Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 180
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Pathog ; 16(5): e1008181, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32453765

RESUMO

Plasmodium sporozoites are the infective stage of the malaria parasite. Though this is a bottleneck for the parasite, the quantitative dynamics of transmission, from mosquito inoculation of sporozoites to patent blood-stage infection in the mammalian host, are poorly understood. Here we utilize a rodent model to determine the probability of malaria infection after infectious mosquito bite, and consider the impact of mosquito parasite load, blood-meal acquisition, probe-time, and probe location, on infection probability. We found that infection likelihood correlates with mosquito sporozoite load and, to a lesser degree, the duration of probing, and is not dependent upon the mosquito's ability to find blood. The relationship between sporozoite load and infection probability is non-linear and can be described by a set of models that include a threshold, with mosquitoes harboring over 10,000 salivary gland sporozoites being significantly more likely to initiate a malaria infection. Overall, our data suggest that the small subset of highly infected mosquitoes may contribute disproportionally to malaria transmission in the field and that quantifying mosquito sporozoite loads could aid in predicting the force of infection in different transmission settings.


Assuntos
Malária/transmissão , Esporozoítos/metabolismo , Animais , Anopheles/metabolismo , Anopheles/parasitologia , Comportamento Alimentar , Feminino , Malária/parasitologia , Camundongos , Mosquitos Vetores/metabolismo , Plasmodium/metabolismo , Plasmodium/patogenicidade , Plasmodium yoelii/metabolismo , Plasmodium yoelii/patogenicidade , Glândulas Salivares/parasitologia , Esporozoítos/fisiologia
2.
PLoS Pathog ; 15(2): e1007599, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30811498

RESUMO

Plasmodium parasites invade and multiply inside red blood cells (RBC). Through a cycle of maturation, asexual replication, rupture and release of multiple infective merozoites, parasitised RBC (pRBC) can reach very high numbers in vivo, a process that correlates with disease severity in humans and experimental animals. Thus, controlling pRBC numbers can prevent or ameliorate malaria. In endemic regions, circulating parasite-specific antibodies associate with immunity to high parasitemia. Although in vitro assays reveal that protective antibodies could control pRBC via multiple mechanisms, in vivo assessment of antibody function remains challenging. Here, we employed two mouse models of antibody-mediated immunity to malaria, P. yoelii 17XNL and P. chabaudi chabaudi AS infection, to study infection-induced, parasite-specific antibody function in vivo. By tracking a single generation of pRBC, we tested the hypothesis that parasite-specific antibodies accelerate pRBC clearance. Though strongly protective against homologous re-challenge, parasite-specific IgG did not alter the rate of pRBC clearance, even in the presence of ongoing, systemic inflammation. Instead, antibodies prevented parasites progressing from one generation of RBC to the next. In vivo depletion studies using clodronate liposomes or cobra venom factor, suggested that optimal antibody function required splenic macrophages and dendritic cells, but not complement C3/C5-mediated killing. Finally, parasite-specific IgG bound poorly to the surface of pRBC, yet strongly to structures likely exposed by the rupture of mature schizonts. Thus, in our models of humoral immunity to malaria, infection-induced antibodies did not accelerate pRBC clearance, and instead co-operated with splenic phagocytes to block subsequent generations of pRBC.


Assuntos
Malária/imunologia , Malária/metabolismo , Plasmodium/crescimento & desenvolvimento , Animais , Anticorpos Antiprotozoários/metabolismo , Modelos Animais de Doenças , Eritrócitos/microbiologia , Eritrócitos/fisiologia , Humanos , Camundongos , Parasitos , Fagócitos , Plasmodium/metabolismo , Plasmodium/patogenicidade , Plasmodium chabaudi/imunologia , Plasmodium chabaudi/patogenicidade , Plasmodium yoelii/imunologia , Plasmodium yoelii/patogenicidade
3.
BMC Infect Dis ; 20(1): 266, 2020 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-32252652

RESUMO

BACKGROUND: Emerging data has suggested that Tregs, Th17, Th1 and Th2 are correlated with early immune mechanisms by controlling Plasmodium infection. Plasmodium infection appeared to impair the antigen presentation and maturation of DCs, leading to attenuation of specific cellular immune response ultimately. Hence, in this study, we aim to evaluate the relevance between DCs and Tregs/Th17 populations in the process and outcomes of infection with Plasmodium yoelii 17XL (P.y17XL). METHODS: DCs detection/analysis dynamically was performed by Tregs depletion or Th17 neutralization in P.y17XL infected BALB/c mice via flow cytometry. Then the levels of cytokines production were detected using enzyme-linked mmunosorbent assay (ELISA). RESULTS: Our results indicated that Tregs depletion or Th17 neutralization in BALB/c mice infected with P.y17XL significantly up-regulated the percentages of mDC and pDC, increased the expressions of major histocompatibility complex (MHC) class II, CD80, CD86 on DCs and the levels of IL-10/IL-12 secreted by DCs, indicating that abnormal amplification of Tregs or Th17 may damage the maturation and function of DCs during the early stage of malaria infection. Interestingly, we also found that the abnormal amplification of Th17, as well as Tregs, could inhibit the maturation of DCs. CONCLUSIONS: Tregs skewing or Th17 amplification during the early stage of malaria infection may inhibit the maturation and function of DCs by modifying the subsets of DCs, expressions of surface molecules on DCs and secretion mode of cytokines.


Assuntos
Células Dendríticas/imunologia , Malária/imunologia , Plasmodium yoelii/patogenicidade , Linfócitos T Reguladores/patologia , Células Th17/parasitologia , Animais , Citocinas/metabolismo , Células Dendríticas/parasitologia , Feminino , Interações Hospedeiro-Parasita , Imunidade Celular , Malária/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/parasitologia , Células Th1/imunologia , Células Th17/patologia
4.
PLoS Pathog ; 13(7): e1006447, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28704525

RESUMO

Identifying the genetic determinants of phenotypes that impact disease severity is of fundamental importance for the design of new interventions against malaria. Here we present a rapid genome-wide approach capable of identifying multiple genetic drivers of medically relevant phenotypes within malaria parasites via a single experiment at single gene or allele resolution. In a proof of principle study, we found that a previously undescribed single nucleotide polymorphism in the binding domain of the erythrocyte binding like protein (EBL) conferred a dramatic change in red blood cell invasion in mutant rodent malaria parasites Plasmodium yoelii. In the same experiment, we implicated merozoite surface protein 1 (MSP1) and other polymorphic proteins, as the major targets of strain-specific immunity. Using allelic replacement, we provide functional validation of the substitution in the EBL gene controlling the growth rate in the blood stages of the parasites.


Assuntos
Antígenos de Protozoários/genética , Malária/imunologia , Malária/parasitologia , Proteína 1 de Superfície de Merozoito/genética , Plasmodium yoelii/genética , Plasmodium yoelii/patogenicidade , Proteínas de Protozoários/genética , Receptores de Superfície Celular/genética , Antígenos de Protozoários/metabolismo , Eritrócitos/parasitologia , Interações Hospedeiro-Parasita , Humanos , Imunidade , Malária/genética , Proteína 1 de Superfície de Merozoito/metabolismo , Plasmodium yoelii/crescimento & desenvolvimento , Plasmodium yoelii/metabolismo , Polimorfismo de Nucleotídeo Único , Proteínas de Protozoários/metabolismo , Receptores de Superfície Celular/metabolismo , Virulência
5.
Cell Microbiol ; 20(5): e12821, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29316140

RESUMO

Anopheles mosquitoes transmit Plasmodium parasites of mammals, including the species that cause malaria in humans. Malaria pathology is caused by rapid multiplication of parasites in asexual intraerythrocytic cycles. Sexual stage parasites are also produced during the intraerythrocytic cycle and are ingested by the mosquito, initiating gametogenesis and subsequent sporogonic stage development. Here, we present a Plasmodium protein, termed microgamete surface protein (MiGS), which has an important role in male gametocyte osmiophilic body (MOB) formation and microgamete function. MiGS is expressed exclusively in male gametocytes and microgametes, in which MiGS localises to the MOB and microgamete surface. Targeted gene disruption of MiGS in a rodent malaria parasite Plasmodium yoelii 17XNL generated knockout parasites (ΔPyMiGS) that proliferate normally in erythrocytes and form male and female gametocytes. The number of MOB in male gametocyte cytoplasm is markedly reduced and the exflagellation of microgametes is impaired in ΔPyMiGS. In addition, anti-PyMiGS antibody severely blocked the parasite development in the Anopheles stephensi mosquito. MiGS might thus be a potential novel transmission-blocking vaccine target candidate.


Assuntos
Gametogênese/genética , Células Germinativas/crescimento & desenvolvimento , Malária/genética , Plasmodium yoelii/genética , Animais , Eritrócitos/parasitologia , Feminino , Células Germinativas/metabolismo , Humanos , Malária/parasitologia , Masculino , Proteínas de Membrana/genética , Plasmodium yoelii/patogenicidade , Roedores/parasitologia
6.
Artigo em Inglês | MEDLINE | ID: mdl-29784841

RESUMO

As a partner antimalarial for artemisinin drug-based combination therapy (ACT), piperaquine (PQ) can be metabolized into two major metabolites, including piperaquine N-oxide (M1) and piperaquine N,N-dioxide (M2). To better understand the antimalarial potency of PQ, the antimalarial activity of the PQ metabolites (M1 and M2) was studied in vitro (in Plasmodium falciparum strains Pf3D7 and PfDd2) and in vivo (in the murine species Plasmodium yoelii) in this study. The recrudescence and survival time of infected mice were also recorded after drug treatment. The pharmacokinetic profiles of PQ and its two metabolites (M1 and M2) were investigated in healthy subjects after oral doses of two widely used ACT regimens, i.e., dihydroartemisinin plus piperaquine phosphate (Duo-Cotecxin) and artemisinin plus piperaquine (Artequick). Remarkable antiplasmodial activity was found for PQ (50% growth-inhibitory concentration [IC50], 4.5 nM against Pf3D7 and 6.9 nM against PfDd2; 90% effective dose [ED90], 1.3 mg/kg of body weight), M1 (IC50, 25.5 nM against Pf3D7 and 38.7 nM against PfDd2; ED90, 1.3 mg/kg), and M2 (IC50, 31.2 nM against Pf3D7 and 33.8 nM against PfDd2; ED90, 2.9 mg/kg). Compared with PQ, M1 showed comparable efficacy in terms of recrudescence and survival time and M2 had relatively weaker antimalarial potency. PQ and its two metabolites displayed a long elimination half-life (∼11 days for PQ, ∼9 days for M1, and ∼4 days for M2), and they accumulated after repeated administrations. The contribution of the two PQ metabolites to the efficacy of piperaquine as a partner drug of ACT for the treatment of malaria should be considered for PQ dose optimization.


Assuntos
Antimaláricos/farmacocinética , Malária/tratamento farmacológico , Plasmodium falciparum/efeitos dos fármacos , Plasmodium yoelii/efeitos dos fármacos , Quinolinas/farmacocinética , Animais , Antimaláricos/sangue , Antimaláricos/farmacologia , Artemisininas/farmacologia , Biotransformação , Esquema de Medicação , Cálculos da Dosagem de Medicamento , Quimioterapia Combinada , Meia-Vida , Voluntários Saudáveis , Humanos , Malária/metabolismo , Malária/mortalidade , Malária/parasitologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Óxidos/sangue , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/patogenicidade , Plasmodium yoelii/crescimento & desenvolvimento , Plasmodium yoelii/patogenicidade , Quinolinas/sangue , Quinolinas/farmacologia , Ratos Wistar , Recidiva , Análise de Sobrevida , Adulto Jovem
7.
Bioconjug Chem ; 29(11): 3606-3613, 2018 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-30247899

RESUMO

The diagnosis and prognosis of the disease associated with lipid irregularity are areas of extreme significance. In this direction, fluoranthene based yellow fluorescent probes (FLUN-550, FLUN-552, FLUN-547) were designed and synthesized by conjugating the ethanolamine headgroup of the phospholipid phosphatidyl-ethanolamine present in biological membranes. Owing to unique photophysical properties and aqueous compatibility, these probes were successfully employed for staining lipid droplets (LDs) in preadipocytes and Leishmania donovani promastigotes. Furthermore, using the fluorescent probes FLUN-550 and FLUN-552 we successfully imaged and quantitatively detected the excess accumulation of lipids in a liver section of Plasmodium yoelii MDR infected mice (3- to 4-fold) and the tissue sections of third stage human cervical cancer patients (1.5- to 2-fold) compared to normal tissues. To the best of our knowledge, this is the first report of yellow fluorescent probes for imaging and quantitative detection of LDs in human cervical cancer tissues. These new yellow fluorescent lipid probes (FLUN-550 and FLUN-552) showed great potential for diagnosis of cervical cancer patients.


Assuntos
Corantes Fluorescentes/metabolismo , Gotículas Lipídicas/metabolismo , Fígado/metabolismo , Fígado/parasitologia , Plasmodium yoelii/patogenicidade , Neoplasias do Colo do Útero/metabolismo , Células 3T3-L1 , Animais , Teoria da Densidade Funcional , Feminino , Humanos , Leishmania donovani/metabolismo , Camundongos , Coloração e Rotulagem
8.
Int J Mol Sci ; 19(5)2018 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-29734652

RESUMO

Curcumin is an antimalarial compound easy to obtain and inexpensive, having shown little toxicity across a diverse population. However, the clinical use of this interesting polyphenol has been hampered by its poor oral absorption, extremely low aqueous solubility and rapid metabolism. In this study, we have used the anionic copolymer Eudragit® S100 to assemble liposomes incorporating curcumin and containing either hyaluronan (Eudragit-hyaluronan liposomes) or the water-soluble dextrin Nutriose® FM06 (Eudragit-nutriosomes). Upon oral administration of the rehydrated freeze-dried nanosystems administered at 25/75 mg curcumin·kg−1·day−1, only Eudragit-nutriosomes improved the in vivo antimalarial activity of curcumin in a dose-dependent manner, by enhancing the survival of all Plasmodium yoelii-infected mice up to 11/11 days, as compared to 6/7 days upon administration of an equal dose of the free compound. On the other hand, animals treated with curcumin incorporated in Eudragit-hyaluronan liposomes did not live longer than the controls, a result consistent with the lower stability of this formulation after reconstitution. Polymer-lipid nanovesicles hold promise for their development into systems for the oral delivery of curcumin-based antimalarial therapies.


Assuntos
Curcumina/administração & dosagem , Sistemas de Liberação de Medicamentos , Lipossomos/administração & dosagem , Malária/tratamento farmacológico , Administração Oral , Animais , Antimaláricos/administração & dosagem , Antimaláricos/química , Curcumina/química , Humanos , Lipossomos/química , Malária/parasitologia , Camundongos , Nanopartículas/administração & dosagem , Nanopartículas/química , Plasmodium yoelii/efeitos dos fármacos , Plasmodium yoelii/patogenicidade
9.
Infect Immun ; 85(9)2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28630062

RESUMO

Repeated stimulation of T cells that occurs in the context of chronic infection results in progressively reduced responsiveness of T cells to pathogen-derived antigens. This phenotype, known as T cell exhaustion, occurs during chronic infections caused by a variety of pathogens, from persistent viruses to parasites. Unlike the memory cells that typically form after successful pathogen clearance following an acute infection, exhausted T cells secrete lower levels of effector cytokines, proliferate less in response to cognate antigen, and upregulate cell surface inhibitory molecules such as PD-1 and LAG-3. The molecular events that lead to the induction of this phenotype have, however, not been fully characterized. In T cells, members of the NFAT family of transcription factors not only are responsible for the expression of many activation-induced genes but also are crucial for the induction of transcriptional programs that inhibit T cell activation and maintain tolerance. Here we show that NFAT1-deficient CD4+ T cells maintain higher proliferative capacity and expression of effector cytokines following Plasmodium yoelii infection and are therefore more resistant to P. yoelii-induced exhaustion than their wild-type counterparts. Consequently, gene expression microarray analysis of CD4+ T cells following P. yoelii-induced exhaustion shows upregulation of effector T cell-associated genes in the absence of NFAT1 compared with wild-type exhausted T cells. Furthermore, adoptive transfer of NFAT1-deficient CD4+ T cells into mice infected with P. yoelii results in increased production of antibodies to cognate antigen. Our results support the idea that NFAT1 is necessary to fully suppress effector responses during Plasmodium-induced CD4+ T cell exhaustion.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Interações Hospedeiro-Patógeno , Tolerância Imunológica , Malária/patologia , Fatores de Transcrição NFATC/metabolismo , Plasmodium yoelii/patogenicidade , Animais , Proliferação de Células , Citocinas/metabolismo , Ativação Linfocitária , Malária/imunologia , Camundongos Endogâmicos C57BL
10.
PLoS Pathog ; 11(2): e1004628, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25658331

RESUMO

In endemic areas with high transmission intensities, malaria infections are very often composed of multiple genetically distinct strains of malaria parasites. It has been hypothesised that this leads to intra-host competition, in which parasite strains compete for resources such as space and nutrients. This competition may have repercussions for the host, the parasite, and the vector in terms of disease severity, vector fitness, and parasite transmission potential and fitness. It has also been argued that within-host competition could lead to selection for more virulent parasites. Here we use the rodent malaria parasite Plasmodium yoelii to assess the consequences of mixed strain infections on disease severity and parasite fitness. Three isogenic strains with dramatically different growth rates (and hence virulence) were maintained in mice in single infections or in mixed strain infections with a genetically distinct strain. We compared the virulence (defined as harm to the mammalian host) of mixed strain infections with that of single infections, and assessed whether competition impacted on parasite fitness, assessed by transmission potential. We found that mixed infections were associated with a higher degree of disease severity and a prolonged infection time. In the mixed infections, the strain with the slower growth rate was often responsible for the competitive exclusion of the faster growing strain, presumably through host immune-mediated mechanisms. Importantly, and in contrast to previous work conducted with Plasmodium chabaudi, we found no correlation between parasite virulence and transmission potential to mosquitoes, suggesting that within-host competition would not drive the evolution of parasite virulence in P. yoelii.


Assuntos
Interações Hospedeiro-Parasita/fisiologia , Malária/microbiologia , Plasmodium yoelii/patogenicidade , Animais , Feminino , Malária/genética , Camundongos , Camundongos Endogâmicos CBA , Plasmodium yoelii/genética , Reação em Cadeia da Polimerase em Tempo Real , Virulência
11.
Cytometry A ; 89(6): 531-42, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27078044

RESUMO

In vivo photoacoustic (PA) flow cytometry (PAFC) has already demonstrated a great potential for the diagnosis of deadly diseases through ultrasensitive detection of rare disease-associated circulating markers in whole blood volume. Here, we demonstrate the first application of this powerful technique for early diagnosis of malaria through label-free detection of malaria parasite-produced hemozoin in infected red blood cells (iRBCs) as high-contrast PA agent. The existing malaria tests using blood smears can detect the disease at 0.001-0.1% of parasitemia. On the contrary, linear PAFC showed a potential for noninvasive malaria diagnosis at an extremely low level of parasitemia of 0.0000001%, which is ∼10(3) times better than the existing tests. Multicolor time-of-flight PAFC with high-pulse repetition rate lasers at wavelengths of 532, 671, and 820 nm demonstrated rapid spectral and spatial identification and quantitative enumeration of individual iRBCs. Integration of PAFC with fluorescence flow cytometry (FFC) provided real-time simultaneous detection of single iRBCs and parasites expressing green fluorescence proteins, respectively. A combination of linear and nonlinear nanobubble-based multicolor PAFC showed capability to real-time control therapy efficiency by counting of iRBCs before, during, and after treatment. Our results suggest that high-sensitivity, high-resolution ultrafast PAFC-FFC platform represents a powerful research tool to provide the insight on malaria progression through dynamic study of parasite-cell interactions directly in bloodstream, whereas portable hand-worn PAFC device could be broadly used in humans for early malaria diagnosis. © 2016 International Society for Advancement of Cytometry.


Assuntos
Eritrócitos/parasitologia , Citometria de Fluxo/métodos , Hemeproteínas/análise , Malária/diagnóstico , Parasitemia/diagnóstico , Técnicas Fotoacústicas/instrumentação , Plasmodium yoelii/crescimento & desenvolvimento , Animais , Computadores de Mão , Orelha/irrigação sanguínea , Orelha/parasitologia , Diagnóstico Precoce , Citometria de Fluxo/instrumentação , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hemeproteínas/biossíntese , Hemeproteínas/química , Interações Hospedeiro-Parasita , Lasers , Malária/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Parasitemia/parasitologia , Técnicas Fotoacústicas/métodos , Plasmodium yoelii/patogenicidade , Esquizontes/química , Esquizontes/fisiologia
12.
Antimicrob Agents Chemother ; 59(6): 3672-4, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25870067

RESUMO

There is an urgent need for new antimalarial agents and strategies to treat and control malaria. This study shows an antiplasmodium effect of tulathromycin in mice infected with Plasmodium yoelii. The administration of tulathromycin around the time of infection prevented the progression of disease in 100% of the animals. In addition, highly parasitized mice treated with tulathromycin showed a decreased parasite burden and cleared the parasite faster than did untreated infected mice.


Assuntos
Antimaláricos/uso terapêutico , Dissacarídeos/uso terapêutico , Compostos Heterocíclicos/uso terapêutico , Malária/tratamento farmacológico , Plasmodium yoelii/efeitos dos fármacos , Plasmodium yoelii/patogenicidade , Animais , Feminino , Camundongos
13.
Cell Microbiol ; 16(10): 1533-48, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24798694

RESUMO

Plasmodium sporozoites are transmitted by Anopheles mosquitoes and first infect the liver of their mammalian host, where they develop as liver stages before the onset of erythrocytic infection and malaria symptoms. Sporozoite entry into hepatocytes is an attractive target for anti-malarial prophylactic strategies but remains poorly understood at the molecular level. Apicomplexan parasites invade host cells by forming a parasitophorous vacuole that is essential for parasite development, a process that involves secretion of apical organelles called rhoptries. We previously reported that the host membrane protein CD81 is required for infection by Plasmodium falciparum and Plasmodium yoelii sporozoites. CD81 acts at an early stage of infection, possibly at the entry step, but the mechanisms involved are still unknown. To investigate the role of CD81 during sporozoite entry, we generated transgenic P. yoelii parasites expressing fluorescent versions of three known rhoptry proteins, RON2, RON4 and RAP2/3. We observed that RON2 and RON4 are lost following rhoptry discharge during merozoite and sporozoite entry. In contrast, our data indicate that RAP2/3 is secreted into the parasitophorous vacuole during infection. We further show that sporozoite rhoptry discharge occurs only in the presence of CD81, providing the first direct evidence for a role of CD81 during sporozoite productive invasion.


Assuntos
Interações Hospedeiro-Parasita/fisiologia , Plasmodium yoelii/patogenicidade , Proteínas de Protozoários/metabolismo , Esporozoítos/patologia , Tetraspanina 28/metabolismo , Animais , Linhagem Celular , Feminino , Proteínas de Fluorescência Verde/genética , Células Hep G2 , Hepatócitos/parasitologia , Humanos , Proteínas Luminescentes/genética , Malária , Camundongos , Camundongos Endogâmicos BALB C , Organismos Geneticamente Modificados , Plasmodium yoelii/genética , Proteínas de Protozoários/biossíntese , Proteínas de Protozoários/genética , Vacúolos/patologia , Proteína Vermelha Fluorescente
14.
Parasite Immunol ; 37(7): 349-61, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25886201

RESUMO

Although thymic atrophy and apoptosis of the double-positive (DP) T cells have been reported in murine malaria, comparative studies investigating the effect of lethal and nonlethal Plasmodium infections on the thymus are lacking. We assessed the effects of P. yoelii lethal (17XL) and nonlethal (17XNL) infections on thymic T cells. Both strains affected the thymus. 17XL infection induced DP T-cell apoptosis and a selective decrease in surface CD8 expression on developing thymocytes. By contrast, more severe but reversible effects were observed during 17XNL infection. DP T cells underwent apoptosis, and proliferation of both DN and DP cells was affected around peak parasitemia. A transient increase in surface CD8 expression on thymic T cells was also observed. Adult thymic organ culture revealed that soluble serum factors, but not IFN-γ or TNF-α, contributed to the observed effects. Thus, lethal and nonlethal malarial infections led to multiple disparate effects on thymus. These parasite-induced thymic changes are expected to impact the naïve T-cell repertoire and the subsequent control of the immune response against the parasite. Further investigations are required to elucidate the mechanism responsible for these disparate effects, especially the reversible involution of the thymus in case of nonlethal infection.


Assuntos
Apoptose/imunologia , Antígenos CD8/metabolismo , Malária/imunologia , Plasmodium yoelii/imunologia , Linfócitos T/imunologia , Animais , Proliferação de Células , Interferon gama/metabolismo , Ativação Linfocitária/imunologia , Malária/parasitologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Parasitemia/imunologia , Plasmodium yoelii/patogenicidade , Timo/parasitologia , Timo/patologia , Fator de Necrose Tumoral alfa/metabolismo
15.
J Immunol ; 191(1): 178-86, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23709683

RESUMO

The protective immunity induced by the whole-killed parasite vaccine against malarial blood-stage infection is dependent on the CD4(+) T cell response. However, the mechanism underlying this robust CD4(+) T cell response elicited by the whole-killed parasite vaccine is still largely unknown. In this study, we observe that immunization with Plasmodium yoelii-parasitized RBC lysate activates complement C5 and generates C5a. However, the protective efficacy against P. yoelii 17XL challenge is considerably reduced, and the malaria-specific CD4(+) T cell activation and memory T cell differentiation are largely suppressed in the C5aR-deficient (C5aR(-/-)) mice. An adoptive transfer assay demonstrates that the reduced protection of C5aR(-/-) mice is closely associated with the severely impaired CD4(+) T cell response. This is further confirmed by the fact that administration of C5aR antagonist significantly reduces the protective efficacy of the immunized B cell-deficient mice. Further study indicates that the defective CD4(+) T cell response in C5aR(-/-) mice is unlikely involved in the expansion of CD4(+)CD25(+)Foxp3(+) T cells, but strongly linked to a defect in dendritic cell (DC) maturation and the ability to allostimulate CD4(+) T cells. These results demonstrate that C5aR signaling is essential for the optimal induction of the malaria-specific CD4(+) T cell response by the whole-killed parasite vaccine through modulation of DCs function, which provides us with new clues to design an effective blood-stage subunit vaccine and helps us to understand the mechanism by which the T cell response is regulated by the complement system.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Vacinas Antimaláricas/imunologia , Malária/imunologia , Malária/patologia , Plasmodium yoelii/imunologia , Receptor da Anafilatoxina C5a/fisiologia , Transdução de Sinais/imunologia , Animais , Linfócitos T CD4-Positivos/parasitologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Memória Imunológica/genética , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Malária/parasitologia , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/sangue , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Plasmodium yoelii/crescimento & desenvolvimento , Plasmodium yoelii/patogenicidade , Receptor da Anafilatoxina C5a/sangue , Receptor da Anafilatoxina C5a/deficiência , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/sangue
16.
Biochem Biophys Res Commun ; 451(1): 15-23, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25063026

RESUMO

Developing novel generations of subunit-based antimalarial vaccines in the form of chemically-defined macromolecule systems for multiple antigen presentation represents a classical problem in the field of vaccine development. Many efforts involving synthesis strategies leading to macromolecule constructs have been based on dendrimer-like systems, the condensation of large building blocks and conventional asymmetric double dimer constructs, all based on lysine cores. This work describes novel symmetric double dimer and condensed linear constructs for presenting selected peptide multi-copies from the apical sushi protein expressed in Plasmodium falciparum. These molecules have been proved to be safe and innocuous, highly antigenic and have shown strong protective efficacy in rodents challenged with two Plasmodium species. Insights into systematic design, synthesis and characterisation have led to such novel antigen systems being used as potential platforms for developing new anti-malarial vaccine candidates.


Assuntos
Antígenos de Protozoários/química , Vacinas Antimaláricas/química , Vacinas Antimaláricas/farmacologia , Plasmodium falciparum/química , Sequência de Aminoácidos , Aminocaproatos/química , Animais , Antígenos de Protozoários/imunologia , Antígenos de Protozoários/metabolismo , Epitopos , Humanos , Malária/prevenção & controle , Malária Falciparum/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/imunologia , Plasmodium berghei/patogenicidade , Plasmodium yoelii/patogenicidade , Conformação Proteica , Multimerização Proteica , Coelhos , Vacinas de Subunidades Antigênicas/química , Vacinas de Subunidades Antigênicas/imunologia
17.
PLoS Pathog ; 8(10): e1002982, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23133375

RESUMO

Plasmodium falciparum malaria is responsible for nearly one million annual deaths worldwide. Because of the difficulty in monitoring the pathogenesis of cerebral malaria in humans, we conducted a study in various mouse models to better understand disease progression in experimental cerebral malaria (ECM). We compared the effect on the integrity of the blood brain barrier (BBB) and the histopathology of the brain of P. berghei ANKA, a known ECM model, P. berghei NK65, generally thought not to induce ECM, P. yoelii 17XL, originally reported to induce human cerebral malaria-like histopathology, and P. yoelii YM. As expected, P. berghei ANKA infection caused neurological signs, cerebral hemorrhages, and BBB dysfunction in CBA/CaJ and Swiss Webster mice, while Balb/c and A/J mice were resistant. Surprisingly, PbNK induced ECM in CBA/CaJ mice, while all other mice were resistant. P. yoelii 17XL and P. yoelii YM caused lethal hyperparasitemia in all mouse strains; histopathological alterations, BBB dysfunction, or neurological signs were not observed. Intravital imaging revealed that infected erythrocytes containing mature parasites passed slowly through capillaries making intimate contact with the endothelium, but did not arrest. Except for relatively rare microhemorrhages, mice with ECM presented no obvious histopathological alterations that would explain the widespread disruption of the BBB. Intravital imaging did reveal, however, that postcapillary venules, but not capillaries or arterioles, from mice with ECM, but not hyperparasitemia, exhibit platelet marginalization, extravascular fibrin deposition, CD14 expression, and extensive vascular leakage. Blockage of LFA-1 mediated cellular interactions prevented leukocyte adhesion, vascular leakage, neurological signs, and death from ECM. The endothelial barrier-stabilizing mediators imatinib and FTY720 inhibited vascular leakage and neurological signs and prolonged survival to ECM. Thus, it appears that neurological signs and coma in ECM are due to regulated opening of paracellular-junctional and transcellular-vesicular fluid transport pathways at the neuroimmunological BBB.


Assuntos
Barreira Hematoencefálica/patologia , Malária Cerebral/patologia , Plasmodium berghei/patogenicidade , Plasmodium yoelii/patogenicidade , Animais , Benzamidas , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/fisiopatologia , Hemorragia Cerebral/tratamento farmacológico , Hemorragia Cerebral/etiologia , Modelos Animais de Doenças , Cloridrato de Fingolimode , Mesilato de Imatinib , Receptores de Lipopolissacarídeos/biossíntese , Antígeno-1 Associado à Função Linfocitária/metabolismo , Malária Cerebral/tratamento farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos CBA , Neuroimunomodulação , Parasitemia , Piperazinas/farmacologia , Plasmodium falciparum/patogenicidade , Propilenoglicóis/farmacologia , Pirimidinas/farmacologia , Esfingosina/análogos & derivados , Esfingosina/farmacologia
18.
Cell Microbiol ; 15(7): 1266-83, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23356439

RESUMO

Malaria's cycle of infection requires parasite transmission between a mosquito vector and a mammalian host. We here demonstrate that the Plasmodium yoelii Pumilio-FBF family member Puf2 allows the sporozoite to remain infectious in the mosquito salivary glands while awaiting transmission. Puf2 mediates this solely through its RNA-binding domain (RBD) likely by stabilizing or hastening the degradation of specific mRNAs. Puf2 traffics to sporozoite cytosolic granules, which are negative for several markers of stress granules and P-bodies, and disappear rapidly after infection of hepatocytes. In contrast to previously described Plasmodium berghei pbpuf2(-) parasites, pypuf2(-) sporozoites have no apparent defect in host infection when tested early in salivary gland residence, but become progressively non-infectious and prematurely transform into EEFs during prolonged salivary gland residence. The premature overexpression of Puf2 in oocysts causes striking deregulation of sporozoite maturation and infectivity while extension of Puf2 expression in liverstages causes no defect, suggesting that the presence of Puf2 alone is not sufficient for its functions. Finally, by conducting the first comparative RNA-seq analysis of Plasmodium sporozoites, we find that Puf2 may play a role in directly or indirectly maintaining the homeostasis of specific transcripts. These findings uncover requirements for maintaining a window of opportunity for the malaria parasite to accommodate the unpredictable moment of transmission from mosquito to mammalian host.


Assuntos
Regulação da Expressão Gênica , Plasmodium yoelii/patogenicidade , Proteínas de Protozoários/metabolismo , RNA/metabolismo , Esporozoítos/metabolismo , Animais , Culicidae , Homeostase , Camundongos , Plasmodium yoelii/genética , Plasmodium yoelii/metabolismo , Proteínas de Protozoários/genética , Estabilidade de RNA
19.
Proc Natl Acad Sci U S A ; 108(31): E374-82, 2011 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-21690382

RESUMO

Plasmodium yoelii is an excellent model for studying malaria pathogenesis that is often intractable to investigate using human parasites; however, genetic studies of the parasite have been hindered by lack of genome-wide linkage resources. Here, we performed 14 genetic crosses between three pairs of P. yoelii clones/subspecies, isolated 75 independent recombinant progeny from the crosses, and constructed a high-resolution linkage map for this parasite. Microsatellite genotypes from the progeny formed 14 linkage groups belonging to the 14 parasite chromosomes, allowing assignment of sequence contigs to chromosomes. Growth-related virulent phenotypes from 25 progeny of one of the crosses were significantly associated with a major locus on chromosome 13 and with two secondary loci on chromosomes 7 and 10. The chromosome 10 and 13 loci are both linked to day 5 parasitemia, and their effects on parasite growth rate are independent but additive. The locus on chromosome 7 is associated with day 10 parasitemia. The chromosome 13 locus spans ~220 kb of DNA containing 51 predicted genes, including the P. yoelii erythrocyte binding ligand, in which a C741Y substitution in the R6 domain is implicated in the change of growth rate. Similarly, the chromosome 10 locus spans ~234 kb with 71 candidate genes, containing a member of the 235-kDa rhoptry proteins (Py235) that can bind to the erythrocyte surface membrane. Atypical virulent phenotypes among the progeny were also observed. This study provides critical tools and information for genetic investigations of virulence and biology of P. yoelii.


Assuntos
Mapeamento Cromossômico/métodos , Genes de Protozoários/genética , Genoma de Protozoário/genética , Plasmodium yoelii/genética , Animais , Cromossomos/genética , Eritrócitos/parasitologia , Feminino , Malária/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Filogenia , Plasmodium yoelii/classificação , Plasmodium yoelii/patogenicidade , Especificidade da Espécie , Virulência/genética
20.
Proc Natl Acad Sci U S A ; 108(27): 11205-10, 2011 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-21690346

RESUMO

Plasmodium spp. parasites cause malaria in 300 to 500 million individuals each year. Disease occurs during the blood-stage of the parasite's life cycle, where the parasite is thought to replicate exclusively within erythrocytes. Infected individuals can also suffer relapses after several years, from Plasmodium vivax and Plasmodium ovale surviving in hepatocytes. Plasmodium falciparum and Plasmodium malariae can also persist after the original bout of infection has apparently cleared in the blood, suggesting that host cells other than erythrocytes (but not hepatocytes) may harbor these blood-stage parasites, thereby assisting their escape from host immunity. Using blood stage transgenic Plasmodium berghei-expressing GFP (PbGFP) to track parasites in host cells, we found that the parasite had a tropism for CD317(+) dendritic cells. Other studies using confocal microscopy, in vitro cultures, and cell transfer studies showed that blood-stage parasites could infect, survive, and replicate within CD317(+) dendritic cells, and that small numbers of these cells released parasites infectious for erythrocytes in vivo. These data have identified a unique survival strategy for blood-stage Plasmodium, which has significant implications for understanding the escape of Plasmodium spp. from immune-surveillance and for vaccine development.


Assuntos
Células Dendríticas/parasitologia , Malária/parasitologia , Plasmodium/crescimento & desenvolvimento , Plasmodium/patogenicidade , Animais , Animais Geneticamente Modificados , Antígenos CD/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/ultraestrutura , Eritrócitos/parasitologia , Feminino , Proteínas de Fluorescência Verde/genética , Humanos , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Plasmodium/imunologia , Plasmodium berghei/genética , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium berghei/patogenicidade , Plasmodium chabaudi/patogenicidade , Plasmodium yoelii/patogenicidade , Proteínas Recombinantes/genética , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA