Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Infect Immun ; 89(11): e0016521, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34310889

RESUMO

Preerythrocytic vaccines prevent malaria by targeting parasites in the clinically silent sporozoite and liver stages and preventing progression to the virulent blood stages. The leading preerythrocytic vaccine, RTS,S/AS01E (Mosquirix), entered implementation programs in 2019 and targets the major sporozoite surface antigen, circumsporozoite protein (CSP). However, in phase III clinical trials, RTS,S conferred partial protection with limited durability, indicating a need to improve CSP-based vaccination. Previously, we identified highly expressed liver-stage proteins that could potentially be used in combination with CSP; they are referred to as preerythrocytic vaccine antigens (PEVAs). Here, we developed heterologous prime-boost CSP vaccination models to confer partial sterilizing immunity against Plasmodium yoelii (protein prime-adenovirus 5 [Ad5] boost) and Plasmodium berghei (DNA prime-Ad5 boost) in mice. When combined as individual antigens with P. yoelii CSP (PyCSP), three of eight P. yoelii PEVAs significantly enhanced sterile protection against sporozoite challenge, compared to PyCSP alone. Similar results were obtained when three P. berghei PEVAs and P. berghei CSP were combined in a single vaccine regimen. In general, PyCSP antibody responses were similar after CSP alone versus CSP plus PEVA vaccinations. Both P. yoelii and P. berghei CSP plus PEVA combination vaccines induced robust CD8+ T cell responses, including signature gamma interferon (IFN-γ) increases. In the P. berghei model system, IFN-γ responses were significantly higher in hepatic versus splenic CD8+ T cells. The addition of novel antigens may enhance the degree and duration of sterile protective immunity conferred by a human vaccine such as RTS,S.


Assuntos
Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/imunologia , Proteínas de Protozoários/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Interferon gama/biossíntese , Ativação Linfocitária , Malária/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Vacinação
2.
Crit Rev Immunol ; 40(4): 311-323, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33426820

RESUMO

The physicochemical properties of an antigen (Ag) influence the type, specificity, as well as duration of emerging immune responses. Like immune responses arising to nominal protein Ags, reactivities to protozoan parasites, Plasmodium falciparum and P. berghei, the causative agents of human and mouse malaria, respectively, are shaped by the form of the parasite. While repeated natural exposures to infectious Plasmodium sporozoites (spzs) typically induce malaria, immunizations with radiation or genetically attenuated forms of Plasmodium spzs induce sterile and durable protective immunity. The immune mechanisms that are responsible for these diametrically opposite outcomes are still not well understood. It has been observed that infectious spzs engage in mechanisms that evade immune recognition and thus prevent protective immune responses from occurring. The responses that develop are characteristic of anti-disease immunity; acquisition of protective immunity against infection is a prolonged process, and it decays once exposure to the parasite ceases. In contrast, repeated exposures to attenuated Plasmodium spzs induce antibodies and CD4 T cells directed primarily to the spz surface Ags and effector and memory CD8 T cells that localize in the liver and are specific for Plasmodium liver-stage Ags. Understanding the precise mechanisms, including early interactions between the spzs and Ag-presenting cells that lead to the manner of Ag processing and presentation, are of key importance as such information would substantially contribute to the successful development of malaria vaccine.


Assuntos
Vacinas Antimaláricas , Malária , Plasmodium , Animais , Antígenos de Protozoários , Linfócitos T CD8-Positivos , Humanos , Malária/prevenção & controle , Camundongos , Esporozoítos
3.
Parasite Immunol ; 41(7): e12622, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30854655

RESUMO

Immunization with radiation-attenuated Plasmodium sporozoites (RAS) induces sterile and long-lasting protective immunity. Although intravenous (IV) route of RAS immunization is reported to induce superior immunity compared to intradermal (ID) injection, its role in the maintenance of sterile immunity is yet to be understood. We investigated whether the route of homologous sporozoite challenge of Plasmodium berghei (Pb) RAS-immunized mice would influence the longevity of protection. C57BL/6 mice immunized with Pb-RAS by IV were 100% protected upon primary IV/ID sporozoite challenge. In contrast, ID immunization resulted in 80% protection, regardless of primary challenge route. Interestingly, the route of primary challenge was found to bring difference in the maintenance of sterile protection. While IV Pb RAS-immunized mice remained protected at all challenges regardless of the route of primary challenge, ID Pb-RAS-immunized mice receiving ID primary challenge became parasitaemic upon secondary IV challenge. Significantly, primary IV challenge of Pb RAS ID-immunized mice resulted in 80% and 50% survival at secondary and tertiary challenges, respectively. According to phenotypically diverse liver CD8+ T cells, the percentages and the numbers of both CD8+ T effector memory and resident memory cells were significantly higher in IV than in ID Pb RAS-immunized mice. IFN-γ-producing CD8+ T cells specific to Pb TRAP130 and MIP-4-Kb-17 were also found significantly higher in IV mice than in ID mice. The enhanced T-cell generation and the longevity of protection appear to be dependent on the parasite load during challenge when infection is tolerated under suboptimal CD8+ T-cell response.


Assuntos
Memória Imunológica , Fígado/imunologia , Malária/imunologia , Plasmodium berghei/imunologia , Esporozoítos/imunologia , Administração Intravenosa , Animais , Antígenos de Protozoários/administração & dosagem , Linfócitos T CD8-Positivos/imunologia , Feminino , Imunização , Injeções Intradérmicas , Fígado/parasitologia , Malária/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Carga Parasitária , Esporozoítos/efeitos da radiação
4.
Front Immunol ; 9: 91, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29434602

RESUMO

We recently identified novel Plasmodium berghei (Pb) liver stage (LS) genes that as DNA vaccines significantly reduce Pb LS parasite burden (LPB) in C57Bl/6 (B6) mice through a mechanism mediated, in part, by CD8 T cells. In this study, we sought to determine fine antigen (Ag) specificities of CD8 T cells that target LS malaria parasites. Guided by algorithms for predicting MHC class I-restricted epitopes, we ranked sequences of 32 Pb LS Ags and selected ~400 peptides restricted by mouse H-2Kb and H-2Db alleles for analysis in the high-throughput method of caged MHC class I-tetramer technology. We identified a 9-mer H-2Kb restricted CD8 T cell epitope, Kb-17, which specifically recognized and activated CD8 T cell responses in B6 mice immunized with Pb radiation-attenuated sporozoites (RAS) and challenged with infectious sporozoites (spz). The Kb-17 peptide is derived from the recently described novel protective Pb LS Ag, PBANKA_1031000 (MIF4G-like protein). Notably, immunization with the Kb-17 epitope delivered in the form of a minigene in the adenovirus serotype 5 vector reduced LPB in mice infected with spz. On the basis of our results, Kb-17 peptide was available for CD8 T cell activation and recall following immunization with Pb RAS and challenge with infectious spz. The identification of a novel MHC class I-restricted epitope from the protective Pb LS Ag, MIF4G-like protein, is crucial for advancing our understanding of immune responses to Plasmodium and by extension, toward vaccine development against malaria.


Assuntos
Antígenos de Protozoários/imunologia , Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Fígado/parasitologia , Malária/imunologia , Malária/parasitologia , Plasmodium berghei/imunologia , Animais , Modelos Animais de Doenças , Feminino , Imunização , Interferon gama/metabolismo , Fígado/imunologia , Malária/metabolismo , Camundongos , Plasmodium berghei/crescimento & desenvolvimento
5.
PLoS One ; 11(7): e0159449, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27434123

RESUMO

Malaria vaccine development has been hampered by the limited availability of antigens identified through conventional discovery approaches, and improvements are needed to enhance the efficacy of the leading vaccine candidate RTS,S that targets the circumsporozoite protein (CSP) of the infective sporozoite. Here we report a transcriptome-based approach to identify novel pre-erythrocytic vaccine antigens that could potentially be used in combination with CSP. We hypothesized that stage-specific upregulated genes would enrich for protective vaccine targets, and used tiling microarray to identify P. falciparum genes transcribed at higher levels during liver stage versus sporozoite or blood stages of development. We prepared DNA vaccines for 21 genes using the predicted orthologues in P. yoelii and P. berghei and tested their efficacy using different delivery methods against pre-erythrocytic malaria in rodent models. In our primary screen using P. yoelii in BALB/c mice, we found that 16 antigens significantly reduced liver stage parasite burden. In our confirmatory screen using P. berghei in C57Bl/6 mice, we confirmed 6 antigens that were protective in both models. Two antigens, when combined with CSP, provided significantly greater protection than CSP alone in both models. Based on the observations reported here, transcriptional patterns of Plasmodium genes can be useful in identifying novel pre-erythrocytic antigens that induce protective immunity alone or in combination with CSP.


Assuntos
Anticorpos Antiprotozoários/imunologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/tratamento farmacológico , Proteínas de Protozoários/imunologia , Animais , Anticorpos Antiprotozoários/uso terapêutico , Antígenos de Protozoários/imunologia , Feminino , Humanos , Vacinas Antimaláricas/genética , Vacinas Antimaláricas/uso terapêutico , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium falciparum/imunologia , Plasmodium falciparum/patogenicidade , Plasmodium yoelii/imunologia , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Vacinas de DNA/uso terapêutico
6.
Eur J Immunol ; 46(4): 885-96, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26703789

RESUMO

MHC class I dependent CD8(+) T cells are essential for protection induced by radiation-attenuated Plasmodium sporozoites (RAS) in murine malaria models. Apart from the mechanism of activation of CD8(+) T cells specific for the circumsporozoite protein, the major sporozoite antigen (Ag), CD8(+) T cells specific for other exoerythrocytic Ags that have been shown to mediate protection have not been thoroughly investigated. Specifically, mechanisms of processing and presentation of exoerythrocytic Ags, which includes liver stage (LS) Ags, remain poorly understood. We hypothesize that as exogenous proteins, LS Ags are processed by mechanisms involving either the TAP-dependent phagosomal-to-cytosol or TAP-independent vacuolar pathway of cross-presentation. We used TAP-deficient mice to investigate whether LS Ag mediated induction of naïve CD8(+) T cells and their recall during sporozoite challenge occur by the TAP-dependent or TAP-independent pathways. On the basis of functional attributes, CD8(+) T cells were activated via the TAP-independent pathway during immunizations with Plasmodium berghei RAS; however, IFN-γ(+) CD8(+) T cells previously induced by P. berghei RAS in TAP-deficient mice failed to be recalled against sporozoite challenge and the mice became parasitemic. On the basis of these observations, we propose that TAP-associated Ag processing is indispensable for sterile protection induced with P. berghei RAS.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Apresentação de Antígeno/imunologia , Antígenos de Protozoários/imunologia , Linfócitos T CD8-Positivos/imunologia , Plasmodium berghei/imunologia , Esporozoítos/imunologia , Transportadores de Cassetes de Ligação de ATP/imunologia , Animais , Linfócitos T CD8-Positivos/citologia , Diferenciação Celular/imunologia , Feminino , Antígenos de Histocompatibilidade Classe I/imunologia , Imunização , Memória Imunológica/imunologia , Interferon gama/biossíntese , Interferon gama/imunologia , Fígado/citologia , Fígado/imunologia , Fígado/parasitologia , Malária/imunologia , Malária/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasmodium berghei/efeitos da radiação , Proteínas de Protozoários/imunologia , Esporozoítos/efeitos da radiação
7.
Methods Mol Biol ; 1325: 39-48, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26450377

RESUMO

Murine models of malaria, such as Plasmodium berghei (Pb) and Plasmodium yoelii (Py), have been used for decades to identify correlates of protection associated with immunization using radiation-attenuated sporozoites (RAS). To date, RAS is the only known immunization regimen to consistently deliver 100 % sterilizing immunity and is considered the "gold standard" of protection against malaria. The ability to isolate lymphocytes directly from the liver of immune mice has facilitated the identification of correlates of protection at the site of infection. Liver CD8 T cells have been identified as a key factor in mediating protection against challenge with infectious Plasmodium sporozoites. Liver CD3 + CD8 T cells can further be divided into subsets based on the expression of specific surface molecules and the increase of CD8 effector memory (TEM) cells (identified by the phenotype CD44(+)CD62L(-)) has been shown to mediate protection by releasing of IFN-γ while CD8 central memory (TCM) cells (CD44(+)CD62L(+)) are important for maintaining long-term protection.Identification of multiple CD8 T cell subsets present in the liver relies on the ability to detect multiple surface markers simultaneously. Polychromatic flow cytometry affords the user with the ability to distinguish multiple lymphocyte populations as well as subsets defined within each population. In this chapter we present a basic 9-color surface staining panel that can be used to identify CD8 TEM, CD8 TCM, short-lived effector cells (SLECs), and memory precursor cells (MPECs) as well as identify those cells which have recently undergone degranulation (surface expression of CD107a). This panel has been designed to allow for the addition of intracellular staining for IFN-γ on other available channels (such as PE) as is discussed in another chapter for analysis of functional CD8 T cell responses.


Assuntos
Vacinas Antimaláricas/imunologia , Malária/imunologia , Biologia Molecular/métodos , Vacinas Atenuadas/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Eritrócitos/imunologia , Humanos , Malária/parasitologia , Malária/prevenção & controle , Vacinas Antimaláricas/uso terapêutico , Camundongos , Plasmodium berghei/imunologia , Plasmodium berghei/patogenicidade , Plasmodium yoelii/imunologia , Plasmodium yoelii/patogenicidade , Subpopulações de Linfócitos T/imunologia , Vacinas Atenuadas/uso terapêutico
8.
Methods Mol Biol ; 1325: 81-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26450381

RESUMO

Direct detection and quantification of liver-stage Plasmodium parasites became possible with the development of quantitative real-time PCR (qPCR). Here we describe the measurement of parasite burden in the livers of mice infected with the rodent malaria species, Plasmodium berghei and Plasmodium yoelii. This method is based on detection of expression of parasite ribosomal 18S RNA and can serve as an endpoint to accurately evaluate the efficacy of vaccines targeting the preerythrocytic stages of malaria. This approach is fast and highly reproducible and allows quantification of liver-stage parasite burden in different mouse strains and different Plasmodium species after infection with a range of sporozoite challenge doses.


Assuntos
Malária/parasitologia , Plasmodium berghei/isolamento & purificação , Plasmodium yoelii/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real/métodos , Animais , Humanos , Fígado/parasitologia , Fígado/patologia , Malária/genética , Malária/patologia , Camundongos , Plasmodium berghei/genética , Plasmodium berghei/patogenicidade , Plasmodium yoelii/genética , Plasmodium yoelii/patogenicidade , RNA Ribossômico 18S/genética
10.
Front Microbiol ; 6: 482, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26074888

RESUMO

Plasmodium falciparum malaria remains one of the most serious health problems globally. Immunization with attenuated parasites elicits multiple cellular effector mechanisms capable of eliminating Plasmodium liver stages. However, malaria liver stage (LS) immunity is complex and the mechanisms effector T cells use to locate the few infected hepatocytes in the large liver in order to kill the intracellular LS parasites remain a mystery to date. Here, we review our current knowledge on the behavior of CD8 effector T cells in the hepatic microvasculature, in malaria and other hepatic infections. Taking into account the unique immunological and lymphogenic properties of the liver, we discuss whether classical granule-mediated cytotoxicity might eliminate infected hepatocytes via direct cell contact or whether cytokines might operate without cell-cell contact and kill Plasmodium LSs at a distance. A thorough understanding of the cellular effector mechanisms that lead to parasite death hence sterile protection is a prerequisite for the development of a successful malaria vaccine to protect the 40% of the world's population currently at risk of Plasmodium infection.

11.
Vaccine ; 32(49): 6683-91, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-24950358

RESUMO

In an attempt to improve the efficacy of the candidate malaria vaccine RTS,S/AS02, two studies were conducted in 1999 in healthy volunteers of RTS,S/AS02 in combination with recombinant Plasmodium falciparum thrombospondin-related anonymous protein (TRAP). In a Phase 1 safety and immunogenicity study, volunteers were randomized to receive TRAP/AS02 (N=10), RTS,S/AS02 (N=10), or RTS,S+TRAP/AS02 (N=20) at 0, 1 and 6-months. In a Phase 2 challenge study, subjects were randomized to receive either RTS,S+TRAP/AS02 (N=25) or TRAP/AS02 (N=10) at 0 and 1-month, or to a challenge control group (N=8). In both studies, the combination vaccine had an acceptable safety profile and was acceptably tolerated. Antigen-specific antibodies, lymphoproliferative responses, and IFN-γ production by ELISPOT assay elicited with the combination vaccine were qualitatively similar to those generated by the single component vaccines. However, post-dose 2 anti-CS antibodies in the RTS,S+TRAP/AS02 vaccine recipients were lower than in the RTS,S/AS02 vaccine recipients. After challenge, 10 of 11 RTS,S+TRAP/AS02 vaccinees, 5 of 5 TRAP/AS02 vaccinees, and 8 of 8 infectivity controls developed parasitemia, with median pre-patent periods of 13.0, 11.0, and 12.0 days, respectively. The absence of any prevention or delay of parasitemia by TRAP/AS02 suggests no apparent added value of TRAP/AS02 as a candidate vaccine. The absence of significant protection or delay of parasitemia in the 11 RTS,S+TRAP/AS02 vaccine recipients contrasts with previous 2 dose studies of RTS,S/AS02. The small sample size did not permit identifying statistically significant differences between the study arms. However, we speculate, within the constraints of the challenge study, that the presence of the TRAP antigen may have interfered with the vaccine efficacy previously observed with this regimen of RTS,S/AS02, and that any future TRAP-based vaccines should consider employing alternative vaccine platforms.


Assuntos
Lipídeo A/análogos & derivados , Vacinas Antimaláricas/efeitos adversos , Vacinas Antimaláricas/imunologia , Malária Falciparum/prevenção & controle , Plasmodium falciparum/imunologia , Saponinas/efeitos adversos , Adolescente , Adulto , Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/imunologia , Proliferação de Células , Combinação de Medicamentos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/epidemiologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , ELISPOT , Feminino , Humanos , Interferon gama/metabolismo , Leucócitos Mononucleares/imunologia , Lipídeo A/administração & dosagem , Lipídeo A/efeitos adversos , Vacinas Antimaláricas/administração & dosagem , Masculino , Pessoa de Meia-Idade , Parasitemia/prevenção & controle , Proteínas de Protozoários/imunologia , Saponinas/administração & dosagem , Resultado do Tratamento , Vacinação/efeitos adversos , Vacinação/métodos , Adulto Jovem
12.
Immunol Lett ; 161(2): 189-95, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24709142

RESUMO

Immunologic memory is one of the cardinal features of antigen-specific immune responses, and the persistence of memory cells contributes to prophylactic immunizations against infectious agents. Adequately maintained memory T and B cell pools assure a fast, effective and specific response against re-infections. However, many aspects of immunologic memory are still poorly understood, particularly immunologic memory inducible by parasites, for example, Plasmodium spp., the causative agents of malaria. For example, memory responses to Plasmodium antigens amongst residents of malaria endemic areas appear to be either inadequately developed or maintained, because persons who survive episodes of childhood malaria remain vulnerable to intermittent malaria infections. By contrast, multiple exposures of humans and laboratory rodents to radiation-attenuated Plasmodium sporozoites (γ-spz) induce sterile and long-lasting protection against experimental sporozoite challenge. Multifactorial immune mechanisms maintain this protracted and sterile protection. While the presence of memory CD4 T cell subsets has been associated with lasting protection in humans exposed to multiple bites from Anopheles mosquitoes infected with attenuated Plasmodium falciparum, memory CD8 T cells maintain protection induced with Plasmodium yoelii and Plasmodium berghei γ-spz in murine models. In this review, we discuss our observations that show memory CD8 T cells specific for antigens expressed by P. berghei liver stage parasites as an indispensable component for the maintenance of protracted protective immunity against experimental malaria infection; moreover, the provision of an Ag-depot assures a quick recall of memory T cells as IFN-γ-producing effector CD8 T cells and IL-4- producing CD4 T cells that collaborate with B cells for an effective antibody response.


Assuntos
Memória Imunológica , Malária/imunologia , Malária/prevenção & controle , Plasmodium/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Antígenos de Protozoários/imunologia , Citocinas/metabolismo , Humanos , Fígado/imunologia , Fígado/parasitologia , Malária/metabolismo , Malária/parasitologia , Parasitemia/imunologia , Parasitemia/metabolismo , Esporozoítos/imunologia , Subpopulações de Linfócitos T/metabolismo
13.
Vaccine ; 31(43): 4975-83, 2013 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-24029408

RESUMO

BACKGROUND: Immunization with genetically engineered, attenuated malaria parasites (GAP) that arrest during liver infection confers sterile protection in mouse malaria models. A first generation Plasmodium falciparum GAP (Pf p52(-)/p36(-) GAP) was previously generated by deletion of two pre-erythrocytic stage-expressed genes (P52 and P36) in the NF54 strain. METHODS: A first-in-human, proof-of-concept, safety and immunogenicity clinical trial in six human volunteers was conducted. Exposure consisted of delivery of Pf p52(-)/p36(-) GAP sporozoites via infected Anopheles mosquito bite with a five-bite/volunteer exposure followed by an approximately 200-bite exposure/volunteer one month later. RESULTS: The exposures were well tolerated with mild to moderate local and systemic reactions. All volunteers remained blood stage negative after low dose exposure. Five volunteers remained blood stage negative after high dose exposure. One volunteer developed peripheral parasitemia twelve days after high dose exposure. Together the findings indicate that Pf p52(-)/p36(-) GAP was severely but not completely attenuated. All six volunteers developed antibodies to CSP. Furthermore, IFN-γ responses to whole sporozoites and multiple antigens were elicited in 5 of 6 volunteers, with both CD4 and CD8 cell cytokine production detected. CONCLUSION: Severe attenuation and favorable immune responses following administration of a first generation Pf p52(-)/p36(-) GAP suggests that further development of live-attenuated strains using genetic engineering should be pursued.


Assuntos
Anopheles/parasitologia , Imunização/métodos , Vacinas Antimaláricas/imunologia , Malária/prevenção & controle , Plasmodium falciparum/imunologia , Esporozoítos/imunologia , Adolescente , Adulto , Animais , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , Feminino , Deleção de Genes , Genes de Protozoários , Voluntários Saudáveis , Humanos , Imunização/efeitos adversos , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/efeitos adversos , Vacinas Antimaláricas/genética , Masculino , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidade , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Adulto Jovem
14.
AIDS ; 27(10): 1674-7, 2013 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-23907270

RESUMO

HIV and malaria overlap geographically, but the full impact of different antiretrovirals on malaria remains poorly understood. We examined the antimalarial activity of the HIV protease inhibitors lopinavir and saquinavir and the non-nucleoside reverse transcriptase inhibitor nevirapine on Plasmodium falciparum liver stages. Our results demonstrate that the HIV PI lopinavir inhibits liver stage parasites at clinically relevant concentrations, that is, at drug levels achieved in HIV-infected patients on standard dosing regimens. Because drugs that inhibit liver stages target parasites when they are present in lower numbers, these results might have implications for eradication efforts.


Assuntos
Inibidores da Protease de HIV/farmacologia , Fígado/parasitologia , Plasmodium falciparum/efeitos dos fármacos , Inibidores da Transcriptase Reversa/farmacologia , Infecções por HIV/complicações , Infecções por HIV/tratamento farmacológico , Inibidores da Protease de HIV/uso terapêutico , Humanos , Técnicas In Vitro , Fígado/efeitos dos fármacos , Lopinavir/farmacologia , Lopinavir/uso terapêutico , Malária Falciparum/complicações , Nevirapina/farmacologia , Nevirapina/uso terapêutico , Inibidores da Transcriptase Reversa/uso terapêutico , Saquinavir/farmacologia , Saquinavir/uso terapêutico
15.
J Immunol ; 190(10): 5128-41, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23589611

RESUMO

Ag-specific memory T cell responses elicited by infections or vaccinations are inextricably linked to long-lasting protective immunity. Studies of protective immunity among residents of malaria endemic areas indicate that memory responses to Plasmodium Ags are not adequately developed or maintained, as people who survive episodes of childhood malaria are still vulnerable to either persistent or intermittent malaria infections. In contrast, multiple exposures to radiation-attenuated Plasmodium berghei sporozoites (Pb γ-spz) induce long-lasting protective immunity to experimental sporozoite challenge. We previously demonstrated that sterile protection induced by Pb γ-spz is MHC class I-dependent and CD8 T cells are the key effectors. IFN-γ(+) CD8 T cells that arise in Pb γ-spz-immunized B6 mice are found predominantly in the liver and are sensitive to levels of liver-stage Ag depot and they express CD44(hi)CD62L(lo) markers indicative of effector/effector memory phenotype. The developmentally related central memory CD8 T (TCM) cells express elevated levels of CD122 (IL-15Rß), which suggests that CD8 TCM cells depend on IL-15 for maintenance. Using IL-15-deficient mice, we demonstrate in this study that although protective immunity is inducible in these mice, protection is short-lived, mainly owing to the inability of CD8 TCM cells to survive in the IL-15-deficient milieu. We present a hypothesis consistent with a model whereby intrahepatic CD8 TCM cells, being maintained by IL-15-mediated survival and basal proliferation, are conscripted into the CD8 effector/effector memory T cell pool during subsequent infections.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Interleucina-15/imunologia , Malária/imunologia , Plasmodium berghei/imunologia , Animais , Feminino , Imunização , Memória Imunológica , Interferon gama/imunologia , Interleucina-15/deficiência , Interleucina-15/genética , Fígado/imunologia , Fígado/parasitologia , Vacinas Antimaláricas/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasmodium berghei/efeitos da radiação , Esporozoítos/imunologia
16.
Methods Mol Biol ; 923: 385-400, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22990793

RESUMO

There is an important role for in vitro assays to better understand the initial steps of malaria infection. In this section, we describe both microscopy-based and flow cytometry-based sporozoite invasion, migration and development assays with the rodent malaria parasites, Plasmodium berghei and Plasmodium yoelii, and the human malaria parasite, Plasmodium falciparum.


Assuntos
Movimento Celular , Citometria de Fluxo/métodos , Hepatócitos/parasitologia , Microscopia/métodos , Plasmodium/crescimento & desenvolvimento , Esporozoítos/metabolismo , Animais , Anopheles/parasitologia , Técnicas de Cultura de Células , Linhagem Celular , Células Hep G2 , Hepatócitos/patologia , Humanos , Malária/parasitologia , Camundongos
17.
Front Immunol ; 3: 370, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23233854

RESUMO

Immunologic memory induced by pathogenic agents or vaccinations is inextricably linked to long-lasting protection. Adequately maintained memory T and B cell pools assure a fast, effective, and specific response against re-infections. Studies of immune responses amongst residents of malaria endemic areas suggest that memory responses to Plasmodia antigens appear to be neither adequately developed nor maintained, because persons who survive episodes of childhood malaria remain vulnerable to persistent or intermittent malaria infections. By contrast, multiple exposures of humans and laboratory rodents to radiation-attenuated Plasmodia sporozoites (γ-spz) induces sterile and long-lasting protection against experimental sporozoite challenge. Protection is associated with MHC-class I-dependent CD8 T cells, the key effectors against pre-erythrocytic stage infection. We have adopted the P. berghei γ-spz mouse model to study memory CD8 T cells that are specific for antigens expressed by Pb liver-stage (LS) parasites and are found predominantly in the liver. On the basis of phenotypic and functional characteristics, we have demonstrated that liver CD8 T cells form two subsets: CD44(hi)CD62L(lo)KLRG-1(+)CD107(+)CD127(-)CD122(lo)CD8 T effector/effector memory (T(E/EM)) cells that are the dominant IFN-γ producers and CD44(hi)CD62L(hi)KLRG-1(-)CD107(-)CD127(+)CD122(hi)CD8 T central memory (T(CM)) cells. In this review, we discuss our observations concerning the role of CD8 T(E/EM) and CD8 T(CM) cells in the maintenance of protracted protective immunity against experimental malaria infection. Finally, we present a hypothesis consistent with a model whereby intrahepatic CD8 T(CM) cells, that are maintained in part by LS-Ag depot and by IL-15-mediated survival and homeostatic proliferation, form a reservoir of cells ready for conscription to CD8 T(E/EM) cells needed to prevent re-infections.

18.
J Infect Dis ; 206(11): 1706-14, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23125449

RESUMO

BACKGROUND: Although nonnucleoside reverse transcriptase inhibitors (NNRTIs) are usually part of first-line treatment regimens for human immunodeficiency virus (HIV), their activity on Plasmodium liver stages remains unexplored. Additionally, trimethoprim-sulfamethoxazole (TMP-SMX), used for opportunistic infection prophylaxis in HIV-exposed infants and HIV-infected patients, reduces clinical episodes of malaria; however, TMP-SMX effect on Plasmodium liver stages requires further study. METHODS: We characterized NNRTI and TMP-SMX effects on Plasmodium liver stages in vivo using Plasmodium yoelii. On the basis of these results, we conducted in vitro studies assessing TMP-SMX effects on the rodent parasites P. yoelii and Plasmodium berghei and on the human malaria parasite Plasmodium falciparum. RESULTS: Our data showed NNRTI treatment modestly reduced P. yoelii liver stage parasite burden and minimally extended prepatent period. TMP-SMX administration significantly reduced liver stage parasite burden, preventing development of patent parasitemia in vivo. TMP-SMX inhibited development of rodent and P. falciparum liver stage parasites in vitro. CONCLUSIONS: NNRTIs modestly affect liver stage Plasmodium parasites, whereas TMP-SMX prevents patent parasitemia. Because drugs that inhibit liver stages target parasites when they are present in lower numbers, these results may have implications for eradication efforts. Understanding HIV drug effects on Plasmodium liver stages will aid in optimizing treatment regimens for HIV-exposed and HIV-infected infected patients in malaria-endemic areas.


Assuntos
Fígado/parasitologia , Malária/tratamento farmacológico , Malária/parasitologia , Plasmodium/efeitos dos fármacos , Inibidores da Transcriptase Reversa/farmacologia , Combinação Trimetoprima e Sulfametoxazol/farmacologia , Animais , Antimaláricos/administração & dosagem , Antimaláricos/farmacologia , Feminino , Humanos , Camundongos , Parasitemia/tratamento farmacológico , Inibidores da Transcriptase Reversa/administração & dosagem , Especificidade da Espécie , Combinação Trimetoprima e Sulfametoxazol/administração & dosagem
19.
Vaccine ; 29(48): 8847-54, 2011 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-21983360

RESUMO

In a Phase 2a trial of the RTS,S/AS vaccine, we described significant association between protection against infection and vaccine-induced CD4 T cells. To determine whether processing of the circumsporozoite protein as a component of the RTS,S particulate antigen yields the same HLA-DR-restricted epitopes as those recognized by CD4 T cells from donors immunized by exposure to attenuated or infectious sporozoites we mapped the specificities of the RTS,S primed CD4 T cells by measuring IFN-γ cultured Elispot responses to pairs of overlapping 15 a.a. peptides that span the protein's C-terminus. Peptide pairs representing the previously described TH2R, T* and CS.T3 epitopes, were immunoprevalent and immunodominant. There was no response to the peptides corresponding to the human thrombospondin homology region. Responses to the CD4 T cell epitopes were restricted by multiple HLA-DR haplotypes. Of note, HLA-DR4 and HLA-DR11 restricted epitopes in the T* region and in the location on the CS protein defined by peptide pair 4, respectively. We conclude that processing of the CS protein derived from the RTS,S antigen leads to the generation of HLA-DR-restricted epitopes that are similar to those identified previously using CD4 T cells from subjects immunized with and protected by attenuated sporozoites or exposed to infectious sporozoites. This may in part account for the protective efficacy of the RTS,S/AS vaccine.


Assuntos
Apresentação de Antígeno , Linfócitos T CD4-Positivos/imunologia , Vacinas Antimaláricas/imunologia , Proteínas de Protozoários/imunologia , Especificidade do Receptor de Antígeno de Linfócitos T , Sequência de Aminoácidos , Antígenos de Protozoários/imunologia , Células Cultivadas , Epitopos de Linfócito T/imunologia , Subtipos Sorológicos de HLA-DR/imunologia , Antígeno HLA-DR4/imunologia , Humanos , Imunização Secundária , Interferon gama/imunologia , Leucócitos Mononucleares/imunologia , Vacinas Antimaláricas/administração & dosagem , Dados de Sequência Molecular , Plasmodium falciparum/imunologia , Ensaios Clínicos Controlados Aleatórios como Assunto , Trombospondina 1/imunologia
20.
PLoS One ; 6(7): e20775, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21779319

RESUMO

A phase 2a RTS,S/AS malaria vaccine trial, conducted previously at the Walter Reed Army Institute of Research, conferred sterile immunity against a primary challenge with infectious sporozoites in 40% of the 80 subjects enrolled in the study. The frequency of Plasmodium falciparum circumsporozoite protein (CSP)-specific CD4(+) T cells was significantly higher in protected subjects as compared to non-protected subjects. Intrigued by these unique vaccine-related correlates of protection, in the present study we asked whether RTS,S also induced effector/effector memory (T(E/EM)) and/or central memory (T(CM)) CD4(+) T cells and whether one or both of these sub-populations is the primary source of cytokine production. We showed for the first time that PBMC from malaria-non-exposed RTS,S-immunized subjects contain both T(E/EM) and T(CM) cells that generate strong IL-2 responses following re-stimulation in vitro with CSP peptides. Moreover, both the frequencies and the total numbers of IL-2-producing CD4(+) T(E/EM) cells and of CD4(+) T(CM) cells from protected subjects were significantly higher than those from non-protected subjects. We also demonstrated for the first time that there is a strong association between the frequency of CSP peptide-reactive CD4(+) T cells producing IL-2 and the titers of CSP-specific antibodies in the same individual, suggesting that IL-2 may be acting as a growth factor for follicular Th cells and/or B cells. The frequencies of CSP peptide-reactive, TNF-α-producing CD4(+) T(E/EM) cells and of CD4(+) T(E/EM) cells secreting both IL-2 and TNF-α were also shown to be higher in protected vs. non-protected individuals. We have, therefore, demonstrated that in addition to TNF-α, IL-2 is also a significant contributing factor to RTS,S/AS vaccine induced immunity and that both T(E/EM) and T(CM) cells are major producers of IL-2.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Interleucina-2/metabolismo , Vacinas Antimaláricas/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Vacinas Antimaláricas/uso terapêutico , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...