Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Microbes Infect ; 26(5-6): 105343, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38670216

RESUMO

Hemozoin is a crystal synthesized by Plasmodium parasites during hemoglobin digestion in the erythrocytic stage. The hemozoin released when the parasites egress from the red blood cell, which is complexed with parasite DNA, is cleared from the circulation by circulating and tissue-resident monocytes and macrophages, respectively. Recently, we reported that intravenous administration of purified hemozoin complexed with Plasmodium berghei DNA (HzPbDNA) resulted in an innate immune response that blocked liver stage development of sporozoites that was dose-dependent and time-limited. Here, we further characterize the organismal, cellular, and molecular events associated with this protective innate response in the liver and report that a large proportion of the IV administered HzPbDNA localized to F4/80+ cells in the liver and that the rapid and strong protection against liver-stage development waned quickly such that by 1 week post-HzPbDNA treatment animals were fully susceptible to infection. RNAseq of the liver after IV administration of HzPbDNA demonstrated that the rapid and robust induction of genes associated with the acute phase response, innate immune activation, cellular recruitment, and IFN-γ signaling observed at day 1 was largely absent at day 7. RNAseq analysis implicated NK cells as the major cellular source of IFN-γ. In vivo cell depletion and IFN-γ neutralization experiments supported the hypothesis that tissue-resident macrophages and NK cells are major contributors to the protective response and the NK cell-derived IFN-γ is key to induction of the mechanisms that block sporozoite development in the liver. These findings advance our understanding of the innate immune responses that prevent liver stage malaria infection.


Assuntos
Hemeproteínas , Imunidade Inata , Interferon gama , Fígado , Malária , Plasmodium berghei , Esporozoítos , Animais , Plasmodium berghei/imunologia , Esporozoítos/imunologia , Malária/imunologia , Malária/prevenção & controle , Malária/parasitologia , Hemeproteínas/imunologia , Camundongos , Fígado/parasitologia , Fígado/imunologia , Interferon gama/imunologia , Interferon gama/metabolismo , Camundongos Endogâmicos C57BL , Macrófagos/imunologia , Macrófagos/parasitologia , DNA de Protozoário/genética , Feminino
2.
Cell Rep ; 38(7): 110367, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35172158

RESUMO

L9 is a potent human monoclonal antibody (mAb) that preferentially binds two adjacent NVDP minor repeats and cross-reacts with NANP major repeats of the Plasmodium falciparum circumsporozoite protein (PfCSP) on malaria-infective sporozoites. Understanding this mAb's ontogeny and mechanisms of binding PfCSP will facilitate vaccine development. Here, we isolate mAbs clonally related to L9 and show that this B cell lineage has baseline NVDP affinity and evolves to acquire NANP reactivity. Pairing the L9 kappa light chain (L9κ) with clonally related heavy chains results in chimeric mAbs that cross-link two NVDPs, cross-react with NANP, and more potently neutralize sporozoites in vivo compared with their original light chain. Structural analyses reveal that the chimeric mAbs bound minor repeats in a type-1 ß-turn seen in other repeat-specific antibodies. These data highlight the importance of L9κ in binding NVDP on PfCSP to neutralize sporozoites and suggest that PfCSP-based immunogens might be improved by presenting ≥2 NVDPs.


Assuntos
Anticorpos Monoclonais/metabolismo , Cadeias Leves de Imunoglobulina/metabolismo , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Proteínas de Protozoários/metabolismo , Sequências Repetitivas de Aminoácidos , Adolescente , Adulto , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/isolamento & purificação , Linhagem da Célula , Culicidae/parasitologia , Feminino , Humanos , Fragmentos Fab das Imunoglobulinas/metabolismo , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Modelos Moleculares , Testes de Neutralização , Peptídeos/química , Peptídeos/metabolismo , Plasmodium falciparum/imunologia , Ligação Proteica , Adulto Jovem
3.
Cell ; 184(17): 4512-4530.e22, 2021 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-34343496

RESUMO

Cytotoxic T lymphocyte (CTL) responses against tumors are maintained by stem-like memory cells that self-renew but also give rise to effector-like cells. The latter gradually lose their anti-tumor activity and acquire an epigenetically fixed, hypofunctional state, leading to tumor tolerance. Here, we show that the conversion of stem-like into effector-like CTLs involves a major chemotactic reprogramming that includes the upregulation of chemokine receptor CXCR6. This receptor positions effector-like CTLs in a discrete perivascular niche of the tumor stroma that is densely occupied by CCR7+ dendritic cells (DCs) expressing the CXCR6 ligand CXCL16. CCR7+ DCs also express and trans-present the survival cytokine interleukin-15 (IL-15). CXCR6 expression and IL-15 trans-presentation are critical for the survival and local expansion of effector-like CTLs in the tumor microenvironment to maximize their anti-tumor activity before progressing to irreversible dysfunction. These observations reveal a cellular and molecular checkpoint that determines the magnitude and outcome of anti-tumor immune responses.


Assuntos
Receptores CXCR6/metabolismo , Linfócitos T Citotóxicos/imunologia , Microambiente Tumoral , Animais , Antígeno B7-H1/metabolismo , Comunicação Celular , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Quimiocina CXCL16 , Células Dendríticas/metabolismo , Interleucina-12/metabolismo , Interleucina-15/metabolismo , Ligantes , Linfonodos/metabolismo , Melanoma/imunologia , Melanoma/patologia , Camundongos Endogâmicos C57BL
4.
Nat Commun ; 12(1): 1063, 2021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33594061

RESUMO

The most advanced P. falciparum circumsporozoite protein-based malaria vaccine, RTS,S/AS01 (RTS,S), confers partial protection but with antibody titers that wane relatively rapidly, highlighting the need to elicit more potent and durable antibody responses. Here, we elucidate crystal structures, binding affinities and kinetics, and in vivo protection of eight anti-NANP antibodies derived from an RTS,S phase 2a trial and encoded by three different heavy-chain germline genes. The structures reinforce the importance of homotypic Fab-Fab interactions in protective antibodies and the overwhelmingly dominant preference for a germline-encoded aromatic residue for recognition of the NANP motif. In this study, antibody apparent affinity correlates best with protection in an in vivo mouse model, with the more potent antibodies also recognizing epitopes with repeating secondary structural motifs of type I ß- and Asn pseudo 310 turns; such insights can be incorporated into design of more effective immunogens and antibodies for passive immunization.


Assuntos
Anticorpos Antiprotozoários/imunologia , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Plasmodium falciparum/imunologia , Sequências Repetitivas de Aminoácidos , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Afinidade de Anticorpos/imunologia , Cristalografia por Raios X , Epitopos/química , Epitopos/imunologia , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/imunologia , Cinética , Camundongos Endogâmicos C57BL , Modelos Moleculares , Parasitos/imunologia , Peptídeos/química , Peptídeos/metabolismo , Ligação Proteica
5.
JCI Insight ; 6(3)2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33332286

RESUMO

CIS43 is a potent neutralizing human mAb that targets a highly conserved "junctional" epitope in the Plasmodium falciparum (Pf) circumsporozoite protein (PfCSP). Enhancing the durability of CIS43 in vivo will be important for clinical translation. Here, 2 approaches were used to improve the durability of CIS43 in vivo while maintaining potent neutralization. First, the Fc domain was modified with the LS mutations (CIS43LS) to increase CIS43 binding affinity for the neonatal Fc receptor (FcRn). CIS43LS and CIS43 showed comparable in vivo protective efficacy. CIS43LS had 9- to 13-fold increased binding affinity for human (6.2 nM versus 54.2 nM) and rhesus (25.1 nM versus 325.8 nM) FcRn at endosomal pH 6.0 compared with CIS43. Importantly, the half-life of CIS43LS in rhesus macaques increased from 22 days to 39 days compared with CIS43. The second approach for sustaining antibody levels of CIS43 in vivo is through adeno-associated virus (AAV) expression. Mice administered once with AAV-expressing CIS43 had sustained antibody levels of approximately 300 µg/mL and mediated protection against sequential malaria challenges up to 36 weeks. Based on these data, CIS43LS has advanced to phase I clinical trials, and AAV delivery provides a potential next-generation approach for malaria prevention.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Malária Falciparum/prevenção & controle , Plasmodium falciparum/imunologia , Substituição de Aminoácidos , Animais , Anticorpos Anti-Idiotípicos/biossíntese , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/farmacocinética , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/genética , Anticorpos Antiprotozoários/administração & dosagem , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/genética , Dependovirus/genética , Feminino , Humanos , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Fragmentos Fc das Imunoglobulinas/genética , Macaca mulatta , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mutagênese Sítio-Dirigida , Proteínas de Protozoários/imunologia
6.
Immunity ; 53(4): 733-744.e8, 2020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-32946741

RESUMO

Discovering potent human monoclonal antibodies (mAbs) targeting the Plasmodium falciparum circumsporozoite protein (PfCSP) on sporozoites (SPZ) and elucidating their mechanisms of neutralization will facilitate translation for passive prophylaxis and aid next-generation vaccine development. Here, we isolated a neutralizing human mAb, L9 that preferentially bound NVDP minor repeats of PfCSP with high affinity while cross-reacting with NANP major repeats. L9 was more potent than six published neutralizing human PfCSP mAbs at mediating protection against mosquito bite challenge in mice. Isothermal titration calorimetry and multiphoton microscopy showed that L9 and the other most protective mAbs bound PfCSP with two binding events and mediated protection by killing SPZ in the liver and by preventing their egress from sinusoids and traversal of hepatocytes. This study defines the subdominant PfCSP minor repeats as neutralizing epitopes, identifies an in vitro biophysical correlate of SPZ neutralization, and demonstrates that the liver is an important site for antibodies to prevent malaria.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antiprotozoários/imunologia , Antimaláricos/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Esporozoítos/imunologia , Adolescente , Adulto , Animais , Linhagem Celular , Linhagem Celular Tumoral , Epitopos/imunologia , Feminino , Células HEK293 , Hepatócitos/imunologia , Hepatócitos/parasitologia , Humanos , Fígado/imunologia , Fígado/parasitologia , Malária/imunologia , Malária/parasitologia , Vacinas Antimaláricas/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Adulto Jovem
7.
Malar J ; 16(1): 110, 2017 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-28279180

RESUMO

BACKGROUND: Primaquine is an anti-malarial used to prevent Plasmodium vivax relapses and malaria transmission. However, PQ metabolites cause haemolysis in patients deficient in the enzyme glucose-6-phosphate dehydrogenase (G6PD). Fifteen PQ-thiazolidinone derivatives, synthesized through one-post reactions from primaquine, arenealdehydes and mercaptoacetic acid, were evaluated in parallel in several biological assays, including ability to block malaria transmission to mosquitoes. RESULTS: All primaquine derivatives (PQ-TZs) exhibited lower cell toxicity than primaquine; none caused haemolysis to normal or G6PD-deficient human erythrocytes in vitro. Sera from mice pretreated with the test compounds thus assumed to have drug metabolites, caused no in vitro haemolysis of human erythrocytes, whereas sera from mice pretreated with primaquine did cause haemolysis. The ability of the PQ-TZs to block malaria transmission was evaluated based on the oocyst production and percentage of mosquitoes infected after a blood meal in drug pre-treated animals with experimental malaria caused by either Plasmodium gallinaceum or Plasmodium berghei; four and five PQ-TZs significantly inhibited sporogony in avian and in rodent malaria, respectively. Selected PQ-TZs were tested for their inhibitory activity on P. berghei liver stage development, in mice and in vitro, one compound (4m) caused a 3-day delay in the malaria pre-patent period. CONCLUSIONS: The compound 4m was the most promising, blocking malaria transmissions and reducing the number of exoerythrocytic forms of P. berghei (EEFs) in hepatoma cells in vitro and in mice in vivo. The same compound also caused a 3-day delay in the malaria pre-patent period.


Assuntos
Eritrócitos/parasitologia , Glucosefosfato Desidrogenase/metabolismo , Malária/tratamento farmacológico , Plasmodium berghei/efeitos dos fármacos , Plasmodium gallinaceum/efeitos dos fármacos , Primaquina/análogos & derivados , Primaquina/farmacologia , Animais , Linhagem Celular Tumoral , Galinhas , Chlorocebus aethiops , Eritrócitos/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Células Hep G2 , Humanos , Malária/transmissão , Malária Aviária/tratamento farmacológico , Malária Aviária/transmissão , Camundongos , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium gallinaceum/crescimento & desenvolvimento
8.
Semin Immunopathol ; 37(3): 211-20, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25917387

RESUMO

Parasitic protozoa cause considerable disease in humans and, due to their intracellular life cycle, induce robust CD8(+) T cell responses. A greater understanding of the factors that promote and maintain CD8(+) T cell-mediated immunity against these pathogens is likely needed for the development of effective vaccines. Immunization with radiation-attenuated sporozoites, the infectious stage of the malaria parasite transmitted by mosquitoes, is an excellent model to study these questions as CD8(+) T cells specific for a single epitope can completely eliminate parasite infection in the liver. Furthermore, live, radiation-attenuated parasites represent the "gold standard" for malaria vaccination. Here, we will highlight recent studies aimed at understanding the factors required for the induction, recruitment, and maintenance of effector and memory CD8(+) T cells against malaria liver stages.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Interações Hospedeiro-Parasita/imunologia , Fígado/imunologia , Linfonodos/imunologia , Malária/imunologia , Plasmodium/imunologia , Imunidade Adaptativa , Apresentação de Antígeno/imunologia , Antígenos de Protozoários , Movimento Celular , Citotoxicidade Imunológica , Humanos , Imunidade Inata , Memória Imunológica , Estágios do Ciclo de Vida/imunologia , Malária/parasitologia , Malária/prevenção & controle , Vacinas Antimaláricas/imunologia , Especificidade de Órgãos/imunologia , Plasmodium/crescimento & desenvolvimento
9.
Infect Immun ; 83(1): 268-75, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25368113

RESUMO

Decades of success with live adenovirus vaccines suggest that replication-competent recombinant adenoviruses (rAds) could serve as effective vectors for immunization against other pathogens. To explore the potential of a live rAd vaccine against malaria, we prepared a viable adenovirus 5 (Ad5) recombinant that displays a B-cell epitope from the circumsporozoite protein (CSP) of Plasmodium falciparum on the virion surface. The recombinant induced P. falciparum sporozoite-neutralizing antibodies in mice. Human adenoviruses do not replicate in mice. Therefore, to examine immunogenicity in a system in which, as in humans, the recombinant replicates, we constructed a similar recombinant in an adenovirus mutant that replicates in monkey cells and immunized four Aotus nancymaae monkeys. The recombinant replicated in the monkeys after intratracheal instillation, the first demonstration of replication of human adenoviruses in New World monkeys. Immunization elicited antibodies both to the Plasmodium epitope and the Ad5 vector. Antibodies from all four monkeys recognized CSP on intact parasites, and plasma from one monkey neutralized sporozoites in vitro and conferred partial protection against P. falciparum sporozoite infection after passive transfer to mice. Prior enteric inoculation of two animals with antigenically wild-type adenovirus primed a response to the subsequent intratracheal inoculation, suggesting a route to optimizing performance. A vaccine is not yet available against P. falciparum, which induces the deadliest form of malaria and kills approximately one million children each year. The live capsid display recombinant described here may constitute an early step in a critically needed novel approach to malaria immunization.


Assuntos
Adenoviridae/genética , Anticorpos Antiprotozoários/sangue , Portadores de Fármacos , Vacinas Antimaláricas/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Animais , Anticorpos Neutralizantes/sangue , Aotidae , Técnicas de Visualização da Superfície Celular , Feminino , Vetores Genéticos , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/genética , Masculino , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
10.
PLoS One ; 9(9): e107764, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25247295

RESUMO

The circumsporozoite protein (CSP) of Plasmodium falciparum is a major surface protein, which forms a dense coat on the sporozoite's surface. Preclinical research on CSP and clinical evaluation of a CSP fragment-based RTS, S/AS01 vaccine have demonstrated a modest degree of protection against P. falciparum, mediated in part by humoral immunity and in part by cell-mediated immunity. Given the partial protective efficacy of the RTS, S/AS01 vaccine in a recent Phase 3 trial, further improvement of CSP-based vaccines is crucial. In this report, we describe the preclinical development of a full-length, recombinant CSP (rCSP)-based vaccine candidate against P. falciparum malaria suitable for current Good Manufacturing Practice (cGMP) production. Utilizing a novel high-throughput Pseudomonas fluorescens expression platform, we demonstrated greater efficacy of full-length rCSP as compared to N-terminally truncated versions, rapidly down-selected a promising lead vaccine candidate, and developed a high-yield purification process to express immunologically active, intact antigen for clinical trial material production. The rCSP, when formulated with various adjuvants, induced antigen-specific antibody responses as measured by enzyme-linked immunosorbent assay (ELISA) and immunofluorescence assay (IFA), as well as CD4+ T-cell responses as determined by ELISpot. The adjuvanted rCSP vaccine conferred protection in mice when challenged with transgenic P. berghei sporozoites containing the P. falciparum repeat region of CSP. Furthermore, heterologous prime/boost regimens with adjuvanted rCSP and an adenovirus type 35-vectored CSP (Ad35CS) showed modest improvements in eliciting CSP-specific T-cell responses and anti-malarial protection, depending on the order of vaccine delivery. Collectively, these data support the importance of further clinical development of adjuvanted rCSP, either as a stand-alone product or as one of the components in a heterologous prime/boost strategy, ultimately acting as an effective vaccine candidate for the mitigation of P. falciparum-induced malaria.


Assuntos
Vacinas Antimaláricas/administração & dosagem , Malária Falciparum/prevenção & controle , Plasmodium falciparum/imunologia , Proteínas de Protozoários/metabolismo , Pseudomonas fluorescens/genética , Proteínas Recombinantes/metabolismo , Adjuvantes Imunológicos/farmacologia , Animais , Modelos Animais de Doenças , Feminino , Células Hep G2 , Humanos , Vacinas Antimaláricas/química , Malária Falciparum/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Organismos Geneticamente Modificados , Proteínas de Protozoários/genética , Pseudomonas fluorescens/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Vacinação/métodos
11.
Proc Natl Acad Sci U S A ; 111(34): 12528-32, 2014 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-25114213

RESUMO

Malaria caused by Plasmodium falciparum kills nearly one million children each year and imposes crippling economic burdens on families and nations worldwide. No licensed vaccine exists, but infection can be prevented by antibodies against the circumsporozoite protein (CSP), the major surface protein of sporozoites, the form of the parasite injected by mosquitoes. We have used vectored immunoprophylaxis (VIP), an adeno-associated virus-based technology, to introduce preformed antibody genes encoding anti-P. falciparum CSP mAb into mice. VIP vector-transduced mice exhibited long-lived mAb expression at up to 1,200 µg/mL in serum, and up to 70% were protected from both i.v. and mosquito bite challenge with transgenic Plasmodium berghei rodent sporozoites that incorporate the P. falciparum target of the mAb in their CSP. Serum antibody levels and protection from mosquito bite challenge were dependent on the dose of the VIP vector. All individual mice expressing CSP-specific mAb 2A10 at 1 mg/mL or more were completely protected, suggesting that in this model system, exceeding that threshold results in consistent sterile protection. Our results demonstrate the potential of VIP as a path toward the elusive goal of immunization against malaria.


Assuntos
Técnicas de Transferência de Genes , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/genética , Malária Falciparum/prevenção & controle , Plasmodium falciparum/imunologia , Animais , Anticorpos Monoclonais Murinos/biossíntese , Anticorpos Monoclonais Murinos/genética , Anticorpos Neutralizantes/biossíntese , Anticorpos Neutralizantes/genética , Anticorpos Antiprotozoários/biossíntese , Anticorpos Antiprotozoários/genética , Dependovirus/genética , Modelos Animais de Doenças , Feminino , Vetores Genéticos , Humanos , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium berghei/genética , Plasmodium berghei/imunologia , Plasmodium falciparum/genética , Esporozoítos/imunologia
12.
Infect Immun ; 81(3): 789-800, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23275094

RESUMO

The Plasmodium falciparum circumsporozoite (CS) protein (CSP) is a major vaccine target for preventing malaria infection. Thus, developing strong and durable antibody and T cell responses against CSP with novel immunogens and potent adjuvants may improve upon the success of current approaches. Here, we compare four distinct full-length P. falciparum CS proteins expressed in Escherichia coli or Pichia pastoris for their ability to induce immunity and protection in mice when administered with long-chain poly(I · C) [poly(I · C)LC] as an adjuvant. CS proteins expressed in E. coli induced high-titer antibody responses against the NANP repeat region and potent CSP-specific CD4(+) T cell responses. Moreover, E. coli-derived CS proteins in combination with poly(I · C)LC induced potent multifunctional (interleukin 2-positive [IL-2(+)], tumor necrosis factor alpha-positive [TNF-α(+)], gamma interferon-positive [IFN-γ(+)]) CD4(+) effector T cell responses in blood, in spleen, and particularly in liver. Using transgenic Plasmodium berghei expressing the repeat region of P. falciparum CSP [Pb-CS(Pf)], we showed that there was a 1- to 4-log decrease in malaria rRNA in the liver following a high-dose challenge and ~50% sterilizing protection with a low-dose challenge compared to control levels. Protection was directly correlated with high-level antibody titers but not CD4(+) T cell responses. Finally, protective immunity was also induced using the Toll-like receptor 4 agonist glucopyranosyl lipid adjuvant-stable emulsion (GLA-SE) as the adjuvant, which also correlated with high antibody titers yet CD4(+) T cell immunity that was significantly less potent than that with poly(I · C)LC. Overall, these data suggest that full-length CS proteins and poly(I · C)LC or GLA-SE offer a simple vaccine formulation to be used alone or in combination with other vaccines for preventing malaria infection.


Assuntos
Anticorpos Antiprotozoários/imunologia , Linfócitos T CD4-Positivos/imunologia , Lipídeos/farmacologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/metabolismo , Receptor 4 Toll-Like/agonistas , Animais , Linfócitos T CD4-Positivos/fisiologia , Relação Dose-Resposta Imunológica , Emulsões , Escherichia coli/genética , Escherichia coli/metabolismo , Regulação da Expressão Gênica , Lipídeos/química , Malária/prevenção & controle , Vacinas Antimaláricas/imunologia , Camundongos , Organismos Geneticamente Modificados , Pichia/genética , Pichia/metabolismo , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Plasmodium falciparum/fisiologia , Proteínas de Protozoários/genética , Fatores de Tempo
13.
Mem. Inst. Oswaldo Cruz ; 106(supl.1): 172-178, Aug. 2011.
Artigo em Inglês | LILACS | ID: lil-597259

RESUMO

CD8+ T cells against malaria liver stages represent a major protective immune mechanism against infection. Following induction in the peripheral lymph nodes by dendritic cells (DCs), these CD8+ T cells migrate to the liver and eliminate parasite infected hepatocytes. The processing and presentation of sporozoite antigen requires TAP mediated transport of major histocompatibility complex class I epitopes to the endoplasmic reticulum. Importantly, in DCs this process is also dependent on endosome-mediated cross presentation while this mechanism is not required for epitope presentation on hepatocytes. Protective CD8+ T cell responses are strongly dependent on the presence of CD4+ T cells and the capacity of sporozoite antigen to persist for a prolonged period of time. While human trials with subunit vaccines capable of inducing antibodies and CD4+ T cell responses have yielded encouraging results, an effective anti-malaria vaccine will likely require vaccine constructs designed to induce protective CD8+ T cells against malaria liver stages.


Assuntos
Animais , Humanos , Camundongos , Antígenos de Protozoários/imunologia , /imunologia , Hepatócitos , Fígado , Malária/imunologia , /imunologia , Epitopos/imunologia , Vacinas Antimaláricas/imunologia , Malária , Malária
14.
Vaccine ; 29(8): 1683-9, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21199707

RESUMO

Adenovirus particles can be engineered to display exogenous peptides on their surfaces by modification of viral capsid proteins, and particles that display pathogen-derived peptides can induce protective immunity. We constructed viable recombinant adenoviruses that display B-cell epitopes from the Plasmodium falciparum circumsporozoite protein (PfCSP) in the major adenovirus capsid protein, hexon. Recombinants induced high-titer antibodies against CSP when injected intraperitoneally into mice. Serum obtained from immunized mice recognized both recombinant PfCSP protein and P. falciparum sporozoites, and neutralized P. falciparum sporozoites in vitro. Replicating adenovirus vaccines have provided economical protection against adenovirus disease for over three decades. The recombinants described here may provide a path to an affordable malaria vaccine in the developing world.


Assuntos
Adenoviridae/imunologia , Anticorpos Neutralizantes/sangue , Anticorpos Antiprotozoários/imunologia , Vacinas Antimaláricas/imunologia , Proteínas de Protozoários/imunologia , Animais , Anopheles/virologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antiprotozoários/sangue , Proteínas do Capsídeo/imunologia , Epitopos de Linfócito B/imunologia , Feminino , Células Hep G2 , Humanos , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Testes de Neutralização , Plasmodium falciparum/imunologia , Proteínas Recombinantes/imunologia
15.
PLoS One ; 6(1): e15948, 2011 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-21245909

RESUMO

CD4(+) helper T cells are critical orchestrators of immune responses to infection and vaccination. During primary responses, naïve CD8(+) T cells may need "CD4 help" for optimal development of memory populations. The immunological factors attributed to CD4 help depend on the context of immunization and vary depending on the priming system. In response to immunization with radiation-attenuated Plasmodium yoelii sporozoites, CD8(+) T cells in BALB/c mice fail to generate large numbers of effector cells without help from CD4(+) T cells--a defect not observed in most systems. Given this unique early dependence on CD4 help, we evaluated the effects of CD4(+) cells on the development of functional properties of CD8(+) T cells and on their ability to abolish infection. First, we determined that this effect was not mediated by CD4(+) non-T cells and did not involve CD1d-restricted NKT cells. We found that CD8(+) T cells induced by sporozoites without CD4 help formed memory populations severely reduced in magnitude that could not limit parasite development in the liver. The inability of these "helpless" memory T cells to protect is not a result of defects in effector function, as their capacity to produce cytokines and undergo cytotoxic degranulation was indistinguishable from control memory T cells. These data indicate that CD4(+) T help may not be necessary to develop the functional attributes of CD8(+) T cells; however they are crucial to ensure the survival of effector and memory cells induced in primary responses.


Assuntos
Linfócitos T CD4-Positivos/parasitologia , Linfócitos T CD8-Positivos/parasitologia , Proliferação de Células , Sobrevivência Celular/imunologia , Esporozoítos/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Citotoxicidade Imunológica , Memória Imunológica/imunologia , Fígado/parasitologia , Malária/imunologia , Camundongos , Camundongos Endogâmicos BALB C
16.
Eur J Immunol ; 40(1): 124-33, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19830730

RESUMO

Synthetic peptides encoding protective pathogen-derived epitopes represent--in principle--an ideal approach to T-cell vaccination. Empirically, however, these strategies have not been successful. In the current study, we profiled the early activation of CD8+ T cells by MHC class I-restricted peptide immunization to better understand the biology of this response. We found that CD8+ T cells proliferated robustly in response to low doses of short synthetic peptides in PBS, but failed to acquire effector function or form memory populations in the absence of the TLR ligand CpG. CpG was unique among TLR ligands in its ability to enhance the response to peptide and its adjuvant effects had strict temporal requirements. Interestingly, CpG treatment modulated T-cell expression of the surface receptors PD-1 and CD25, providing insight into its possible adjuvant mechanism. The effects of CpG on peptide immunization were dramatically enhanced in the absence of B cells, demonstrating a unique system of regulation of T-cell responses by these lymphocytes. The results reported here provide insight into the complex response to a simple vaccination regimen, as well as a framework for a rational peptide-based vaccine design to both exploit and overcome targeted aspects of the immune response.


Assuntos
Linfócitos B/imunologia , Linfócitos T CD8-Positivos/imunologia , Ilhas de CpG , Antígenos de Histocompatibilidade Classe I/imunologia , Animais , Linfócitos T CD8-Positivos/citologia , Proliferação de Células , Feminino , Imunização , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Peptídeos/imunologia
17.
FEMS Immunol Med Microbiol ; 52(2): 237-46, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18177343

RESUMO

Live Salmonella vaccines are limited in use by the inherent toxicity of the lipopolysaccharide. The waaN gene encodes a myristyl transferase required for the secondary acylation of lipid A in lipopolysaccharide. A waaN mutant exhibits reduced induction of the inflammatory cytokines associated with lipopolysaccharide toxicity. Here the characteristics of a Salmonella enterica serovar Typhimurium aroA waaN mutant (SK100) in vitro and in vivo compared with its parent aroA strain (SL3261) were described. Phenotypic analysis of purified lipopolysaccharide obtained from SK100 confirmed that the physical and biological activities of the lipopolysaccharide had been altered. Nevertheless both strains had similar patterns of colonization and persistence in mice and significantly the aroA waaN mutant was equally as effective as the parent at protecting against challenge with wild-type S. Typhimurium. Furthermore, a SK100 strain was constructed expressing both tetanus toxin fragment C and the circumsporozoite protein of a malaria parasite. In marked contrast to its isogenic parent, the new attenuated strain induces significantly enhanced immune responses against the circumsporozoite protein. The waaN mutation enhances the ability of this strain to elicit immune responses towards guest antigens. This study provides important insights into the development of safe and effective multivalent Salmonella vaccines.


Assuntos
Vacinas Antimaláricas/imunologia , Vacinas contra Salmonella/imunologia , Salmonella typhimurium/genética , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Antiprotozoários/sangue , Proteínas de Bactérias/genética , Linhagem Celular , Contagem de Colônia Microbiana , Feminino , Lipopolissacarídeos/isolamento & purificação , Lipopolissacarídeos/toxicidade , Fígado/microbiologia , Macrófagos/microbiologia , Vacinas Antimaláricas/genética , Camundongos , Camundongos Endogâmicos BALB C , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Polissacarídeos Bacterianos/isolamento & purificação , Polissacarídeos Bacterianos/toxicidade , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Infecções por Salmonella/imunologia , Infecções por Salmonella/prevenção & controle , Vacinas contra Salmonella/genética , Salmonella typhimurium/imunologia , Salmonella typhimurium/patogenicidade , Baço/microbiologia , Toxina Tetânica/genética , Toxina Tetânica/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Vacinas Tíficas-Paratíficas/genética , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia
18.
Nat Med ; 13(9): 1035-41, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17704784

RESUMO

The success of immunization with irradiated sporozoites is unparalleled among the current vaccination approaches against malaria, but its mechanistic underpinnings have yet to be fully elucidated. Using a model mimicking natural infection by Plasmodium yoelii, we delineated early events governing the development of protective CD8(+) T-cell responses to the circumsporozoite protein. We demonstrate that dendritic cells in cutaneous lymph nodes prime the first cohort of CD8(+) T cells after an infectious mosquito bite. Ablation of these lymphoid sites greatly impairs subsequent development of protective immunity. Activated CD8(+) T cells then travel to systemic sites, including the liver, in a sphingosine-1-phosphate (S1P)-dependent fashion. These effector cells, however, no longer require bone marrow-derived antigen-presenting cells for protection; instead, they recognize antigen on parenchymal cells-presumably parasitized hepatocytes. Therefore, we report an unexpected dichotomy in the tissue restriction of host responses during the development and execution of protective immunity to Plasmodium.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Fígado/parasitologia , Linfonodos/imunologia , Malária/imunologia , Pele/imunologia , Animais , Antígenos de Protozoários/imunologia , Medula Óssea/imunologia , Humanos , Depleção Linfocítica , Camundongos , Plasmodium yoelii/imunologia , Esplenectomia
19.
J Leukoc Biol ; 81(4): 1102-10, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17200144

RESUMO

IL-4 has distinct effects on the differentiation and functional properties of CD8+ T cells. In vivo studies have shown that it is critical for the development of protective memory responses against tumors and infections by Leishmania and Plasmodium parasites. The intracellular signaling events mediated by IL-4/IL-4 receptor (IL-4R) interactions on CD4+ T cells have been studied extensively; however, the nature of IL-4-induced signaling on CD8+ T cells has not been characterized. Using naïve, activated, as well as differentiated CD8+ T cells, we show that IL-4 has a strong in vivo and in vitro antiapoptotic effect on activated and resting CD8+ T cells. We demonstrate that IL-4 induces the phosphorylation of the IL-4R, which is followed by the activation of at least two distinct intracellular signaling cascades: the Jak1/STAT6 and the insulin receptor substrate/PI-3K/protein kinase B pathways. We also found that IL-4 induces the Jak3-mediated phosphorylation and nuclear migration of STAT1, STAT3, and STAT5 in naïve, activated, as well as differentiated, IFN-gamma-producing CD8+ T cells. The induction of this broad signaling activity in CD8+ T cells coincides with a transcriptional activity of suppressors of cytokine signaling genes, which are decreased significantly in comparison with CD4+ T cells. To our knowledge, this report constitutes the first comprehensive analysis of the signaling events that shape CD8+ T cell responses to IL-4.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Interleucina-4/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Transdução de Sinais , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Animais , Apoptose , Linfócitos T CD8-Positivos/efeitos dos fármacos , Ativação Enzimática , Janus Quinases/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição STAT/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Transcrição Gênica
20.
Vaccine ; 25(14): 2567-74, 2007 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-16914237

RESUMO

A workshop on viral vectors for malaria vaccine development, organized by the PATH Malaria Vaccine Initiative, was held in Bethesda, MD on October 20, 2005. Recent advancements in viral-vectored malaria vaccine development and emerging vector technologies were presented and discussed. Classic viral vectors such as poxvirus, adenovirus and alphavirus vectors have been successfully used to deliver malaria antigens. Some of the vaccine candidates have demonstrated their potential in inducing malaria-specific immunity in animal models and human trials. In addition, emerging viral-vector technologies, such as measles virus (MV), vesicular stomatitis virus (VSV) and yellow fever (YF) virus, may also be useful for malaria vaccine development. Studies in animal models suggest that each viral vector is unique in its ability to induce humoral and/or cellular immune responses. Those studies have also revealed that optimization of Plasmodium genes for mammalian expression is an important aspect of vaccine design. Codon-optimization, surface-trafficking, de-glycosylation and removal of toxic domains can lead to improved immunogenicity. Understanding the vector's ability to induce an immune response and the expression of malaria antigens in mammalian cells will be critical in designing the next generation of viral-vectored malaria vaccines.


Assuntos
Vetores Genéticos/genética , Vacinas Antimaláricas/biossíntese , Vacinas Sintéticas/biossíntese , Vírus/genética , Adenoviridae/genética , Alphavirus/genética , Vacinas Antimaláricas/imunologia , Vírus do Sarampo/genética , Poxviridae/genética , Vacinas Sintéticas/imunologia , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Febre Amarela/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA