Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Annu Rev Immunol ; 32: 157-87, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24655294

RESUMO

Malaria is a mosquito-borne disease caused by parasites of the obligate intracellular Apicomplexa phylum the most deadly of which, Plasmodium falciparum, prevails in Africa. Malaria imposes a huge health burden on the world's most vulnerable populations, claiming the lives of nearly one million children and pregnant women each year. Although there is keen interest in eradicating malaria, we do not yet have the necessary tools to meet this challenge, including an effective malaria vaccine and adequate vector control strategies. Here we review what is known about the mechanisms at play in immune resistance to malaria in both the human and mosquito hosts at each step in the parasite's complex life cycle with a view toward developing the tools that will contribute to the prevention of disease and death and, ultimately, to the goal of malaria eradication. In so doing, we hope to inspire immunologists to participate in defeating this devastating disease.


Assuntos
Culicidae/imunologia , Interações Hospedeiro-Patógeno/imunologia , Malária/imunologia , Plasmodium/imunologia , Animais , Culicidae/parasitologia , Humanos , Estágios do Ciclo de Vida , Malária/parasitologia , Malária/prevenção & controle , Plasmodium/crescimento & desenvolvimento , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/imunologia
2.
J Infect Dis ; 222(10): 1681-1691, 2020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-32687161

RESUMO

BACKGROUND: A previous RTS,S/AS01B vaccine challenge trial demonstrated that a 3-dose (0-1-7-month) regimen with a fractional third dose can produce high vaccine efficacy (VE) in adults challenged 3 weeks after vaccination. This study explored the VE of different delayed fractional dose regimens of adult and pediatric RTS,S/AS01 formulations. METHODS: A total of 130 participants were randomized into 5 groups. Four groups received 3 doses of RTS,S/AS01B or RTS,S/AS01E on a 0-1-7-month schedule, with the final 1 or 2 doses being fractional (one-fifth dose volume). One group received 1 full (month 0) and 1 fractional (month 7) dose of RTS,S/AS01E. Immunized and unvaccinated control participants underwent Plasmodium falciparum-infected mosquito challenge (controlled human malaria infection) 3 months after immunization, a timing chosen to potentially discriminate VEs between groups. RESULTS: The VE of 3-dose formulations ranged from 55% (95% confidence interval, 27%-72%) to 76% (48%-89%). Groups administered equivalent formulations of RTS,S/AS01E and RTS,S/AS01B demonstrated comparable VE. The 2-dose group demonstrated lower VE (29% [95% confidence interval, 6%-46%]). All regimens were well tolerated and immunogenic, with trends toward higher anti-circumsporozoite antibody titers in participants protected against infection. CONCLUSIONS: RTS,S/AS01E can provide VE comparable to an equivalent RTS,S/AS01B regimen in adults, suggesting a universal formulation may be considered. Results also suggest that the 2-dose regimen is inferior to the 3-dose regimens evaluated. CLINICAL TRIAL REGISTRATION: NCT03162614.


Assuntos
Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/imunologia , Malária/imunologia , Malária/prevenção & controle , Adolescente , Adulto , Feminino , Humanos , Esquemas de Imunização , Controle de Infecções , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Masculino , Pessoa de Meia-Idade , Plasmodium falciparum/imunologia , Vacinação , Adulto Jovem
3.
Malar J ; 19(1): 10, 2020 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-31910830

RESUMO

BACKGROUND: Immunization with attenuated malaria sporozoites protects humans from experimental malaria challenge by mosquito bite. Protection in humans is strongly correlated with the production of T cells targeting a heterogeneous population of pre-erythrocyte antigen proteoforms, including liver stage antigens. Currently, few T cell epitopes derived from Plasmodium falciparum, the major aetiologic agent of malaria in humans are known. METHODS: In this study both in vitro and in vivo malaria liver stage models were used to sequence host and pathogen proteoforms. Proteoforms from these diverse models were subjected to mild acid elution (of soluble forms), multi-dimensional fractionation, tandem mass spectrometry, and top-down bioinformatics analysis to identify proteoforms in their intact state. RESULTS: These results identify a group of host and malaria liver stage proteoforms that meet a 5% false discovery rate threshold. CONCLUSIONS: This work provides proof-of-concept for the validity of this mass spectrometry/bioinformatic approach for future studies seeking to reveal malaria liver stage antigens towards vaccine development.


Assuntos
Fígado/parasitologia , Malária Falciparum/imunologia , Plasmodium falciparum/imunologia , Animais , Antígenos de Protozoários/imunologia , Modelos Animais de Doenças , Epitopos de Linfócito T , Feminino , Hepatócitos , Imunidade Celular , Vacinas Antimaláricas/imunologia , Malária Falciparum/prevenção & controle , Espectrometria de Massas , Camundongos , Proteômica , Albumina Sérica Humana
4.
PLoS Pathog ; 13(8): e1006487, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28771605

RESUMO

Studies have demonstrated cross-reactivity of anti-dengue virus (DENV) antibodies in human sera against Zika virus (ZIKV), promoting increased ZIKV infection in vitro. However, the correlation between in vitro and in vivo findings is not well characterized. Thus, we evaluated the impact of heterotypic flavivirus immunity on ZIKV titers in biofluids of rhesus macaques. Animals previously infected (≥420 days) with DENV2, DENV4, or yellow fever virus were compared to flavivirus-naïve animals following infection with a Brazilian ZIKV strain. Sera from DENV-immune macaques demonstrated cross-reactivity with ZIKV by antibody-binding and neutralization assays prior to ZIKV infection, and promoted increased ZIKV infection in cell culture assays. Despite these findings, no significant differences between flavivirus-naïve and immune animals were observed in viral titers, neutralizing antibody levels, or immune cell kinetics following ZIKV infection. These results indicate that prior infection with heterologous flaviviruses neither conferred protection nor increased observed ZIKV titers in this non-human primate ZIKV infection model.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Infecções por Flavivirus/imunologia , Infecção por Zika virus/imunologia , Animais , Reações Cruzadas/imunologia , Ensaio de Imunoadsorção Enzimática , Flavivirus/imunologia , Infecções por Flavivirus/patologia , Macaca mulatta , Reação em Cadeia da Polimerase , Zika virus/imunologia , Infecção por Zika virus/patologia
5.
Proc Natl Acad Sci U S A ; 112(5): 1273-80, 2015 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-25552553

RESUMO

The malaria parasite, Plasmodium, must survive and develop in the mosquito vector to be successfully transmitted to a new host. The Plasmodium falciparum Pfs47 gene is critical for malaria transmission. Parasites that express Pfs47 (NF54 WT) evade mosquito immunity and survive, whereas Pfs47 knockouts (KO) are efficiently eliminated by the complement-like system. Two alternative approaches were used to investigate the mechanism of action of Pfs47 on immune evasion. First, we examined whether Pfs47 affected signal transduction pathways mediating mosquito immune responses, and show that the Jun-N-terminal kinase (JNK) pathway is a key mediator of Anopheles gambiae antiplasmodial responses to P. falciparum infection and that Pfs47 disrupts JNK signaling. Second, we used microarrays to compare the global transcriptional responses of A. gambiae midguts to infection with WT and KO parasites. The presence of Pfs47 results in broad and profound changes in gene expression in response to infection that are already evident 12 h postfeeding, but become most prominent at 26 h postfeeding, the time when ookinetes invade the mosquito midgut. Silencing of 15 differentially expressed candidate genes identified caspase-S2 as a key effector of Plasmodium elimination in parasites lacking Pfs47. We provide experimental evidence that JNK pathway regulates activation of caspases in Plasmodium-invaded midgut cells, and that caspase activation is required to trigger midgut epithelial nitration. Pfs47 alters the cell death pathway of invaded midgut cells by disrupting JNK signaling and prevents the activation of several caspases, resulting in an ineffective nitration response that makes the parasite undetectable by the mosquito complement-like system.


Assuntos
Anopheles/imunologia , Apoptose/fisiologia , MAP Quinase Quinase 4/metabolismo , Plasmodium falciparum/fisiologia , Animais , Anopheles/parasitologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiologia , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Proteínas de Protozoários/fisiologia
6.
PLoS Pathog ; 9(9): e1003622, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24039583

RESUMO

The innate immune system of Anopheles gambiae mosquitoes limits Plasmodium infection through multiple molecular mechanisms. For example, midgut invasion by the parasite triggers an epithelial nitration response that promotes activation of the complement-like system. We found that suppression of the JNK pathway, by silencing either Hep, JNK, Jun or Fos expression, greatly enhanced Plasmodium infection; while overactivating this cascade, by silencing the suppressor Puckered, had the opposite effect. The JNK pathway limits infection via two coordinated responses. It induces the expression of two enzymes (HPx2 and NOX5) that potentiate midgut epithelial nitration in response to Plasmodium infection and regulates expression of two key hemocyte-derived immune effectors (TEP1 and FBN9). Furthermore, the An. gambiae L3-5 strain that has been genetically selected to be refractory (R) to Plasmodium infection exhibits constitutive overexpression of genes from the JNK pathway, as well as midgut and hemocyte effector genes. Silencing experiments confirmed that this cascade mediates, to a large extent, the drastic parasite elimination phenotype characteristic of this mosquito strain. In sum, these studies revealed the JNK pathway as a key regulator of the ability of An. gambiae mosquitoes to limit Plasmodium infection and identified several effector genes mediating these responses.


Assuntos
Anopheles/imunologia , Proteínas de Insetos/imunologia , MAP Quinase Quinase 4/imunologia , Plasmodium berghei/imunologia , Transdução de Sinais/imunologia , Animais , Anopheles/parasitologia , NADPH Oxidases/imunologia , Proteínas Proto-Oncogênicas c-fos/imunologia , Proteínas Proto-Oncogênicas c-jun/imunologia
7.
J Am Mosq Control Assoc ; 31(2): 149-54, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26181690

RESUMO

Animal models are often used to study hematophagous insect feeding behavior and evaluate products such as topical repellents. However, when these models are used the study animals often experience significant drops in core body temperature because of the effects of anesthesia. This study used a guinea pig model to investigate whether maintaining a normothermic core body temperature during anesthesia influenced the rate of Anopheles stephensi and Phlebotomus papatasi blood feeding. Experiments were conducted with anesthetized animals that had their body temperatures either maintained with a warming device or were allowed to drop naturally. Results showed that when guinea pigs were actively warmed by a heating device, An. stephensi feeding behavior was similar at the beginning and end of anesthesia. However, when a warming device was not used, fewer An. stephensi took a blood meal after the animals' temperatures had dropped. Phlebotomus papatasi were not as sensitive to changes in temperature and feeding rates were similar whether a warming device was used or not. These results are discussed and it is recommended that warming devices are used when conducting feeding experiments with insects sensitive to changes in host body temperature, such as An. stephensi.


Assuntos
Anestesia Geral , Anopheles/fisiologia , Temperatura Corporal , Comportamento Alimentar/fisiologia , Temperatura Alta , Phlebotomus/fisiologia , Animais , Cobaias
8.
PLoS Pathog ; 8(6): e1002737, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22685401

RESUMO

The Anopheles gambiae immune response against Plasmodium falciparum, an etiological agent of human malaria, has been identified as a source of potential anti-Plasmodium genes and mechanisms to be exploited in efforts to control the malaria transmission cycle. One such mechanism is the Imd pathway, a conserved immune signaling pathway that has potent anti-P. falciparum activity. Silencing the expression of caspar, a negative regulator of the Imd pathway, or over-expressing rel2, an Imd pathway-controlled NFkappaB transcription factor, confers a resistant phenotype on A. gambiae mosquitoes that involves an array of immune effector genes. However, unexplored features of this powerful mechanism that may be essential for the implementation of a malaria control strategy still remain. Using RNA interference to singly or dually silence caspar and other components of the Imd pathway, we have identified genes participating in the anti-Plasmodium signaling module regulated by Caspar, each of which represents a potential target to achieve over-activation of the pathway. We also determined that the Imd pathway is most potent against the parasite's ookinete stage, yet also has reasonable activity against early oocysts and lesser activity against late oocysts. We further demonstrated that caspar silencing alone is sufficient to induce a robust anti-P. falciparum response even in the relative absence of resident gut microbiota. Finally, we established the relevance of the Imd pathway components and regulated effectors TEP1, APL1, and LRIM1 in parasite infection intensity-dependent defense, thereby shedding light on the relevance of laboratory versus natural infection intensity models. Our results highlight the physiological considerations that are integral to a thoughtful implementation of Imd pathway manipulation in A. gambiae as part of an effort to limit the malaria transmission cycle, and they reveal a variety of previously unrecognized nuances in the Imd-directed immune response against P. falciparum.


Assuntos
Anopheles/imunologia , Anopheles/parasitologia , Proteínas de Insetos/imunologia , Malária/imunologia , Plasmodium falciparum/imunologia , Animais , Insetos Vetores/imunologia , Malária Falciparum/prevenção & controle , Interferência de RNA , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais
9.
PLoS Pathog ; 6(2): e1000767, 2010 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-20169188

RESUMO

A universal feature of metazoan sexual development is the generation of oocyte P granules that withhold certain mRNA species from translation to provide coding potential for proteins during early post-fertilization development. Stabilisation of translationally quiescent mRNA pools in female Plasmodium gametocytes depends on the RNA helicase DOZI, but the molecular machinery involved in the silencing of transcripts in these protozoans is unknown. Using affinity purification coupled with mass-spectrometric analysis we identify a messenger ribonucleoprotein (mRNP) from Plasmodium berghei gametocytes defined by DOZI and the Sm-like factor CITH (homolog of worm CAR-I and fly Trailer Hitch). This mRNP includes 16 major factors, including proteins with homologies to components of metazoan P granules and archaeal proteins. Containing translationally silent transcripts, this mRNP integrates eIF4E and poly(A)-binding protein but excludes P body RNA degradation factors and translation-initiation promoting eIF4G. Gene deletion mutants of 2 core components of this mRNP (DOZI and CITH) are fertilization-competent, but zygotes fail to develop into ookinetes in a female gametocyte-mutant fashion. Through RNA-immunoprecipitation and global expression profiling of CITH-KO mutants we highlight CITH as a crucial repressor of maternally supplied mRNAs. Our data define Plasmodium P granules as an ancient mRNP whose protein core has remained evolutionarily conserved from single-cell organisms to germ cells of multi-cellular animals and stores translationally silent mRNAs that are critical for early post-fertilization development during the initial stages of mosquito infection. Therefore, translational repression may offer avenues as a target for the generation of transmission blocking strategies and contribute to limiting the spread of malaria.


Assuntos
Regulação da Expressão Gênica/fisiologia , Plasmodium berghei/fisiologia , Proteínas de Protozoários/fisiologia , Interferência de RNA/fisiologia , Animais , Southern Blotting , Western Blotting , Feminino , Citometria de Fluxo , Expressão Gênica , Perfilação da Expressão Gênica , Células Germinativas , Imunoprecipitação , Filogenia , RNA Mensageiro/genética , Ribonucleoproteínas/fisiologia , Desenvolvimento Sexual , Zigoto
10.
PLoS Pathog ; 5(3): e1000335, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19282971

RESUMO

Immune responses mounted by the malaria vector Anopheles gambiae are largely regulated by the Toll and Imd (immune deficiency) pathways via the NF-kappaB transcription factors Rel1 and Rel2, which are controlled by the negative regulators Cactus and Caspar, respectively. Rel1- and Rel2-dependent transcription in A. gambiae has been shown to be particularly critical to the mosquito's ability to manage infection with the rodent malaria parasite Plasmodium berghei. Using RNA interference to deplete the negative regulators of these pathways, we found that Rel2 controls resistance of A. gambiae to the human malaria parasite Plasmodium falciparum, whereas Rel 1 activation reduced infection levels. The universal relevance of this defense system across Anopheles species was established by showing that caspar silencing also prevents the development of P. falciparum in the major malaria vectors of Asia and South America, A. stephensi and A. albimanus, respectively. Parallel studies suggest that while Imd pathway activation is most effective against P. falciparum, the Toll pathway is most efficient against P. berghei, highlighting a significant discrepancy between the human pathogen and its rodent model. High throughput gene expression analyses identified a plethora of genes regulated by the activation of the two Rel factors and revealed that the Toll pathway played a more diverse role in mosquito biology than the Imd pathway, which was more immunity-specific. Further analyses of key anti-Plasmodium factors suggest they may be responsible for the Imd pathway-mediated resistance phenotype. Additionally, we found that the fitness cost caused by Rel2 activation through caspar gene silencing was undetectable in sugar-fed, blood-fed, and P. falciparum-infected female A. gambiae, while activation of the Toll pathway's Rel1 had a major impact. This study describes for the first time a single gene that influences an immune mechanism that is able to abort development of P. falciparum in Anopheline species. Further, this study addresses aspects of the molecular, evolutionary, and physiological consequences of the observed phenotype. These findings have implications for malaria control since broad-spectrum immune activation in diverse anopheline species offers a viable and strategic approach to develop novel malaria control methods worldwide.


Assuntos
Anopheles/genética , Anopheles/imunologia , Proteínas de Insetos/imunologia , Insetos Vetores/genética , Insetos Vetores/imunologia , Plasmodium falciparum/fisiologia , Sequência de Aminoácidos , Animais , Anopheles/parasitologia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo , Insetos Vetores/parasitologia , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Transdução de Sinais/fisiologia , Fatores de Transcrição
11.
Vaccine ; 39(43): 6398-6406, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34593270

RESUMO

BACKGROUND: We previously demonstrated that RTS,S/AS01B and RTS,S/AS01E vaccination regimens including at least one delayed fractional dose can protect against Plasmodium falciparum malaria in a controlled human malaria infection (CHMI) model, and showed inferiority of a two-dose versus three-dose regimen. In this follow-on trial, we evaluated whether fractional booster vaccination extended or induced protection in previously protected (P-Fx) or non-protected (NP-Fx) participants. METHODS: 49 participants (P-Fx: 25; NP-Fx: 24) received a fractional (1/5th dose-volume) RTS,S/AS01E booster 12 months post-primary regimen. They underwent P. falciparum CHMI three weeks later and were then followed for six months for safety and immunogenicity. RESULTS: Overall vaccine efficacy against re-challenge was 53% (95% CI: 37-65%), and similar for P-Fx (52% [95% CI: 28-68%]) and NP-Fx (54% [95% CI: 29-70%]). Efficacy appeared unaffected by primary regimen or previous protection status. Anti-CS (repeat region) antibody geometric mean concentrations (GMCs) increased post-booster vaccination. GMCs were maintained over time in primary three-dose groups but declined in the two-dose group. Protection after re-challenge was associated with higher anti-CS antibody responses. The booster was well-tolerated. CONCLUSIONS: A fractional RTS,S/AS01E booster given one year after completion of a primary two- or three-dose RTS,S/AS01 delayed fractional dose regimen can extend or induce protection against CHMI. CLINICAL TRIAL REGISTRATION: NCT03824236. linked to this article can be found on the Research Data as well as Figshare https://figshare.com/s/ee025150f9d1ac739361.


Assuntos
Vacinas Antimaláricas , Malária Falciparum , Malária , Anticorpos Antiprotozoários , Humanos , Malária Falciparum/prevenção & controle , Plasmodium falciparum , Vacinação
12.
PLoS One ; 16(9): e0256980, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34495988

RESUMO

BACKGROUND: A DNA-prime/human adenovirus serotype 5 (HuAd5) boost vaccine encoding Plasmodium falciparum (Pf) circumsporozoite protein (PfCSP) and Pf apical membrane antigen-1 (PfAMA1), elicited protection in 4/15 (27%) of subjects against controlled human malaria infection (CHMI) that was statistically associated with CD8+ T cell responses. Subjects with high level pre-existing immunity to HuAd5 were not protected, suggesting an adverse effect on vaccine efficacy (VE). We replaced HuAd5 with chimpanzee adenovirus 63 (ChAd63), and repeated the study, assessing both the two-antigen (CSP, AMA1 = CA) vaccine, and a novel three-antigen (CSP, AMA1, ME-TRAP = CAT) vaccine that included a third pre-erythrocytic stage antigen [malaria multiple epitopes (ME) fused to the Pf thrombospondin-related adhesive protein (TRAP)] to potentially enhance protection. METHODOLOGY: This was an open label, randomized Phase 1 trial, assessing safety, tolerability, and VE against CHMI in healthy, malaria naïve adults. Forty subjects (20 each group) were to receive three monthly CA or CAT DNA priming immunizations, followed by corresponding ChAd63 boost four months later. Four weeks after the boost, immunized subjects and 12 infectivity controls underwent CHMI by mosquito bite using the Pf3D7 strain. VE was assessed by determining the differences in time to parasitemia as detected by thick blood smears up to 28-days post CHMI and utilizing the log rank test, and by calculating the risk ratio of each treatment group and subtracting from 1, with significance calculated by the Cochran-Mantel-Haenszel method. RESULTS: In both groups, systemic adverse events (AEs) were significantly higher after the ChAd63 boost than DNA immunizations. Eleven of 12 infectivity controls developed parasitemia (mean 11.7 days). In the CA group, 15 of 16 (93.8%) immunized subjects developed parasitemia (mean 12.0 days). In the CAT group, 11 of 16 (63.8%) immunized subjects developed parasitemia (mean 13.0 days), indicating significant protection by log rank test compared to infectivity controls (p = 0.0406) and the CA group (p = 0.0229). VE (1 minus the risk ratio) in the CAT group was 25% compared to -2% in the CA group. The CA and CAT vaccines induced robust humoral (ELISA antibodies against CSP, AMA1 and TRAP, and IFA responses against sporozoites and Pf3D7 blood stages), and cellular responses (IFN-γ FluoroSpot responses to CSP, AMA1 and TRAP) that were not associated with protection. CONCLUSIONS: This study demonstrated that the ChAd63 CAT vaccine exhibited significant protective efficacy, and confirmed protection was afforded by adding a third antigen (T) to a two-antigen (CA) formulation to achieve increased VE. Although the ChAd63-CAT vaccine was associated with increased frequencies of systemic AEs compared to the CA vaccine and, historically, compared to the HuAd5 vectored malaria vaccine encoding CSP and AMA1, they were transient and associated with increased vector dosing.


Assuntos
Vacinas contra Adenovirus/imunologia , Adenovirus dos Símios/imunologia , Antígenos de Protozoários/imunologia , DNA de Protozoário/imunologia , DNA Recombinante/imunologia , Imunização Secundária/métodos , Vacinas Antimaláricas/imunologia , Malária Falciparum/prevenção & controle , Proteínas de Membrana/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Vacinas de DNA/imunologia , Vacinas contra Adenovirus/administração & dosagem , Vacinas contra Adenovirus/efeitos adversos , Adenovirus dos Símios/genética , Adulto , Antígenos de Protozoários/genética , Linfócitos T CD8-Positivos/imunologia , DNA de Protozoário/genética , Epitopos/genética , Epitopos/imunologia , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/imunologia , Voluntários Saudáveis , Humanos , Imunogenicidade da Vacina/imunologia , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/efeitos adversos , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Masculino , Proteínas de Membrana/genética , Proteínas de Protozoários/genética , Resultado do Tratamento , Vacinas de DNA/administração & dosagem , Vacinas de DNA/efeitos adversos , Adulto Jovem
13.
Curr Mol Med ; 9(2): 116-30, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19275622

RESUMO

Malaria is one of today's most serious diseases with an enormous socioeconomic impact. While anti-malarial drugs have existed for some time and vaccines development may be underway, the most successful malaria eradication programs have thus far relied on attacking the mosquito vector that spreads the disease causing agent Plasmodium. Here we will review past, current and future perspectives of malaria vector control strategies and how these approaches have taken a promising turn thanks recent advances in functional genomics and molecular biology.


Assuntos
Insetos Vetores , Malária , Controle de Mosquitos/métodos , Animais , Animais Geneticamente Modificados , Comportamento Animal/fisiologia , Ritmo Circadiano/fisiologia , Culicidae/anatomia & histologia , Culicidae/fisiologia , Humanos , Luz , Malária/prevenção & controle , Malária/transmissão , Vacinas Antimaláricas/uso terapêutico
14.
PLoS Negl Trop Dis ; 14(4): e0008191, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32267846

RESUMO

Dengue virus (DENV) is transmitted by infectious mosquitoes during blood-feeding via saliva containing biologically-active proteins. Here, we examined the effect of varying DENV infection modality in rhesus macaques in order to improve the DENV nonhuman primate (NHP) challenge model. NHPs were exposed to DENV-1 via subcutaneous or intradermal inoculation of virus only, intradermal inoculation of virus and salivary gland extract, or infectious mosquito feeding. The infectious mosquito feeding group exhibited delayed onset of viremia, greater viral loads, and altered clinical and immune responses compared to other groups. After 15 months, NHPs in the subcutaneous and infectious mosquito feeding groups were re-exposed to either DENV-1 or DENV-2. Viral replication and neutralizing antibody following homologous challenge were suggestive of sterilizing immunity, whereas heterologous challenge resulted in productive, yet reduced, DENV-2 replication and boosted neutralizing antibody. These results show that a more transmission-relevant exposure modality resulted in viral replication closer to that observed in humans.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Dengue/imunologia , Animais , Dengue/virologia , Vírus da Dengue/fisiologia , Modelos Animais de Doenças , Feminino , Cinética , Macaca mulatta/imunologia , Mosquitos Vetores/virologia , RNA Viral/sangue , Glândulas Salivares/virologia , Vacinação , Carga Viral , Viremia/prevenção & controle , Replicação Viral
15.
PLoS One ; 15(6): e0233840, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32555601

RESUMO

BACKGROUND: Immunization with radiation-attenuated sporozoites (RAS) by mosquito bite provides >90% sterile protection against Plasmodium falciparum (Pf) malaria in humans. RAS invade hepatocytes but do not replicate. CD8+ T cells recognizing parasite-derived peptides on the surface of infected hepatocytes are likely the primary protective mechanism. We conducted a randomized clinical trial of RAS immunization to assess safety, to achieve 50% vaccine efficacy (VE) against controlled human malaria infection (CHMI), and to generate reagents from protected and non-protected subjects for future identification of protective immune mechanisms and antigens. METHODS: Two cohorts (Cohort 1 and Cohort 2) of healthy, malaria-naïve, non-pregnant adults age 18-50 received five monthly immunizations with infected (true-immunized, n = 21) or non-infected (mock-immunized, n = 5) mosquito bites and underwent homologous CHMI at 3 weeks. Immunization parameters were selected for 50% protection based on prior clinical data. Leukapheresis was done to collect plasma and peripheral blood mononuclear cells. RESULTS: Adverse event rates were similar in true- and mock-immunized subjects. Two true- and two mock-immunized subjects developed large local reactions likely caused by mosquito salivary gland antigens. In Cohort 1, 11 subjects received 810-1235 infected bites; 6/11 (55%) were protected against CHMI vs. 0/3 mock-immunized and 0/6 infectivity controls (VE 55%). In Cohort 2, 10 subjects received 839-1131 infected bites with a higher first dose and a reduced fifth dose; 9/10 (90%) were protected vs. 0/2 mock-immunized and 0/6 controls (VE 90%). Three/3 (100%) protected subjects administered three booster immunizations were protected against repeat CHMI vs. 0/6 controls (VE 100%). Cohort 2 uniquely showed a significant rise in IFN-γ responses after the third and fifth immunizations and higher antibody responses to CSP. CONCLUSIONS: PfRAS were generally safe and well tolerated. Cohort 2 had a higher first dose, reduced final dose, higher antibody responses to CSP and significant rise of IFN-γ responses after the third and fifth immunizations. Whether any of these factors contributed to increased protection in Cohort 2 requires further investigation. A cryobank of sera and cells from protected and non-protected individuals was generated for future immunological studies and antigen discovery. TRIAL REGISTRATION: ClinicalTrials.gov NCT01994525.


Assuntos
Mordeduras e Picadas de Insetos/imunologia , Malária/prevenção & controle , Esporozoítos/imunologia , Vacinação/métodos , Vacinas Atenuadas/efeitos adversos , Adulto , Animais , Anopheles/parasitologia , Anopheles/fisiologia , Feminino , Raios gama , Humanos , Malária/imunologia , Masculino , Pessoa de Meia-Idade , Mosquitos Vetores/parasitologia , Mosquitos Vetores/fisiologia , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/imunologia , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/imunologia , Esporozoítos/patogenicidade , Esporozoítos/efeitos da radiação , Vacinação/efeitos adversos
16.
J Med Entomol ; 56(5): 1359-1367, 2019 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-31134270

RESUMO

Attractive toxic sugar baits (ATSBs) can be an effective vector control tool, especially in areas where aerial or aquatic applications of pesticides are undesirable or impractical. In general, there is a need to develop novel or alternative insecticides for vector control, and there is a demand from consumers for more 'natural' pest control products. Sodium ascorbate (SA) is a naturally occurring antioxidant compound, found in fruits and vegetables, and is available commercially in the United States as a food additive and supplement. In this study, we continuously exposed groups of adult Aedes aegypti (L.), Anopheles stephensi Liston (Diptera: Culicidae), Phlebotomus papatasi Scopoli, and Lutzomyia longipalpis (Lutz & Neiva; Diptera: Psychodidae) to ATSBs containing SA in concentrations of 6, 8, 10, and 20%, and tracked their mortality over 10 d. We also exposed insects to a 20% SA-ATSB on a single day to determine the effect of a single exposure to the bait on mortality. Concentrations of ≥8% SA significantly reduced survival of both mosquito species over 10 d compared with sugar-fed controls. Sand fly mortality was inconsistent. A single exposure to 20% SA significantly reduced the survival of An. stephensi. Mosquitoes exposed to SA exhibited elevated catalase levels and cell death. The use of SA in ATSBs may be most effective in areas where sugar sources are scarce, and where mosquito species frequently sugar-feed. SA sugar baits may be a particularly attractive option for the general public looking to control mosquito populations using 'natural' alternatives to synthetic insecticides.


Assuntos
Ácido Ascórbico , Culicidae , Controle de Insetos , Inseticidas , Psychodidae , Açúcares , Animais , Relação Dose-Resposta a Droga , Feminino , Masculino , Controle de Mosquitos
17.
Dev Comp Immunol ; 32(5): 519-31, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18036658

RESUMO

Immunoglobulin superfamily (IgSF) proteins are known for their ability to specifically recognize and adhere to other molecules, mediating cell-surface reception and pathogen recognition. Mammalian IgSF proteins such as antibodies are among the best characterized molecules of the immune system; in contrast, the involvement of invertebrate IgSF members in immunity has not been broadly studied. Analysis of the predicted Anopheles gambiae transcriptome identified 138 proteins that have at least one immunoglobulin domain. Challenge with Plasmodium, Gram-negative or Gram-positive bacteria resulted in significant regulation of 85 IgSF genes, indicating potential roles for these molecules in infection responses and immunity. Based on sequence and expression data, six infection-responsive with immunoglobulin domain (IRID 1-6) genes were chosen and functionally characterized with regard to their role in innate immunity. Reverse-genetic gene-silencing assays showed IRID3, IRID5 and IRID6 contribute to viability upon bacterial infection while IRID4 and IRID6 are involved in limiting Plasmodium falciparum infection.


Assuntos
Anopheles/imunologia , Imunidade Inata , Imunoglobulinas/fisiologia , Proteínas de Insetos/fisiologia , Animais , Anopheles/microbiologia , Anopheles/parasitologia , Drosophila melanogaster/imunologia , Regulação da Expressão Gênica , Genes de Imunoglobulinas , Especificidade de Órgãos , Plasmodium falciparum/crescimento & desenvolvimento
18.
JCI Insight ; 2(1): e89154, 2017 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-28097230

RESUMO

BACKGROUND: A radiation-attenuated Plasmodium falciparum (Pf) sporozoite (SPZ) malaria vaccine, PfSPZ Vaccine, protected 6 of 6 subjects (100%) against homologous Pf (same strain as in the vaccine) controlled human malaria infection (CHMI) 3 weeks after 5 doses administered intravenously. The next step was to assess protective efficacy against heterologous Pf (different from Pf in the vaccine), after fewer doses, and at 24 weeks. METHODS: The trial assessed tolerability, safety, immunogenicity, and protective efficacy of direct venous inoculation (DVI) of 3 or 5 doses of PfSPZ Vaccine in non-immune subjects. RESULTS: Three weeks after final immunization, 5 doses of 2.7 × 105 PfSPZ protected 12 of 13 recipients (92.3% [95% CI: 48.0, 99.8]) against homologous CHMI and 4 of 5 (80.0% [10.4, 99.5]) against heterologous CHMI; 3 doses of 4.5 × 105 PfSPZ protected 13 of 15 (86.7% [35.9, 98.3]) against homologous CHMI. Twenty-four weeks after final immunization, the 5-dose regimen protected 7 of 10 (70.0% [17.3, 93.3]) against homologous and 1 of 10 (10.0% [-35.8, 45.6]) against heterologous CHMI; the 3-dose regimen protected 8 of 14 (57.1% [21.5, 76.6]) against homologous CHMI. All 22 controls developed Pf parasitemia. PfSPZ Vaccine was well tolerated, safe, and easy to administer. No antibody or T cell responses correlated with protection. CONCLUSIONS: We have demonstrated for the first time to our knowledge that PfSPZ Vaccine can protect against a 3-week heterologous CHMI in a limited group of malaria-naive adult subjects. A 3-dose regimen protected against both 3-week and 24-week homologous CHMI (87% and 57%, respectively) in this population. These results provide a foundation for developing an optimized immunization regimen for preventing malaria. TRIAL REGISTRATION: ClinicalTrials.gov NCT02215707. FUNDING: Support was provided through the US Army Medical Research and Development Command, Military Infectious Diseases Research Program, and the Naval Medical Research Center's Advanced Medical Development Program.


Assuntos
Malária Falciparum/terapia , Plasmodium falciparum/efeitos dos fármacos , Esporozoítos/efeitos dos fármacos , Vacinas Atenuadas/administração & dosagem , Administração Intravenosa , Adulto , Feminino , Humanos , Malária Falciparum/prevenção & controle , Masculino , Plasmodium falciparum/genética , Esporozoítos/genética , Linfócitos T/imunologia , Vacinas Atenuadas/uso terapêutico , Sequenciamento Completo do Genoma/métodos
19.
Nat Med ; 22(6): 614-23, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27158907

RESUMO

An attenuated Plasmodium falciparum (Pf) sporozoite (SPZ) vaccine, PfSPZ Vaccine, is highly protective against controlled human malaria infection (CHMI) 3 weeks after immunization, but the durability of protection is unknown. We assessed how vaccine dosage, regimen, and route of administration affected durable protection in malaria-naive adults. After four intravenous immunizations with 2.7 × 10(5) PfSPZ, 6/11 (55%) vaccinated subjects remained without parasitemia following CHMI 21 weeks after immunization. Five non-parasitemic subjects from this dosage group underwent repeat CHMI at 59 weeks, and none developed parasitemia. Although Pf-specific serum antibody levels correlated with protection up to 21-25 weeks after immunization, antibody levels waned substantially by 59 weeks. Pf-specific T cell responses also declined in blood by 59 weeks. To determine whether T cell responses in blood reflected responses in liver, we vaccinated nonhuman primates with PfSPZ Vaccine. Pf-specific interferon-γ-producing CD8 T cells were present at ∼100-fold higher frequencies in liver than in blood. Our findings suggest that PfSPZ Vaccine conferred durable protection to malaria through long-lived tissue-resident T cells and that administration of higher doses may further enhance protection.


Assuntos
Anticorpos Antiprotozoários/imunologia , Linfócitos T CD8-Positivos/imunologia , Imunogenicidade da Vacina/imunologia , Fígado/imunologia , Vacinas Antimaláricas/uso terapêutico , Malária Falciparum/prevenção & controle , Parasitemia/prevenção & controle , Plasmodium falciparum/imunologia , Administração Intravenosa , Adolescente , Adulto , Animais , Ensaio de Imunoadsorção Enzimática , Feminino , Voluntários Saudáveis , Humanos , Imunoglobulina G/imunologia , Interferon gama/imunologia , Fígado/citologia , Macaca mulatta , Vacinas Antimaláricas/imunologia , Masculino , Pessoa de Meia-Idade , Parasitemia/imunologia , Esporozoítos/imunologia , Linfócitos T/imunologia , Adulto Jovem
20.
US Army Med Dep J ; : 16-24, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26276942

RESUMO

Thirty years ago, the Entomology Branch at the Walter Reed Army Institute of Research (WRAIR) performed the first controlled human malaria infection, in which lab-reared mosquitoes were infected with lab-cultured malaria parasites and allowed to feed on human volunteers. The development of this model was a turning point for pre-erythrocytic malaria vaccine research and, through decades of refinement, has supported 30 years of efficacy testing of a suite of antimalarial vaccines and drugs. In this article, we present a historical overview of the research that enabled the first challenge to occur and the modifications made to the challenge over time, a summary of the 104 challenges performed by WRAIR from the first into 2015, and a prospective look at what the next generation of challenges might entail.


Assuntos
Academias e Institutos/história , Vacinas Antimaláricas , Malária/prevenção & controle , Medicina Militar , Projetos de Pesquisa , Voluntários Saudáveis , História do Século XX , Experimentação Humana/história , Humanos , Vacinas Antimaláricas/história , Vacinas Antimaláricas/uso terapêutico , Medicina Militar/história , Medicina Militar/métodos , Estados Unidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA