS-based glycoconjugates are promising candidates for the development of subunit vaccines for immunization against melioidosis.


Assuntos
Antígenos de Bactérias/imunologia , Cápsulas Bacterianas/imunologia , Vacinas Bacterianas/imunologia , Burkholderia pseudomallei/imunologia , Melioidose/prevenção & controle , Animais , Anticorpos Antibacterianos/sangue , Vacinas Bacterianas/administração & dosagem , Feminino , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sobrevida , Vacinas Conjugadas/administração & dosagem , Vacinas Conjugadas/imunologia
6.
Front Immunol ; 15: 1277526, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38605961

RESUMO

This study evaluated a depot-formulated cytokine-based adjuvant to improve the efficacy of the recombinant F1V (rF1V) plague vaccine and examined the protective response following aerosol challenge in a murine model. The results of this study showed that co-formulation of the Alhydrogel-adsorbed rF1V plague fusion vaccine with the depot-formulated cytokines recombinant human interleukin 2 (rhuIL-2) and/or recombinant murine granulocyte macrophage colony-stimulating factor (rmGM-CSF) significantly enhances immunogenicity and significant protection at lower antigen doses against a lethal aerosol challenge. These results provide additional support for the co-application of the depot-formulated IL-2 and/or GM-CSF cytokines to enhance vaccine efficacy.


Assuntos
Vacina contra a Peste , Yersinia pestis , Humanos , Animais , Camundongos , Citocinas , Antígenos de Bactérias , Vacinas Sintéticas , Aerossóis
7.
Proteomics ; 13(22): 3298-308, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24115457

RESUMO

The use of bacterial systems for recombinant protein production has advantages of simplicity, time and cost over competing systems. However, widely used bacterial expression systems (e.g. Escherichia coli, Pseudomonas fluorescens) are not able to secrete soluble proteins directly into the culture medium. This limits yields and increases downstream processing time and costs. In contrast, Bacillus spp. secrete native enzymes directly into the culture medium at grams-per-litre quantities, although the yields of some recombinant proteins are severely limited. We have engineered the Bacillus subtilis genome to generate novel strains with precise deletions in the genes encoding ten extracytoplasmic proteases that affect recombinant protein secretion, which lack chromosomal antibiotic resistance genes. The deletion sites and presence of single nucleotide polymorphisms were confirmed by sequencing. The strains are stable and were used in industrial-scale fermenters for the production of the Bacillus anthracis vaccine protein, protective antigen, the productivity of which is extremely low in the unmodified strain. We also show that the deletion of so-called quality control proteases appears to influence cell-wall synthesis, resulting in the induction of the cell-wall stress regulon that encodes another quality control protease.


Assuntos
Bacillus subtilis/metabolismo , Proteínas de Bactérias/análise , Engenharia Genética/métodos , Proteoma/análise , Proteínas Recombinantes/metabolismo , Antígenos de Bactérias/análise , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Bacillus subtilis/genética , Proteínas de Bactérias/química , Toxinas Bacterianas/análise , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Espaço Extracelular/química , Espaço Extracelular/metabolismo , Deleção de Genes , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Proteoma/química , Proteínas Recombinantes/análise , Proteínas Recombinantes/genética
8.
Front Microbiol ; 14: 1247041, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38029221

RESUMO

Mice were immunized with a combination of self-amplifying (sa) RNA constructs for the F1 and V antigens of Yersinia pestis at a dose level of 1 µg or 5 µg or with the respective protein sub-units as a reference vaccine. The immunization of outbred OF1 mice on day 0 and day 28 with the lowest dose used (1 µg) of each of the saRNA constructs in lipid nanoparticles protected 5/7 mice against subsequent sub-cutaneous challenge on day 56 with 180 cfu (2.8 MLD) of a 2021 clinical isolate of Y. pestis termed 10-21/S whilst 5/7 mice were protected against 1800cfu (28MLD) of the same bacteria on day 56. By comparison, only 1/8 or 1/7 negative control mice immunized with 10 µg of irrelevant haemagglutin RNA in lipid nanoparticles (LNP) survived the challenge with 2.8 MLD or 28 MLD Y. pestis 10-21/S, respectively. BALB/c mice were also immunized with the same saRNA constructs and responded with the secretion of specific IgG to F1 and V, neutralizing antibodies for the V antigen and developed a recall response to both F1 and V. These data represent the first report of an RNA vaccine approach using self-amplifying technology and encoding both of the essential virulence antigens, providing efficacy against Y. pestis. This saRNA vaccine for plague has the potential for further development, particularly since its amplifying nature can induce immunity with less boosting. It is also amenable to rapid manufacture with simpler downstream processing than protein sub-units, enabling rapid deployment and surge manufacture during disease outbreaks.

9.
Viruses ; 15(6)2023 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-37376607

RESUMO

Venezuelan equine encephalitis virus (VEEV) is a disease typically confined to South and Central America, whereby human disease is characterised by a transient systemic infection and occasionally severe encephalitis, which is associated with lethality. Using an established mouse model of VEEV infection, the encephalitic aspects of the disease were analysed to identify biomarkers associated with inflammation. Sequential sampling of lethally challenged mice (infected subcutaneously) confirmed a rapid onset systemic infection with subsequent spread to the brain within 24 h of the challenge. Changes in inflammatory biomarkers (TNF-α, CCL-2, and CCL-5) and CD45+ cell counts were found to correlate strongly to pathology (R>0.9) and present previously unproven biomarkers for disease severity in the model, more so than viral titre. The greatest level of pathology was observed within the olfactory bulb and midbrain/thalamus. The virus was distributed throughout the brain/encephalon, often in areas not associated with pathology. The principal component analysis identified five principal factors across two independent experiments, with the first two describing almost half of the data: (1) confirmation of a systemic Th1-biased inflammatory response to VEEV infection, and (2) a clear correlation between specific inflammation of the brain and clinical signs of disease. Targeting strongly associated biomarkers of deleterious inflammation may ameliorate or even eliminate the encephalitic syndrome of this disease.


Assuntos
Vírus da Encefalite Equina Venezuelana , Encefalomielite Equina Venezuelana , Humanos , Cavalos , Camundongos , Animais , Fator de Necrose Tumoral alfa , Vírus da Encefalite Equina Venezuelana/fisiologia , Encéfalo , Inflamação/patologia , Quimiocinas , Leucócitos
10.
Adv Appl Microbiol ; 81: 209-41, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22958531

RESUMO

Plague has been a scourge of humanity, responsible for the deaths of millions. The etiological agent, Yersinia pestis, has evolved relatively recently from an enteropathogen, Yersinia pseudotuberculosis. The evolution of the plague pathogen has involved a complex series of genetic acquisitions, deletions, and rearrangements in its transition from an enteric niche to becoming a systemic, flea-vectored pathogen. With the advent of modern molecular biology techniques, we are starting to understand how the organism adapts to the diverse niches it encounters and how to combat the threat it poses.


Assuntos
Peste , Yersinia pestis , Animais , Proteínas de Bactérias/genética , Evolução Biológica , Humanos , Sifonápteros , Virulência , Yersinia pseudotuberculosis
11.
J Immunol ; 184(7): 3814-21, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20208010

RESUMO

There has been a long history of defining T cell epitopes to track viral immunity and to design rational vaccines, yet few data of this type exist for bacterial infections. Bacillus anthracis, the causative agent of anthrax, is both an endemic pathogen in many regions and a potential biological warfare threat. T cell immunity in naturally infected anthrax patients has not previously been characterized, which is surprising given concern about the ability of anthrax toxins to subvert or ablate adaptive immunity. We investigated CD4 T cell responses in patients from the Kayseri region of Turkey who were previously infected with cutaneous anthrax. Responses to B. anthracis protective Ag and lethal factor (LF) were investigated at the protein, domain, and epitope level. Several years after antibiotic-treated anthrax infection, strong T cell memory was detectable, with no evidence of the expected impairment in specific immunity. Although serological responses to existing anthrax vaccines focus primarily on protective Ag, the major target of T cell immunity in infected individuals and anthrax-vaccinated donors was LF, notably domain IV. Some of these anthrax epitopes showed broad binding to several HLA class alleles, but others were more constrained in their HLA binding patterns. Of specific CD4 T cell epitopes targeted within LF domain IV, one is preferentially seen in the context of bacterial infection, as opposed to vaccination, suggesting that studies of this type will be important in understanding how the human immune system confronts serious bacterial infection.


Assuntos
Antraz/imunologia , Antígenos de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Linfócitos T CD4-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Vacinas contra Antraz/imunologia , Bacillus anthracis/imunologia , Ensaio de Imunoadsorção Enzimática , Humanos , Memória Imunológica
12.
Front Immunol ; 13: 1017385, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36389793

RESUMO

The development of a safe and effective vaccine against Yersinia pestis, the causative organism for plague disease, remains an important global health priority. Studies have demonstrated effective immune-based protection against plague challenge that is induced by plague antigen subunit vaccination in an aqueous alhydrogel formulation; however, whether these candidate vaccines in this formulation and presentation, induce long-lasting immunological memory in the form of durable cellular and antibody recall responses has not been fully demonstrated. In this study, we analyzed germinal center T follicular helper and germinal center B cell responses following F1V and F1 + V plague subunit immunization of mice with vaccines formulated in various adjuvants. Our data demonstrate that recombinant plague protein immunization formulated with IL-2/GM-CSF cytokines bound to alhydrogel adjuvant drive an increase in the magnitude of the germinal center T follicular helper and germinal center B cell responses following primary immunization, compared to vaccines formulated with Alhydrogel adjuvant alone. In contrast, plague protein subunit immunization combined with CpG ODN bound to alhydrogel increased the magnitude and duration of the germinal center Tfh and B cell responses following booster immunization. Importantly, enhanced germinal center Tfh and B cell responses correlated with long-lasting and high F1V-specific antibody titers and more robust antibody recall responses to F1V re-exposure. These findings indicate that vaccine formulations that drive enhancement of the germinal center Tfh and B cell responses are critical for inducing durable plague-specific humoral immunity.


Assuntos
Hidróxido de Alumínio , Centro Germinativo , Camundongos , Animais , Células T Auxiliares Foliculares , Vacinação , Adjuvantes Imunológicos/farmacologia , Vacinas de Subunidades Antigênicas , Imunização Secundária , Vacinas Sintéticas
13.
Viruses ; 14(8)2022 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-36016391

RESUMO

A transduced mouse model of SARS-CoV-2 infection was established using Balb/c mice. This was achieved through the adenovirus-vectored delivery of the hACE2 gene, to render the mice transiently susceptible to the virus. The model was characterised in terms of the dissemination of hACE2 receptor expression, the dissemination of three SARS-CoV-2 virus variants in vivo up to 10 days following challenge, the resulting histopathology and the clinical signs induced in the mice. In transduced mice, the infection was short-term, with a rapid loss in body weight starting at day 2 with maximum weight loss at day 4, followed by subsequent recovery until day 10. The induced expression of the hACE2 receptor was evident in the lungs, but, upon challenge, the SARS-CoV-2 virus disseminated beyond the lungs to spleen, liver and kidney, peaking at day 2 post infection. However, by day 10 post infection, the virus was undetectable. The lung histopathology was characterised by bronchial and alveolar inflammation, which was still present at day 10 post infection. Transduced mice had differential responses to viral variants ranking CVR-Glasgow 1 > Victoria-1 > England-2 isolates in terms of body weight loss. The transduced mouse model provides a consistent and manipulatable model of SARS-CoV-2 infection to screen viral variants for their relative virulence and possible interventions.


Assuntos
COVID-19 , SARS-CoV-2 , Enzima de Conversão de Angiotensina 2/genética , Animais , Modelos Animais de Doenças , Pulmão , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Peptidil Dipeptidase A/metabolismo , SARS-CoV-2/genética
14.
Vaccine ; 40(42): 6163-6178, 2022 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-36153153

RESUMO

We undertook a Phase 4 clinical trial to assess the effect of time interval between booster doses on serological responses to AVP. The primary objective was to evaluate responses to a single booster dose in two groups of healthy adults who had previously received a complete 4-dose primary course. Group A had received doses on schedule while Group B had not had one for ≥2 years. Secondary objectives were to evaluate the safety and tolerability of AVP booster doses, and to gain information on correlates of protection to aid future anthrax vaccine development. Blood samples were taken on Day 1 before dosing, and on Days 8, 15, 29 and 120, to measure Toxin Neutralisation Assay (TNA) NF50 values and concentrations of IgG antibodies against Protective Antigen (PA), Lethal Factor (LF) and Edema Factor (EF) by ELISA. For each serological parameter, fold changes from baseline following the trial AVP dose were greater in Group B than Group A at every time-point studied. Peak responses correlated positively with time since last AVP dose (highest values being observed after intervals of ≥10 years), and negatively with number of previous doses (highest values occurring in individuals who had received a primary course only). In 2017, having reviewed these results, the Joint Committee on Vaccination and Immunisation (JCVI) updated UK anthrax vaccination guidelines, extending the interval between routine AVP boosters from one to 10 years. Booster doses of AVP induce significant IgG responses against the three anthrax toxin components, particularly PA and LF. Similarly high responses were observed in TNA, a recognised surrogate for anthrax vaccine efficacy. Analysis of the 596 TNA results showed that anti-PA and anti-LF IgG make substantial independent contributions to neutralisation of anthrax lethal toxin. AVP may therefore have advantages over anthrax vaccines that depend on generating immunity to PA alone.


Assuntos
Vacinas contra Antraz , Antraz , Bacillus anthracis , Adulto , Antraz/prevenção & controle , Anticorpos Antibacterianos , Antígenos de Bactérias , Humanos , Imunoglobulina G , Vacinação/métodos
15.
Vaccines (Basel) ; 10(10)2022 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-36298436

RESUMO

The causative agent of anthrax, Bacillus anthracis, evades the host immune response and establishes infection through the production of binary exotoxins composed of Protective Antigen (PA) and one of two subunits, lethal factor (LF) or edema factor (EF). The majority of vaccination strategies have focused upon the antibody response to the PA subunit. We have used a panel of humanised HLA class II transgenic mouse strains to define HLA-DR-restricted and HLA-DQ-restricted CD4+ T cell responses to the immunodominant epitopes of PA. This was correlated with the binding affinities of epitopes to HLA class II molecules, as well as the responses of two human cohorts: individuals vaccinated with the Anthrax Vaccine Precipitated (AVP) vaccine (which contains PA and trace amounts of LF), and patients recovering from cutaneous anthrax infections. The infected and vaccinated cohorts expressing different HLA types were found to make CD4+ T cell responses to multiple and diverse epitopes of PA. The effects of HLA polymorphism were explored using transgenic mouse lines, which demonstrated differential susceptibility, indicating that HLA-DR1 and HLA-DQ8 alleles conferred protective immunity relative to HLA-DR15, HLA-DR4 and HLA-DQ6. The HLA transgenics enabled a reductionist approach, allowing us to better define CD4+ T cell epitopes. Appreciating the effects of HLA polymorphism on the variability of responses to natural infection and vaccination is vital in planning protective strategies against anthrax.

16.
Immunother Adv ; 1(1): ltab020, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35919741

RESUMO

Two monoclonal antibodies directed to the V antigen of Yersinia pestis have been tested for protective efficacy in a murine model of bubonic plague. Mice were infected with a current clinical isolate from Madagascar, designated Y. pestis 10-21/S. Mab7.3, delivered to mice intra-periteoneally at either 24 h prior to, or 24 h post-infection, was fully protective, building on many studies which have demonstrated the protective efficacy of this Mab against a number of different clinical isolates of Y. pestis. Mab 29.3, delivered intra-peritoneally at either -24 h or +24 h, protected 4/5 mice in either condition; this has demonstrated the protective efficacy of this Mab in vivo for the first time. These results add to the cumulative data about Mab7.3, which is currently being humanized and highlight its potential as a human immunotherapeutic for plague, which is an enduring endemic disease in Madagascar and other regions of Africa, Asia, and South America.

17.
Infect Immun ; 78(10): 4356-62, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20660611

RESUMO

Yersinia pestis is the causative agent of plague, a rapidly fatal infectious disease that has not been eradicated worldwide. The capsular Caf1 protein of Y. pestis is a protective antigen under development as a recombinant vaccine. However, little is known about the specificity of human T-cell responses for Caf1. We characterized CD4 T-cell epitopes of Caf1 in "humanized" HLA-DR1 transgenic mice lacking endogenous major histocompatibility complex class II molecules. Mice were immunized with Caf1 or each of a complete set of overlapping synthetic peptides, and CD4 T-cell immunity was measured with respect to proliferative and gamma interferon T-cell responses and recognition by a panel of T-cell hybridomas, as well as direct determination of binding affinities of Caf1 peptides to purified HLA-DR molecules. Although a number of DR1-restricted epitopes were identified following Caf1 immunization, the response was biased toward a single immunodominant epitope near the C terminus of Caf1. In addition, potential promiscuous epitopes, including the immunodominant epitope, were identified by their ability to bind multiple common HLA alleles, with implications for the generation of multivalent vaccines against plague for use in humans.


Assuntos
Proteínas de Bactérias/imunologia , Linfócitos T CD4-Positivos/imunologia , Antígenos HLA/metabolismo , Antígeno HLA-DR1/imunologia , Yersinia pestis/imunologia , Animais , Epitopos , Antígenos HLA/genética , Humanos , Camundongos , Camundongos Transgênicos , Ligação Proteica
18.
Nature ; 430(6998): 417, 2004 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-15272490

RESUMO

Mecsas and colleagues suggest that a deficiency in the chemokine receptor CCR5 in humans is unlikely to confer protection against plague, based on their study of Yersinia pestis infection in Ccr5-deficient mice. They were testing the hypothesis that a mutation in the CCR5 gene, frequently found in Caucasians, may have been selected for in the past because it provided protection against (bubonic) plague; the mutation, called CCR5Delta32, is characterized by a 32-base-pair deletion. We have also tested this hypothesis by using Y. pestis infection in mice and, in addition, we have done phagocytosis experiments with macrophages from wild-type and Ccr5-deficient mice. Although, like Mecsas et al., we did not see any difference in the survival of the two groups of mice, we did find that there was a significantly reduced uptake of Y. pestis by Ccr5-deficient macrophages in vitro. Our results indicate that the role of Ccr5 in Y. pestis infection may therefore be more complex than previously thought.


Assuntos
Evolução Molecular , Peste/genética , Peste/metabolismo , Receptores CCR5/genética , Receptores CCR5/metabolismo , Animais , Humanos , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Modelos Biológicos , Fagocitose , Peste/microbiologia , Receptores CCR5/deficiência , Deleção de Sequência/genética , Taxa de Sobrevida , Virulência , População Branca/genética , Yersinia pestis/patogenicidade , Yersinia pestis/fisiologia
19.
J Control Release ; 324: 644-656, 2020 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-32512014

RESUMO

The field of brain drug delivery faces many challenges that hinder development and testing of novel therapies for clinically important central nervous system disorders. Chief among them is how to deliver large biologics across the highly restrictive blood-brain barrier. Non-ionic surfactant vesicles (NISV) have long been used as a drug delivery platform for cutaneous applications and have benefits over comparable liposomes in terms of greater stability, lower cost and suitability for large scale production. Here we describe a glucosamine-coated NISV, for blood-brain barrier GLUT1 targeting, capable of traversing the barrier and delivering active antibody to cells within the brain. In vitro, we show glucosamine vesicle transcytosis across the blood-brain barrier with intact cargo, which is partially dynamin-dependent, but is clathrin-independent and does not associate with sorting endosome marker EEA1. Uptake of vesicles into astrocytes follows a more classical pathway involving dynamin, clathrin, sorting endosomes and Golgi trafficking where the cargo is released intracellularly. In vivo, glucosamine-coated vesicles are superior to uncoated or transferrin-coated vesicles for delivering cargo to the mouse brain. Finally, mice infected with Venezuelan equine encephalitis virus (VEEV) were successfully treated with anti-VEEV monoclonal antibody Hu1A3B-7 delivered in glucosamine-coated vesicles and had improved survival and reduced brain tissue virus levels. An additional benefit was that the treatment also reduced viral load in peripheral tissues. The data generated highlights the huge potential of glucosamine-decorated NISV as a drug delivery platform with wider potential applications.


Assuntos
Barreira Hematoencefálica , Vírus da Encefalite Equina Venezuelana , Animais , Glucosamina , Cavalos , Camundongos , Tensoativos , Transcitose
20.
Artigo em Inglês | MEDLINE | ID: mdl-32373552

RESUMO

As the ongoing outbreak in the Democratic Republic of Congo illustrates, Ebola virus disease continues to pose a significant risk to humankind and this necessitates the continued development of therapeutic options. One option that warrants evaluation is that of defective genomes; these can potentially parasitize resources from the wild-type virus and may even be packaged for repeated co-infection cycles. Deletion and copy-back defective genomes have been identified and reported in the literature. As a crude, mixed preparation these were found to have limiting effects on cytopathology. Here we have used synthetic virology to clone and manufacture two deletion defective genomes. These genomes were tested with Ebola virus using in vitro cell culture and shown to inhibit viral replication; however, and against expectations, the defective genomes were not released in biologically significant numbers. We propose that EBOV might have yet unknown mechanisms to prevent parasitisation by defective interfering particles beyond the known mechanism that prevents sequential infection of the same cell. Understanding this mechanism would be necessary in any development of a defective interfering particle-based therapy.


Assuntos
Ebolavirus , Doença pelo Vírus Ebola , Congo , Ebolavirus/genética , Genoma Viral , Humanos , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA