Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 213
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Immunol Rev ; 293(1): 270-282, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31709558

RESUMO

After many decades of research, an effective vaccine for malaria is still not available. Most research efforts have focused on identifying a key target antigen and then using powerful adjuvants to generate specific antibodies that can block parasites from entering host cells (hepatocytes, red blood cells). However, the inability to generate sufficiently potent antibody responses has led to significant disappointment with current vaccine programs. An additional challenge for sub-unit vaccines is that key vaccine antigens are highly polymorphic. These challenges have spurred radically different approaches to malaria vaccine development. Many of these involve the use of "whole parasites"-either extracted from mosquitoes or cultured. With these, every parasite molecule for that particular strain is included in the vaccine. This strategy is showing great promise following several clinical trials with irradiated sporozoites. However, a whole-parasite approach to a blood stage vaccine has not advanced as quickly. This article outlines the history, the different approaches that are being taken and the challenges associated with whole parasite blood stage vaccines and discusses recent exciting developments as these vaccines now move into the clinic.


Assuntos
Interações Hospedeiro-Parasita/imunologia , Vacinas Antimaláricas/imunologia , Malária/imunologia , Malária/parasitologia , Plasmodium/crescimento & desenvolvimento , Plasmodium/imunologia , Animais , Animais Geneticamente Modificados , Variação Biológica da População/imunologia , Humanos , Imunidade , Estágios do Ciclo de Vida , Malária/prevenção & controle , Pesquisa Translacional Biomédica , Vacinas de Subunidades Antigênicas/imunologia
2.
Small ; 19(8): e2205819, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36564365

RESUMO

Immunogenic carrier proteins such as the non-toxic diphtheria toxin variant, cross-reacting material 197 (CRM197), are widely used in subunit vaccine formulations to boost immunogenicity of chemically conjugated antigens. Conjugate vaccines are inherently expensive due to laborious manufacturing steps. Here, this work develops a particulate vaccine platform based on using engineered Escherichia coli to assemble CRM197-antigen fusion proteins into discrete submicron-sized particles. This approach enables precise loading of diverse antigens and epitopes enhancing their immunogenicity. A cost-effective, high-yield, and scalable biomanufacturing process is developed. Purified particulate CRM197-antigen vaccines are ambient-temperature stable. CRM197 particles incorporating pathogen-specific antigens or epitopes from SARS-CoV-2, Streptococcus pyogenes (group A), and Mycobacterium tuberculosis induced cell-mediated and humoral immune responses mediating protective immunity in respective animal models of infection. The CRM197 particle vaccine platform is versatile, enabling co-delivery of selected antigens/epitopes together with immunogenic CRM197 as discrete stable particles avoiding laborious manufacture of soluble CRM197 and antigen followed by chemical conjugation.


Assuntos
COVID-19 , Animais , SARS-CoV-2 , Proteínas de Bactérias/química , Vacinas Sintéticas , Vacinas Conjugadas , Antígenos , Epitopos
3.
J Infect Dis ; 219(1): 110-120, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30534974

RESUMO

Background: In pregnancy, Plasmodium falciparum parasites express the surface antigen VAR2CSA, which mediates adherence of red blood cells to chondroitin sulfate A (CSA) in the placenta. VAR2CSA antibodies are generally acquired during infection in pregnancy and are associated with protection from placental malaria. We observed previously that men and children in Colombia also had antibodies to VAR2CSA, but the origin of these antibodies was unknown. Here, we tested whether infection with Plasmodium vivax is an alternative mechanism of acquisition of VAR2CSA antibodies. Methods: We analyzed sera from nonpregnant Colombians and Brazilians exposed to P. vivax and monoclonal antibodies raised against P. vivax Duffy binding protein (PvDBP). Cross-reactivity to VAR2CSA was characterized by enzyme-linked immunosorbent assay, immunofluorescence assay, and flow cytometry, and antibodies were tested for inhibition of parasite binding to CSA. Results: Over 50% of individuals had antibodies that recognized VAR2CSA. Affinity-purified PvDBP human antibodies and a PvDBP monoclonal antibody recognized VAR2CSA, showing that PvDBP can give rise to cross-reactive antibodies. Importantly, the monoclonal antibody inhibited parasite binding to CSA, which is the primary in vitro correlate of protection from placental malaria. Conclusions: These data suggest that PvDBP induces antibodies that functionally recognize VAR2CSA, revealing a novel mechanism of cross-species immune recognition to falciparum malaria.


Assuntos
Antígenos de Protozoários/imunologia , Antígenos de Superfície/imunologia , Reações Cruzadas/imunologia , Malária Falciparum/imunologia , Malária Vivax/imunologia , Plasmodium falciparum/imunologia , Plasmodium vivax/imunologia , Proteínas de Protozoários/imunologia , Receptores de Superfície Celular/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Antiprotozoários/sangue , Criança , Sulfatos de Condroitina , Colômbia , Eritrócitos/parasitologia , Eutérios/imunologia , Feminino , Humanos , Imunidade , Gravidez
4.
Infect Immun ; 87(1)2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30323025

RESUMO

Naturally acquired immunity to malaria is robust and protective against all strains of the same species of Plasmodium This develops as a result of repeated natural infection, taking several years to develop. Evidence suggests that apoptosis of immune lymphocytes due to uncontrolled parasite growth contributes to the slow acquisition of immunity. To hasten and augment the development of natural immunity, we studied controlled infection immunization (CII) using low-dose exposure to different parasite species (Plasmodium chabaudi, P. yoelii, or P. falciparum) in two rodent systems (BALB/c and C57BL/6 mice) and in human volunteers, with drug therapy commencing at the time of initiation of infection. CIIs with infected erythrocytes and in conjunction with doxycycline or azithromycin, which are delayed death drugs targeting the parasite's apicoplast, allowed extended exposure to parasites at low levels. In turn, this induced strong protection against homologous challenge in all immunized mice. We show that P. chabaudi/P. yoelii infection initiated at the commencement of doxycycline therapy leads to cellular or antibody-mediated protective immune responses in mice, with a broad Th1 cytokine response providing the best correlate of protection against homologous and heterologous species of PlasmodiumP. falciparum CII with doxycycline was additionally tested in a pilot clinical study (n = 4) and was found to be well tolerated and immunogenic, with immunological studies primarily detecting increased cell-associated immune responses. Furthermore, we report that a single dose of the longer-acting drug, azithromycin, given to mice (n = 5) as a single subcutaneous treatment at the initiation of infection controlled P. yoelii infection and protected all mice against subsequent challenge.


Assuntos
Antimaláricos/administração & dosagem , Malária/tratamento farmacológico , Malária/imunologia , Plasmodium chabaudi/imunologia , Plasmodium falciparum/imunologia , Plasmodium yoelii/imunologia , Vacinação/métodos , Imunidade Adaptativa , Animais , Azitromicina/administração & dosagem , Citocinas/metabolismo , Modelos Animais de Doenças , Doxiciclina/administração & dosagem , Feminino , Humanos , Malária/prevenção & controle , Malária Falciparum , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Plasmodium chabaudi/crescimento & desenvolvimento , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium yoelii/crescimento & desenvolvimento , Células Th1/imunologia , Adulto Jovem
5.
Immunity ; 33(4): 555-66, 2010 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-21029965

RESUMO

The concept of a malaria vaccine has sparked great interest for decades; however, the challenge is proving to be a difficult one. Immune dysregulation by Plasmodium and the ability of the parasite to mutate critical epitopes in surface antigens have proved to be strong defense weapons. This has led to reconsideration of polyvalent and whole parasite strategies and ways to enhance cellular immunity to malaria that may be more likely to target conserved antigens and an expanded repertoire of antigens. These and other concepts will be discussed in this review.


Assuntos
Vacinas Antimaláricas/imunologia , Animais , Antígenos de Protozoários/imunologia , Linfócitos T CD8-Positivos/imunologia , Desenho de Fármacos , Humanos , Evasão da Resposta Imune , Plasmodium/imunologia , Vacinas de DNA/imunologia , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/imunologia
6.
J Immunol ; 199(8): 2794-2802, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28904125

RESUMO

Peptides offer enormous promise as vaccines to prevent and protect against many infectious and noninfectious diseases. However, to date, limited vaccine efficacy has been reported and none have been licensed for human use. Innovative ways to enhance their immunogenicity are being tested, but rational sequence modification as a means to improve immune responsiveness has been neglected. Our objective was to establish a two-step generic protocol to modify defined amino acids of a helical peptide epitope to create a superior immunogen. Peptide variants of p145, a conserved helical peptide epitope from the M protein of Streptococcus pyogenes, were designed by exchanging one amino acid at a time, without altering their α-helical structure, which is required for correct antigenicity. The immunogenicities of new peptides were assessed in outbred mice. Vaccine efficacy was assessed in a skin challenge and invasive disease model. Out of 86 variants of p145, seven amino acid substitutions were selected and made the basis of the design for 18 new peptides. Of these, 13 were more immunogenic than p145; 7 induced Abs with significantly higher affinity for p145 than Abs induced by p145 itself; and 1 peptide induced more than 10,000-fold greater protection following challenge than the parent peptide. This peptide also only required a single immunization (compared with three immunizations with the parent peptide) to induce complete protection against invasive streptococcal disease. This study defines a strategy to rationally improve the immunogenicity of peptides and will have broad applicability to the development of vaccines for infectious and noninfectious diseases.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Transporte/metabolismo , Fragmentos de Peptídeos/metabolismo , Infecções Estreptocócicas/imunologia , Vacinas Estreptocócicas/imunologia , Streptococcus pyogenes/imunologia , Animais , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Humanos , Imunidade Humoral , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Mutação/genética , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Engenharia de Proteínas , Infecções Estreptocócicas/prevenção & controle , Vacinas de Subunidades Antigênicas
7.
Infect Immun ; 86(1)2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28923897

RESUMO

Controlled human malaria infection (CHMI) entails deliberate infection with malaria parasites either by mosquito bite or by direct injection of sporozoites or parasitized erythrocytes. When required, the resulting blood-stage infection is curtailed by the administration of antimalarial drugs. Inducing a malaria infection via inoculation with infected blood was first used as a treatment (malariotherapy) for neurosyphilis in Europe and the United States in the early 1900s. More recently, CHMI has been applied to the fields of malaria vaccine and drug development, where it is used to evaluate products in well-controlled early-phase proof-of-concept clinical studies, thus facilitating progression of only the most promising candidates for further evaluation in areas where malaria is endemic. Controlled infections have also been used to immunize against malaria infection. Historically, CHMI studies have been restricted by the need for access to insectaries housing infected mosquitoes or suitable malaria-infected individuals. Evaluation of vaccine and drug candidates has been constrained in these studies by the availability of a limited number of Plasmodium falciparum isolates. Recent advances have included cryopreservation of sporozoites, the manufacture of well-characterized and genetically distinct cultured malaria cell banks for blood-stage infection, and the availability of Plasmodium vivax-specific reagents. These advances will help to accelerate malaria vaccine and drug development by making the reagents for CHMI more widely accessible and also enabling a more rigorous evaluation with multiple parasite strains and species. Here we discuss the different applications of CHMI, recent advances in the use of CHMI, and ongoing challenges for consideration.


Assuntos
Malária/imunologia , Malária/parasitologia , Plasmodium/imunologia , Animais , Antimaláricos/uso terapêutico , Culicidae/parasitologia , Eritrócitos/parasitologia , Humanos , Malária/tratamento farmacológico , Vacinas Antimaláricas/imunologia , Plasmodium/efeitos dos fármacos , Esporozoítos/efeitos dos fármacos , Esporozoítos/imunologia
8.
BMC Med ; 16(1): 184, 2018 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-30293531

RESUMO

BACKGROUND: The continuing morbidity and mortality associated with infection with malaria parasites highlights the urgent need for a vaccine. The efficacy of sub-unit vaccines tested in clinical trials in malaria-endemic areas has thus far been disappointing, sparking renewed interest in the whole parasite vaccine approach. We previously showed that a chemically attenuated whole parasite asexual blood-stage vaccine induced CD4+ T cell-dependent protection against challenge with homologous and heterologous parasites in rodent models of malaria. METHODS: In this current study, we evaluated the immunogenicity and safety of chemically attenuated asexual blood-stage Plasmodium falciparum (Pf) parasites in eight malaria-naïve human volunteers. Study participants received a single dose of 3 × 107 Pf pRBC that had been treated in vitro with the cyclopropylpyrolloindole analogue, tafuramycin-A. RESULTS: We demonstrate that Pf asexual blood-stage parasites that are completely attenuated are immunogenic, safe and well tolerated in malaria-naïve volunteers. Following vaccination with a single dose, species and strain transcending Plasmodium-specific T cell responses were induced in recipients. This included induction of Plasmodium-specific lymphoproliferative responses, T cells secreting the parasiticidal cytokines, IFN-γ and TNF, and CD3+CD45RO+ memory T cells. Pf-specific IgG was not detected. CONCLUSIONS: This is the first clinical study evaluating a whole parasite blood-stage malaria vaccine. Following administration of a single dose of completely attenuated Pf asexual blood-stage parasites, Plasmodium-specific T cell responses were induced while Pf-specific antibodies were not detected. These results support further evaluation of this chemically attenuated vaccine in humans. TRIAL REGISTRATION: Trial registration: ACTRN12614000228684 . Registered 4 March 2014.


Assuntos
Vacinas Antimaláricas/imunologia , Malária Falciparum/prevenção & controle , Vacinas Atenuadas/imunologia , Adulto , Animais , Anticorpos Antiprotozoários/imunologia , Humanos , Imunidade Celular/imunologia , Masculino , Projetos Piloto , Plasmodium falciparum/imunologia , Linfócitos T/imunologia , Vacinação/métodos
9.
PLoS Pathog ; 12(12): e1006122, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28027314

RESUMO

The immunobiology underlying the slow acquisition of skin immunity to group A streptococci (GAS), is not understood, but attributed to specific virulence factors impeding innate immunity and significant antigenic diversity of the type-specific M-protein, hindering acquired immunity. We used a number of epidemiologically distinct GAS strains to model the development of acquired immunity. We show that infection leads to antibody responses to the serotype-specific determinants on the M-protein and profound protective immunity; however, memory B cells do not develop and immunity is rapidly lost. Furthermore, antibodies do not develop to a conserved M-protein epitope that is able to induce immunity following vaccination. However, if re-infected with the same strain within three weeks, enduring immunity and memory B-cells (MBCs) to type-specific epitopes do develop. Such MBCs can adoptively transfer protection to naïve recipients. Thus, highly protective M-protein-specific MBCs may never develop following a single episode of pyoderma, contributing to the slow acquisition of immunity and to streptococcal endemicity in at-risk populations.


Assuntos
Memória Imunológica/imunologia , Pioderma/imunologia , Pioderma/microbiologia , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/microbiologia , Animais , Ensaio de Imunoadsorção Enzimática , Camundongos , Streptococcus pyogenes
10.
J Immunol ; 197(12): 4518-4526, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27913644

RESUMO

The development of vaccines to protect against parasites is difficult, in large part due to complex host-parasite interactions that have evolved over millennia. Parasitic factors such as antigenic variation and host factors such as age, transmission intensity, and genetic influences are all thought to contribute to the limited efficacy of parasite vaccines. A developing theme in field studies investigating antiparasitic immunity is the emergence, establishment, and maintenance of immunoregulatory networks that shape the immune responses to new infections, as well as vaccines, thereby influencing disease outcome. In this review, we will examine why parasite vaccine candidates perform poorly in target populations and, in particular, the role of immunoregulatory networks in influencing antimalarial immunity and vaccine efficacy. We will focus our discussion on malaria, the most important parasitic disease of humans, but also highlight the broader impact of immunoregulatory networks on vaccine efficacy.


Assuntos
Imunidade , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Malária/imunologia , Animais , Variação Antigênica , Antígenos de Protozoários/imunologia , Interações Hospedeiro-Parasita , Humanos , Imunomodulação , Malária/prevenção & controle , Vacinação em Massa , Plasmodium falciparum/imunologia
11.
J Immunol ; 196(8): 3364-74, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26969753

RESUMO

Cluster of virulence responder/sensor (CovR/S) mutant group A streptococci (GAS) are serious human pathogens of multiple M protein strains that upregulate expression of virulence factors, including the IL-8 proteaseStreptococcus pyogenescell envelope proteinase (SpyCEP), thus blunting neutrophil-mediated killing and enabling ingress of bacteria from a superficial wound to deep tissue. We previously showed that a combination vaccine incorporating J8-DT (conserved peptide vaccine from the M protein) and a recombinant SpyCEP fragment protects against CovR/S mutants. To enhance the vaccine's safety profile, we identified a minimal epitope (S2) that was the target for anti-SpyCEP Abs that could protect IL-8 from SpyCEP-mediated proteolysis. Abs from healthy humans and from mice experimentally infected with GAS also recognized S2, albeit at low titers. Native SpyCEP may be poorly immunogenic (cryptic or subdominant), and it would be to the organism's advantage if the host did not induce a strong Ab response against it. However, S2 conjugated to diphtheria toxoid is highly immunogenic and induces Abs that recognize and neutralize SpyCEP. Hence, we describe a two-component peptide vaccine that induces Abs (anti-S2) that protect IL-8 from proteolysis and other Abs (anti-J8) that cause strain-independent killing in the presence of neutrophils. We show that either component alone is ineffectual in preventing skin infection and bacteremia due to CovR/S mutants but that the combination induces complete protection. This protection correlated with a significant influx of neutrophils to the infection site. The data strongly suggest that the lack of natural immunity to hypervirulent GAS strains in humans could be rectified by this combination vaccine.


Assuntos
Antígenos de Bactérias/imunologia , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas de Transporte/imunologia , Peptídeo Hidrolases/imunologia , Infecções Estreptocócicas/imunologia , Vacinas Estreptocócicas/imunologia , Streptococcus pyogenes/imunologia , Animais , Anticorpos Antibacterianos/imunologia , Bacteriemia/imunologia , Bacteriemia/microbiologia , Bacteriemia/prevenção & controle , Proteínas de Bactérias/imunologia , Toxoide Diftérico/imunologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/imunologia , Peptídeo Hidrolases/biossíntese , Peptídeo Hidrolases/genética , Pele/microbiologia , Dermatopatias Bacterianas/imunologia , Dermatopatias Bacterianas/microbiologia , Dermatopatias Bacterianas/prevenção & controle , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/prevenção & controle , Streptococcus pyogenes/patogenicidade , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/imunologia , Fatores de Virulência/biossíntese , Fatores de Virulência/genética , Fatores de Virulência/imunologia
12.
Infect Immun ; 85(7)2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28438976

RESUMO

Malaria vaccine development has been dominated by the subunit approach; however, many subunit vaccine candidates have had limited efficacy in settings of malaria endemicity. As our search for an efficacious malaria vaccine continues, the development of a whole-organism vaccine is now receiving much scrutiny. One strategy currently being explored in the development of a whole-organism vaccine involves chemical attenuation of the malaria parasite. In vivo and in vitro chemical attenuation of both liver-stage and blood-stage Plasmodium parasites has been investigated. Here, we discuss both approaches of chemical attenuation in the development of a whole-organism vaccine against malaria.


Assuntos
Antimaláricos/metabolismo , Vacinas Antimaláricas/imunologia , Plasmodium/efeitos dos fármacos , Plasmodium/imunologia , Animais , Vacinas Atenuadas/imunologia
13.
Infect Immun ; 85(6)2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28320838

RESUMO

Plasmodium vivax malaria remains a major public health problem. The requirements for acquisition of protective immunity to the species are not clear. Dendritic cells (DC) are essential for immune cell priming but also perform immune regulatory functions, along with regulatory T cells (Treg). An important function of DC involves activation of the kynurenine pathway via indoleamine 2,3-dioxygenase (IDO). Using a controlled human experimental infection study with blood-stage P. vivax, we characterized plasmacytoid DC (pDC) and myeloid DC (mDC) subset maturation, CD4+ CD25+ CD127lo Treg activation, and IDO activity. Blood samples were collected from six healthy adults preinoculation, at peak parasitemia (day 14; ∼31,400 parasites/ml), and 24 and 48 h after antimalarial treatment. CD1c+ and CD141+ mDC and pDC numbers markedly declined at peak parasitemia, while CD16+ mDC numbers appeared less affected. HLA-DR expression was selectively reduced on CD1c+ mDC, increased on CD16+ mDC, and was unaltered on pDC. Plasma IFN-γ increased significantly and was correlated with an increased kynurenine/tryptophan (KT) ratio, a measure of IDO activity. At peak parasitemia, Treg presented an activated CD4+ CD25+ CD127lo CD45RA- phenotype and upregulated TNFR2 expression. In a mixed-effects model, the KT ratio was positively associated with an increase in activated Treg. Our data demonstrate that a primary P. vivax infection exerts immune modulatory effects by impairing HLA-DR expression on CD1c+ mDC while activating CD16+ mDC. Induction of the kynurenine pathway and increased Treg activation, together with skewed mDC maturation, suggest P. vivax promotes an immunosuppressive environment, likely impairing the development of a protective host immune response.


Assuntos
Células Dendríticas/imunologia , Antígenos HLA-DR/imunologia , Cinurenina/metabolismo , Ativação Linfocitária , Malária Vivax/imunologia , Linfócitos T Reguladores/imunologia , Adulto , Biomarcadores/sangue , Feminino , Voluntários Saudáveis , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Masculino , Plasmodium vivax , Triptofano/metabolismo , Regulação para Cima , Adulto Jovem
14.
Chembiochem ; 18(6): 545-553, 2017 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-28075053

RESUMO

Four group A streptococcal glycolipopeptide vaccine candidates with different lipidic adjuvanting moieties were prepared and characterized. The immunogenicity of the compounds was evaluated by macrophage and dendritic cell uptake studies and by in vivo quantification of systemic IgG antibody by ELISA. Three of the candidates showed significant induction of the IgG response.


Assuntos
Adjuvantes Imunológicos/síntese química , Imunoglobulina G/sangue , Lipídeos/imunologia , Vacinas Estreptocócicas/síntese química , Vacinas Estreptocócicas/imunologia , Adjuvantes Imunológicos/química , Animais , Ensaio de Imunoadsorção Enzimática , Lipídeos/síntese química , Lipídeos/química , Camundongos , Tamanho da Partícula , Peptídeos/síntese química , Peptídeos/química , Peptídeos/imunologia , Vacinas Estreptocócicas/química
15.
J Immunol ; 194(12): 5915-25, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25980008

RESUMO

Infections caused by Streptococcus pyogenes (group A Streptococcus [GAS]) are highly prevalent in the tropics, in developing countries, and in the Indigenous populations of developed countries. These infections and their sequelae are responsible for almost 500,000 lives lost prematurely each year. A synthetic peptide vaccine (J8-DT) from the conserved region of the M protein has shown efficacy against disease that follows i.p. inoculation of bacteria. By developing a murine model for infection that closely mimics human skin infection, we show that the vaccine can protect against pyoderma and subsequent bacteremia caused by multiple GAS strains, including strains endemic in Aboriginal communities in the Northern Territory of Australia. However, the vaccine was ineffective against a hypervirulent cluster of virulence responder/sensor mutant GAS strain; this correlated with the strain's ability to degrade CXC chemokines, thereby preventing neutrophil chemotaxis. By combining J8-DT with an inactive form of the streptococcal CXC protease, S. pyogenes cell envelope proteinase, we developed a combination vaccine that is highly effective in blocking CXC chemokine degradation and permits opsonic Abs to kill the bacteria. Mice receiving the combination vaccine were strongly protected against pyoderma and bacteremia, as evidenced by a 100-1000-fold reduction in bacterial burden following challenge. To our knowledge, a vaccine requiring Abs to target two independent virulence factors of an organism is unique.


Assuntos
Bacteriemia , Pioderma/prevenção & controle , Vacinas Estreptocócicas/imunologia , Streptococcus/imunologia , Vacinas Sintéticas/imunologia , Animais , Anticorpos Antibacterianos/imunologia , Antígenos de Bactérias/química , Antígenos de Bactérias/imunologia , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas de Transporte/química , Proteínas de Transporte/imunologia , Modelos Animais de Doenças , Feminino , Imunidade Inata , Memória Imunológica , Macrófagos/imunologia , Camundongos , Neutrófilos/imunologia , Peptídeos/imunologia , Pioderma/patologia , Proteínas Recombinantes/imunologia , Infecções Cutâneas Estafilocócicas/patologia , Infecções Cutâneas Estafilocócicas/prevenção & controle , Vacinas Estreptocócicas/genética , Vacinas Sintéticas/genética
16.
Infect Immun ; 84(8): 2274-2288, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27245410

RESUMO

The development of a vaccine is essential for the elimination of malaria. However, despite many years of effort, a successful vaccine has not been achieved. Most subunit vaccine candidates tested in clinical trials have provided limited efficacy, and thus attenuated whole-parasite vaccines are now receiving close scrutiny. Here, we test chemically attenuated Plasmodium yoelii 17X and demonstrate significant protection following homologous and heterologous blood-stage challenge. Protection against blood-stage infection persisted for at least 9 months. Activation of both CD4(+) and CD8(+) T cells was shown after vaccination; however, in vivo studies demonstrated a pivotal role for both CD4(+) T cells and B cells since the absence of either cell type led to loss of vaccine-induced protection. In spite of significant activation of circulating CD8(+) T cells, liver-stage immunity was not evident. Neither did vaccine-induced CD8(+) T cells contribute to blood-stage protection; rather, these cells contributed to pathogenesis, since all vaccinated mice depleted of both CD4(+) and CD8(+) T cells survived a challenge infection. This study provides critical insight into whole-parasite vaccine-induced immunity and strong support for testing whole-parasite vaccines in humans.


Assuntos
Vacinas Antimaláricas/imunologia , Malária/imunologia , Plasmodium yoelii/imunologia , Vacinas Atenuadas/imunologia , Animais , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/imunologia , Reações Cruzadas/imunologia , Feminino , Imunização , Memória Imunológica , Estágios do Ciclo de Vida , Ativação Linfocitária/imunologia , Malária/prevenção & controle , Camundongos , Plasmodium yoelii/crescimento & desenvolvimento , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo
17.
Infect Immun ; 84(5): 1403-1412, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26902728

RESUMO

Dendritic cells (DCs) are sentinels of the immune system that uniquely prime naive cells and initiate adaptive immune responses. CD1c (BDCA-1) myeloid DCs (CD1c(+) mDCs) highly express HLA-DR, have a broad Toll-like receptor (TLR) repertoire, and secrete immune modulatory cytokines. To better understand immune responses to malaria, CD1c(+) mDC maturation and cytokine production were examined in healthy volunteers before and after experimental intravenous Plasmodium falciparum infection with 150- or 1,800-parasite-infected red blood cells (pRBCs). After either dose, CD1c(+) mDCs significantly reduced HLA-DR expression in prepatent infections. Circulating CD1c(+) mDCs did not upregulate HLA-DR after pRBC or TLR ligand stimulation and exhibited reduced CD86 expression. At peak parasitemia, CD1c(+) mDCs produced significantly more tumor necrosis factor (TNF), whereas interleukin-12 (IL-12) production was unchanged. Interestingly, only the 1,800-pRBC dose caused a reduction in the circulating CD1c(+) mDC count with evidence of apoptosis. The 1,800-pRBC dose produced no change in T cell IFN-γ or IL-2 production at peak parasitemia or at 3 weeks posttreatment. Overall, CD1c(+) mDCs are compromised by P. falciparum exposure, with impaired HLA-DR and CD86 expression, and have an increased capacity for TNF but not IL-12 production. A first prepatent P. falciparum infection is sufficient to modulate CD1c(+) mDC responsiveness, likely contributing to hampered effector T cell cytokine responses and assisting parasite immune evasion.


Assuntos
Antígenos CD1/análise , Antígeno B7-2/análise , Células Dendríticas/química , Células Dendríticas/imunologia , Glicoproteínas/análise , Antígenos HLA-DR/análise , Malária Falciparum/patologia , Fator de Necrose Tumoral alfa/metabolismo , Adulto , Estudos de Coortes , Feminino , Voluntários Saudáveis , Humanos , Masculino , Plasmodium falciparum/imunologia , Adulto Jovem
18.
Infect Immun ; 84(9): 2689-96, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27382019

RESUMO

Plasmodium falciparum is the most virulent human malaria parasite because of its ability to cytoadhere in the microvasculature. Nonhuman primate studies demonstrated relationships among knob expression, cytoadherence, and infectivity. This has not been examined in humans. Cultured clinical-grade P. falciparum parasites (NF54, 7G8, and 3D7B) and ex vivo-derived cell banks were characterized. Knob and knob-associated histidine-rich protein expression, CD36 adhesion, and antibody recognition of parasitized erythrocytes (PEs) were evaluated. Parasites from the cell banks were administered to malaria-naive human volunteers to explore infectivity. For the NF54 and 3D7B cell banks, blood was collected from the study participants for in vitro characterization. All parasites were infective in vivo However, infectivity of NF54 was dramatically reduced. In vitro characterization revealed that unlike other cell bank parasites, NF54 PEs lacked knobs and did not cytoadhere. Recognition of NF54 PEs by immune sera was observed, suggesting P. falciparum erythrocyte membrane protein 1 expression. Subsequent recovery of knob expression and CD36-mediated adhesion were observed in PEs derived from participants infected with NF54. Knobless cell bank parasites have a dramatic reduction in infectivity and the ability to adhere to CD36. Subsequent infection of malaria-naive volunteers restored knob expression and CD36-mediated cytoadherence, thereby showing that the human environment can modulate virulence.


Assuntos
Adesão Celular/fisiologia , Malária Falciparum/parasitologia , Parasitos/metabolismo , Peptídeos/metabolismo , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/metabolismo , Adolescente , Adulto , Animais , Membrana Eritrocítica/parasitologia , Eritrócitos/parasitologia , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem
19.
Malar J ; 15: 3, 2016 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-26729268

RESUMO

BACKGROUND: The spectrum of techniques to detect malaria parasites in whole blood is limited to measuring parasites in circulation. One approach that is currently used to enumerate total parasite bio-burden involves the use of bio-luminescent parasites. As an alternative approach, this study describes the use of a commercial ELISA human parasite lactate dehydrogenase (pLDH) detection kit to estimate total parasite bio-burden in murine malaria models. METHODS: The cross reactivity of pLDH in a commercial human malaria pLDH diagnostic kit was established in different components of blood for different murine malaria models. The use of pLDH as a measure of parasite bio-burden was evaluated by examining pLDH in relation to peripheral blood parasitaemia as determined by microscopy and calculating total parasite bio-burden using a bio-luminescent Plasmodium berghei ANKA luciferase parasite. RESULTS: The pLDH antigen was detected in all four murine Plasmodium species and in all components of Plasmodium-infected blood. A significant correlation (r = 0.6922, P value <0.0001) was observed between total parasite bio-burden, measured as log average radiance, and concentration of pLDH units. CONCLUSIONS: This high throughput assay is a suitable measure of total parasite bio-burden in murine malaria infections. Unlike existing methods, it permits the estimation of both circulating and sequestered parasites, allowing a more accurate assessment of parasite bio-burden.


Assuntos
L-Lactato Desidrogenase/sangue , Malária/sangue , Malária/diagnóstico , Plasmodium berghei/enzimologia , Animais , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Parasitemia/sangue , Parasitemia/diagnóstico , Proteínas de Protozoários/sangue
20.
Bioorg Med Chem ; 24(14): 3095-101, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27246859

RESUMO

Infection with Group A Streptococcus (GAS) can result in a range of different illnesses, some of which are fatal. Currently, our efforts to develop a vaccine against GAS focuses on the lipid core peptide (LCP) system, a subunit vaccine containing a lipoamino acid (LAA) moiety which allows the stimulation of systemic antibody activity. In the present study, a peptide (J14) representing the B-cell epitope from the GAS M protein was incorporated alongside a universal T-helper epitope (P25) in four LCP constructs of different spatial orientation or LAA lengths. Through structure-activity studies, it was discovered that while the alteration of the LCP orientation had a weaker effect on immunostimulation, increasing the LAA side chain length within the construct increased antibody responses in murine models. Furthermore, the mice immunised with the lead LCP construct were also able to maintain antibody activity throughout the course of five months. These findings highlight the importance of LAA moieties in the development of intranasal peptide vaccines and confirmed that its side chain length has an effect on the immunogenicity of the structure.


Assuntos
Vacinas Bacterianas/química , Vacinas Bacterianas/farmacologia , Lipídeos/química , Peptídeos/química , Streptococcus pyogenes/imunologia , Sequência de Aminoácidos , Animais , Ensaio de Imunoadsorção Enzimática , Epitopos/química , Camundongos , Microscopia Eletrônica de Transmissão , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA