Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
N Engl J Med ; 385(13): 1163-1171, 2021 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-34551228

RESUMO

BACKGROUND: In the six Southeast Asian countries that make up the Greater Mekong Subregion, Plasmodium falciparum has developed resistance to derivatives of artemisinin, the main component of first-line treatments for malaria. Clinical resistance to artemisinin monotherapy in other global regions, including Africa, would be problematic. METHODS: In this longitudinal study conducted in Northern Uganda, we treated patients who had P. falciparum infection with intravenous artesunate (a water-soluble artemisinin derivative) and estimated the parasite clearance half-life. We evaluated ex vivo susceptibility of the parasite using a ring-stage survival assay and genotyped resistance-related genes. RESULTS: From 2017 through 2019, a total of 14 of 240 patients who received intravenous artesunate had evidence of in vivo artemisinin resistance (parasite clearance half-life, >5 hours). Of these 14 patients, 13 were infected with P. falciparum parasites with mutations in the A675V or C469Y allele in the kelch13 gene. Such mutations were associated with prolonged parasite clearance half-lives (geometric mean, 3.95 hours for A675V and 3.30 hours for C469Y, vs. 1.78 hours for wild-type allele; P<0.001 and P = 0.05, respectively). The ring-stage survival assay showed a higher frequency of parasite survival among organisms with the A675V allele than among those with the wild-type allele. The prevalence of parasites with kelch13 mutations increased significantly, from 3.9% in 2015 to 19.8% in 2019, due primarily to the increased frequency of the A675V and C469Y alleles (P<0.001 and P = 0.004, respectively). Single-nucleotide polymorphisms flanking the A675V mutation in Uganda were substantially different from those in Southeast Asia. CONCLUSIONS: The independent emergence and local spread of clinically artemisinin-resistant P. falciparum has been identified in Africa. The two kelch13 mutations may be markers for detection of these resistant parasites. (Funded by the Japan Society for the Promotion of Science and others.).


Assuntos
Antimaláricos/uso terapêutico , Artemisininas/uso terapêutico , Resistência a Medicamentos/genética , Malária Falciparum/tratamento farmacológico , Mutação , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Humanos , Estudos Longitudinais , Polimorfismo de Nucleotídeo Único , Uganda
2.
Front Cell Infect Microbiol ; 11: 672691, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34222045

RESUMO

Elucidation of the mechanisms of drug resistance in malaria parasites is crucial for combatting the emergence and spread of resistant parasites, which can be achieved by tracing resistance-associated mutations and providing useful information for drug development. Previously, we produced a novel genetic tool, a Plasmodium berghei mutator (PbMut), whose base substitution rate is 36.5 times higher than that of wild-type parasites. Here, we report the isolation of a mutant with reduced susceptibility to piperaquine (PPQ) from PbMut under PPQ pressure by sequential nine-cycle screening and named it PbMut-PPQ-R-P9. The ED50 of PbMut-PPQ-R-P9 was 1.79 times higher than that of wild-type parasites, suggesting that its PPQ resistance is weak. In the 1st screen, recrudescence occurred in the mice infected with PbMut but not in those infected with wild-type parasites, suggesting earlier emergence of PPQ-resistant parasites from PbMut. Whole-genome sequence analysis of PbMut-PPQ-R-P9 clones revealed that eight nonsynonymous mutations were conserved in all clones, including N331I in PbCRT, the gene encoding chloroquine resistance transporter (CRT). The PbCRT(N331I) mutation already existed in the parasite population after the 2nd screen and was predominant in the population after the 8th screen. An artificially inserted PbCRT(N331I) mutation gave rise to reduced PPQ susceptibility in genome-edited parasites (PbCRT-N331I). The PPQ susceptibility and growth rates of PbCRT-N331I parasites were significantly lower than those of PbMut-PPQ-R-P9, implying that additional mutations in the PbMut-PPQ-R9 parasites could compensate for the fitness cost of the PbCRT(N331I) mutation and contribute to reduced PPQ susceptibility. In summary, PbMut could serve as a novel genetic tool for predicting gene mutations responsible for drug resistance. Further study on PbMut-PPQ-R-P9 could identify genetic changes that compensate for fitness costs owing to drug resistance acquisition.


Assuntos
Antimaláricos , Malária Falciparum , Malária , Parasitos , Animais , Antimaláricos/farmacologia , Malária/tratamento farmacológico , Camundongos , Plasmodium berghei/genética , Plasmodium falciparum , Proteínas de Protozoários/genética , Quinolinas , Roedores
3.
Acta Trop ; 222: 106049, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34273314

RESUMO

Genetic changes conferring drug resistance are generally believed to impose fitness costs to pathogens in the absence of the drug. However, the fitness of resistant parasites against sulfadoxine/pyrimethamine has been inconclusive in Plasmodium falciparum. This is because resistance is conferred by the complex combination of mutations in dihydropteroate synthase (dhps) and dihydrofolate reductase (dhfr), which makes it difficult to separately assess the extent and magnitude of the costs imposed by mutations in dhps and dhfr. To assess the fitness costs imposed by sulfadoxine resistance alone, we generated a transgenic rodent malaria parasite, P. berghei clone harboring an A394G mutation in dhps (PbDHPS-A394G), corresponding to the causative mutation for sulfadoxine resistance in P. falciparum (PfDHPS-A437G). A four-day suppressive test confirmed that the PbDHPS-A394G clone was resistant to sulfadoxine. PbDHPS-A394G and wild-type clones showed similar growth rates and gametocyte production. This observation was confirmed in competitive experiments in which PbDHPS-A394G and wild-type clones were co-infected into mice to directly assess the survival competition between them. In the mosquitoes, there were no significant differences in oocyst production between PbDHPS-A394G and wild-type. These results indicate that the PbDHPS-A394G mutation alters the parasites to sulfadoxine resistance but may not impose fitness disadvantages during the blood stages in mice and oocyst formation in mosquitoes. These results partly explain the persistence of the PfDHPS-A437G mutant in the natural parasite populations.


Assuntos
Antimaláricos , Resistência a Medicamentos , Sulfadoxina , Tetra-Hidrofolato Desidrogenase , Animais , Antimaláricos/farmacologia , Di-Hidropteroato Sintase/genética , Combinação de Medicamentos , Resistência a Medicamentos/genética , Camundongos , Mutação , Plasmodium berghei/efeitos dos fármacos , Plasmodium berghei/enzimologia , Plasmodium berghei/genética , Pirimetamina/farmacologia , Sulfadoxina/farmacologia , Tetra-Hidrofolato Desidrogenase/genética
4.
Sci Rep ; 11(1): 6455, 2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33742008

RESUMO

Scabies is a highly contagious skin disease caused by the mite Sarcoptes scabiei that affects many mammals. However, the sensitivity of traditional tests for scabies diagnosis in humans is less than 50%. To simplify the diagnosis of scabies, methods that are simple, sensitive, specific, and cost-effective are required. We developed an immunodiagnostic test based on S. scabiei var. nyctereutis RNA-seq data collected from Japanese raccoon dogs with sarcoptic mange. Three candidate antigens-a highly expressed hypothetical protein "QR98_0091190," another mite allergen known as "SMIPP-Cc," and an abundant "vitellogenin-like protein"-were evaluated by western-blot analysis. A lateral flow immunoassay, using specific antibodies against the vitellogenin-like protein, successfully detected scabies in the skin flakes of S. scabiei-infected raccoon dogs. This assay can potentially diagnose scabies more accurately in wildlife, as well as in humans.


Assuntos
Alérgenos/imunologia , Proteínas de Artrópodes/imunologia , Testes Imunológicos/métodos , Sarcoptes scabiei/imunologia , Escabiose/diagnóstico , Transcriptoma , Alérgenos/genética , Animais , Proteínas de Artrópodes/genética , Cães Guaxinins/parasitologia , Sarcoptes scabiei/genética , Sarcoptes scabiei/patogenicidade , Pele/parasitologia
5.
Parasitol Int ; 81: 102277, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33370608

RESUMO

In Uganda, artemether-lumefantrine was introduced as an artemisinin-based combination therapy (ACT) for malaria in 2006. We have previously reported a moderate decrease in ex vivo efficacy of lumefantrine in Northern Uganda, where we also detected ex vivo artemisinin-resistant Plasmodium falciparum. Therefore, it is necessary to search for candidate partner alternatives for ACT. Here, we investigated ex vivo susceptibility to four ACT partner drugs as well as quinine and chloroquine, in 321 cases between 2013 and 2018. Drug-resistant mutations in pfcrt and pfmdr1 were also determined. Ex vivo susceptibility to amodiaquine, quinine, and chloroquine was well preserved, whereas resistance to mefloquine was found in 45.8%. There were few cases of multi-drug resistance. Reduced sensitivity to mefloquine and lumefantrine was significantly associated with the pfcrt K76 wild-type allele, in contrast to the association between chloroquine resistance and the K76T allele. Pfmdr1 duplication was not detected in any of the cases. Amodiaquine, a widely used partner drug for ACT in African countries, may be the first promising alternative in case lumefantrine resistance emerges. Therapeutic use of mefloquine may not be recommended in this area. This study also emphasizes the need for sustained monitoring of antimalarial susceptibility in Northern Uganda to develop proper treatment strategies.


Assuntos
Antimaláricos/farmacologia , Resistência a Medicamentos , Plasmodium falciparum/efeitos dos fármacos , Amodiaquina/farmacologia , Artemisininas/farmacologia , Cloroquina/farmacologia , Lumefantrina/farmacologia , Mefloquina/farmacologia , Quinina/farmacologia , Uganda
6.
PLoS Pathog ; 16(12): e1009133, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33320907

RESUMO

The rapid and aggressive spread of artemisinin-resistant Plasmodium falciparum carrying the C580Y mutation in the kelch13 gene is a growing threat to malaria elimination in Southeast Asia, but there is no evidence of their spread to other regions. We conducted cross-sectional surveys in 2016 and 2017 at two clinics in Wewak, Papua New Guinea (PNG) where we identified three infections caused by C580Y mutants among 239 genotyped clinical samples. One of these mutants exhibited the highest survival rate (6.8%) among all parasites surveyed in ring-stage survival assays (RSA) for artemisinin. Analyses of kelch13 flanking regions, and comparisons of deep sequencing data from 389 clinical samples from PNG, Indonesian Papua and Western Cambodia, suggested an independent origin of the Wewak C580Y mutation, showing that the mutants possess several distinctive genetic features. Identity by descent (IBD) showed that multiple portions of the mutants' genomes share a common origin with parasites found in Indonesian Papua, comprising several mutations within genes previously associated with drug resistance, such as mdr1, ferredoxin, atg18 and pnp. These findings suggest that a P. falciparum lineage circulating on the island of New Guinea has gradually acquired a complex ensemble of variants, including kelch13 C580Y, which have affected the parasites' drug sensitivity. This worrying development reinforces the need for increased surveillance of the evolving parasite populations on the island, to contain the spread of resistance.


Assuntos
Anti-Infecciosos , Artemisininas , Resistência a Medicamentos/genética , Genes de Protozoários/genética , Plasmodium falciparum/genética , Anti-Infecciosos/uso terapêutico , Artemisininas/uso terapêutico , Estudos Transversais , Humanos , Malária Falciparum/tratamento farmacológico , Malária Falciparum/parasitologia , Mutação , Papua Nova Guiné
7.
Malar J ; 19(1): 76, 2020 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-32070358

RESUMO

BACKGROUND: Usage of chloroquine was discontinued from the treatment of Plasmodium falciparum infection in almost all endemic regions because of global spread of resistant parasites. Since the first report in Malawi, numerous epidemiological studies have demonstrated that the discontinuance led to re-emergence of chloroquine-susceptible P. falciparum, suggesting a possible role in future malaria control. However, most studies were cross-sectional, with few studies looking at the persistence of chloroquine recovery in long term. This study fills the gap by providing, for a period of at least 6 years, proof of persistent re-emergence/stable recovery of susceptible parasite populations using both molecular and phenotypic methods. METHODS: Ex vivo drug-susceptibility assays to chloroquine (n = 319) and lumefantrine (n = 335) were performed from 2013 to 2018 in Gulu, Northern Uganda, where chloroquine had been removed from the official malaria treatment regimen since 2006. Genotyping of pfcrt and pfmdr1 was also performed. RESULTS: Chloroquine resistance (≥ 100 nM) was observed in only 3 (1.3%) samples. Average IC50 values for chloroquine were persistently low throughout the study period (17.4-24.9 nM). Parasites harbouring pfcrt K76 alleles showed significantly lower IC50s to chloroquine than the parasites harbouring K76T alleles (21.4 nM vs. 43.1 nM, p-value = 3.9 × 10-8). Prevalence of K76 alleles gradually increased from 71% in 2013 to 100% in 2018. CONCLUSION: This study found evidence of stable persistence of chloroquine susceptibility with the fixation of pfcrt K76 in Northern Uganda after discontinuation of chloroquine in the region. Accumulation of similar evidence in other endemic areas in Uganda could open channels for possible future re-use of chloroquine as an option for malaria treatment or prevention.


Assuntos
Antimaláricos/farmacologia , Cloroquina/farmacologia , Resistência a Medicamentos , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/genética , Uganda
8.
J Econ Entomol ; 113(1): 427-434, 2020 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-31693096

RESUMO

In most insects dependent on food resources that deplete seasonally, mechanisms exist to protect against starvation. Insects overcome periods of food depletion using diapause-associated physiological mechanisms, such as increased energy resources in fat bodies and suppression of metabolism. Because autophagy supplies energy resources through the degradation of intracellular components, we hypothesized that it might be an additional strategy to combat starvation during overwintering. In this study, we measured the abundance of the proteins involved in the signaling pathway of autophagy during overwintering in adults of the bean bug Riptortus pedestris (Fabricius) (Hemiptera: Alydidae), which must withstand the periodic depletion of its host plants from late fall to early spring. Although the levels of gamma-aminobutyric acid receptor-associated protein (GABARAP) markedly increased after the cessation of food supply, AMP-activated protein kinase (AMPK) and target of rapamycin (TOR) were not found to be associated with food depletion. Thus, food depletion appears to induce autophagy independent of AMPK and TOR. The GABARAP levels significantly increased universally when the food supply ceased, irrespective of the diapause status of adults and low-temperature conditions. In overwintering diapause adults under seminatural conditions, the GABARAP levels significantly increased during early spring. Thus, autophagy appears to assist the survival of the bean bugs under natural conditions of food deficiency.


Assuntos
Autofagia , Fabaceae , Heterópteros , Animais , Proteínas Relacionadas à Autofagia , Receptores de GABA
9.
Malar J ; 17(1): 434, 2018 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-30477515

RESUMO

BACKGROUND: Chloroquine treatment for Plasmodium falciparum has been discontinued in almost all endemic regions due to the spread of resistant isolates. Reversal of chloroquine susceptibility after chloroquine discontinuation has been reported in dozens of endemic regions. However, this phenomenon has been mostly observed in Africa and is not well documented in other malaria endemic regions. To investigate this, an ex vivo study on susceptibility to chloroquine and lumefantrine was conducted during 2016-2018 in Wewak, Papua New Guinea where chloroquine had been removed from the official malaria treatment regimen in 2010. Genotyping of pfcrt and pfmdr1 was also performed. RESULTS: In total, 368 patients were enrolled in this study. Average IC50 values for chloroquine were 106.6, 80.5, and 87.6 nM in 2016, 2017, and 2018, respectively. These values were not significantly changed from those obtained in 2002/2003 (108 nM). The majority of parasites harboured a pfcrt K76T the mutation responsible for chloroquine resistance. However, a significant upward trend was observed in the frequency of the K76 (wild) allele from 2.3% in 2016 to 11.7% in 2018 (P = 0.008; Cochran-Armitage trend test). CONCLUSIONS: Eight years of chloroquine withdrawal has not induced a significant recovery of susceptibility in Papua New Guinea. However, an increasing tendency of parasites harbouring chloroquine-susceptible K76 suggests a possibility of resurgence of chloroquine susceptibility in the future.


Assuntos
Antimaláricos/farmacologia , Antimaláricos/uso terapêutico , Cloroquina/farmacologia , Cloroquina/uso terapêutico , Resistência a Medicamentos , Malária Falciparum/tratamento farmacológico , Plasmodium falciparum/efeitos dos fármacos , Adolescente , Adulto , Criança , Pré-Escolar , Estudos Transversais , Uso de Medicamentos , Feminino , Genótipo , Técnicas de Genotipagem , Humanos , Lactente , Recém-Nascido , Concentração Inibidora 50 , Lumefantrina/farmacologia , Lumefantrina/uso terapêutico , Malária Falciparum/parasitologia , Masculino , Proteínas de Membrana Transportadoras/genética , Pessoa de Meia-Idade , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Papua Nova Guiné , Testes de Sensibilidade Parasitária , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Adulto Jovem
10.
Emerg Infect Dis ; 24(4): 718-726, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29553316

RESUMO

Because ≈90% of malaria cases occur in Africa, emergence of artemisinin-resistant Plasmodium falciparum in Africa poses a serious public health threat. To assess emergence of artemisinin-resistant parasites in Uganda during 2014-2016, we used the recently developed ex vivo ring-stage survival assay, which estimates ring-stage-specific P. falciparum susceptibility to artemisinin. We conducted 4 cross-sectional surveys to assess artemisinin sensitivity in Gulu, Uganda. Among 194 isolates, survival rates (ratio of viable drug-exposed parasites to drug-nonexposed controls) were high (>10%) for 4 isolates. Similar rates have been closely associated with delayed parasite clearance after drug treatment and are considered to be a proxy for the artemisinin-resistant phenotype. Of these, the PfKelch13 mutation was observed in only 1 isolate, A675V. Population genetics analysis suggested that these possibly artemisinin-resistant isolates originated in Africa. Large-scale surveillance of possibly artemisinin-resistant parasites in Africa would provide useful information about treatment outcomes and help regional malaria control.


Assuntos
Antimaláricos/farmacologia , Artemisininas/farmacologia , Resistência a Medicamentos , Malária Falciparum/epidemiologia , Malária Falciparum/parasitologia , Plasmodium falciparum/efeitos dos fármacos , Antimaláricos/uso terapêutico , Artemisininas/uso terapêutico , Pré-Escolar , Estudos Transversais , Feminino , Genótipo , História do Século XXI , Humanos , Malária Falciparum/história , Malária Falciparum/mortalidade , Masculino , Mutação , Fenótipo , Plasmodium falciparum/genética , Taxa de Sobrevida , Uganda/epidemiologia , Sequenciamento Completo do Genoma
11.
Malar J ; 16(1): 23, 2017 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-28068997

RESUMO

BACKGROUND: Individual drug treatment may select resistant parasites in the human body, a process termed in vivo selection. Some single nucleotide polymorphisms in Plasmodium falciparum chloroquine-resistance transporter (pfcrt) and multidrug resistance gene 1 (pfmdr1) genes have been reportedly selected after artemether-lumefantrine treatment. However, there is a paucity of data regarding in vivo selection of P. falciparum Kelch propeller domain (pfkelch13) polymorphisms, responsible for artemisinin-resistance in Asia, and six putative background mutations for artemisinin resistance; D193Y in ferredoxin, T484I in multiple resistance protein 2, V127M in apicoplast ribosomal protein S10, I356T in pfcrt, V1157L in protein phosphatase and C1484F in phosphoinositide-binding protein. METHODS: Artemether-lumefantrine efficacy study with a follow-up period of 28 days was conducted in northern Uganda in 2014. The above-mentioned genotypes were comparatively analysed before drug administration and on days; 3, 7, and 28 days after treatment. RESULTS: In 61 individuals with successful follow-up, artemether-lumefantrine treatment regimen was very effective with PCR adjusted efficacy of 95.2%. Among 146 isolates obtained before treatment, wild-type alleles were observed in 98.6% of isolates in pfkelch13 and in all isolates in the six putative background genes except I356T in pfcrt, which had 2.4% of isolates as mixed infections. In vivo selection study revealed that all isolates detected in the follow-up period harboured wild type alleles in pfkelch13 and the six background genes. CONCLUSION: Mutations in pfkelch13 and the six background genes may not play an important role in the in vivo selection after artemether-lumefantrine treatment in Uganda. Different mechanisms might rather be associated with the existence of parasites after treatment.


Assuntos
Antimaláricos/uso terapêutico , Artemisininas/uso terapêutico , Resistência a Medicamentos , Etanolaminas/uso terapêutico , Fluorenos/uso terapêutico , Malária Falciparum/tratamento farmacológico , Plasmodium falciparum/efeitos dos fármacos , Proteínas de Protozoários/genética , Seleção Genética , Adolescente , Adulto , Combinação Arteméter e Lumefantrina , Criança , Pré-Escolar , Combinação de Medicamentos , Feminino , Humanos , Lactente , Malária Falciparum/parasitologia , Masculino , Mutação , Plasmodium falciparum/isolamento & purificação , Polimorfismo Genético , Uganda , Adulto Jovem
12.
Expert Rev Anti Infect Ther ; 14(1): 125-35, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26535806

RESUMO

Although artemisinin combination therapies have been deployed as a first-line treatment for uncomplicated malaria in almost all endemic countries, artemisinin-resistant parasites have emerged and have gradually spread across the Greater Mekong subregions. There is growing concern that the resistant parasites may migrate to or emerge indigenously in sub-Saharan Africa, which might provoke a global increase in malaria-associated morbidity and mortality. Therefore, development of molecular markers that enable identification of artemisinin resistance with high sensitivity is urgently required to combat this issue. In 2014, a potential artemisinin-resistance responsible gene, Plasmodium falciparum kelch13, was discovered. Here, we review the genetic features of P. falciparum kelch13 and discuss its related resistant mechanisms and potential as a molecular marker.


Assuntos
Antimaláricos/farmacologia , Artemisininas/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Plasmodium falciparum/efeitos dos fármacos , Proteínas de Protozoários/genética , África Subsaariana/epidemiologia , Sudeste Asiático/epidemiologia , Biomarcadores/metabolismo , Resistência a Medicamentos/genética , Regulação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Malária Falciparum/tratamento farmacológico , Malária Falciparum/epidemiologia , Malária Falciparum/parasitologia , Fosfatidilinositol 3-Quinase/genética , Fosfatidilinositol 3-Quinase/metabolismo , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas de Protozoários/metabolismo , Transdução de Sinais , Ubiquitina/genética , Ubiquitina/metabolismo , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Resposta a Proteínas não Dobradas/genética
13.
PLoS One ; 10(8): e0135186, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26252206

RESUMO

A male-specific component, 11-cis-vaccenyl acetate (cVA) works as an anti-aphrodisiac pheromone in Drosophila melanogaster. The presence of cVA on a male suppresses the courtship motivation of other males and contributes to suppression of male-male homosexual courtship, while the absence of cVA on a female stimulates the sexual motivation of nearby males and enhances the male-female interaction. However, little is known how a male distinguishes the presence or absence of cVA on a target fly from either self-produced cVA or secondhand cVA from other males in the vicinity. In this study, we demonstrate that male flies have keen sensitivity to cVA; therefore, the presence of another male in the area reduces courtship toward a female. This reduced level of sexual motivation, however, could be overcome by pretest odor exposure via olfactory habituation to cVA. Real-time imaging of cVA-responsive sensory neurons using the neural activity sensor revealed that prolonged exposure to cVA decreased the levels of cVA responses in the primary olfactory center. Pharmacological and genetic screening revealed that signal transduction via GABAA receptors contributed to this olfactory habituation. We also found that the habituation experience increased the copulation success of wild-type males in a group. In contrast, transgenic males, in which GABA input in a small subset of local neurons was blocked by RNAi, failed to acquire the sexual advantage conferred by habituation. Thus, we illustrate a novel phenomenon in which olfactory habituation positively affects sexual capability in a competitive environment.


Assuntos
Drosophila melanogaster/fisiologia , Receptores de GABA-A/metabolismo , Comportamento Sexual Animal , Olfato/fisiologia , Animais , Animais Geneticamente Modificados , Proteínas de Drosophila/metabolismo , Feminino , Habituação Psicofisiológica , Masculino , Microscopia de Fluorescência , Rede Nervosa , Neurônios/metabolismo , Neurotransmissores/metabolismo , Odorantes , Ácidos Oleicos/química , Feromônios/química , Feromônios/metabolismo , Interferência de RNA , Receptores de Superfície Celular/metabolismo , Receptores Odorantes/metabolismo , Transdução de Sinais
14.
PLoS One ; 10(3): e0119475, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25793260

RESUMO

Understanding the transmission and movement of Plasmodium parasites is crucial for malaria elimination and prevention of resurgence. Located at the limit of malaria transmission in the Pacific, Vanuatu is an ideal candidate for elimination programs due to low endemicity and the isolated nature of its island setting. We analyzed the variation in the merozoite surface protein 1 (msp1) and the circumsporozoite protein (csp) of P. falciparum and P. vivax populations to examine the patterns of gene flow and population structures among seven sites on five islands in Vanuatu. Genetic diversity was in general higher in P. vivax than P. falciparum from the same site. In P. vivax, high genetic diversity was likely maintained by greater extent of gene flow among sites and among islands. Consistent with the different patterns of gene flow, the proportion of genetic variance found among islands was substantially higher in P. falciparum (28.81-31.23%) than in P. vivax (-0.53-3.99%). Our data suggest that the current island-by-island malaria elimination strategy in Vanuatu, while adequate for P. falciparum elimination, might need to be complemented with more centrally integrated measures to control P. vivax movement across islands.


Assuntos
Malária Falciparum/epidemiologia , Malária Falciparum/parasitologia , Malária Vivax/epidemiologia , Malária Vivax/parasitologia , Plasmodium falciparum/genética , Plasmodium vivax/genética , Fluxo Gênico , Variação Genética , Genótipo , Geografia , Humanos , Dados de Sequência Molecular , Proteínas de Protozoários/genética , Vanuatu/epidemiologia
15.
Parasitol Int ; 64(3): 274-81, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25316604

RESUMO

Although the human malaria parasite Plasmodium vivax is closely related to Asian Old World monkey malaria parasites, there are no reports of P. vivax infections in macaques. In this study, we compared the infectivity of P. vivax and Plasmodium cynomolgi in Japanese macaques (Macaca fuscata) and in cynomolgus macaques (Macaca fascicularis). The Japanese macaques were highly susceptible to P. cynomolgi but not to P. vivax, whereas cynomolgus macaques showed mild/limited P. cynomolgi infection and were, also, not susceptible to P. vivax. Serotyping and amino acid sequence comparison of erythrocyte surface Duffy antigen/receptor for chemokines (DARC) indicate that the Japanese macaque DARC sequence is nearly identical to that of rhesus (Macaca mulatta) and cynomolgus macaques. This suggests that the macaques share a common mechanism for preventing P. vivax infection. Comparison of amino acid sequences of the Duffy-binding-like (DBL) domain from several different Plasmodium species suggests that P. vivax DBLs will not bind to macaque DARCs, which can explain the lack of P. vivax infectivity. The DBL sequence analyses also suggest that P. cynomolgi DBLs may target Japanese macaque erythrocytes through a DARC-independent interaction.


Assuntos
Antígenos de Protozoários/genética , Macaca/parasitologia , Malária Vivax/veterinária , Malária/veterinária , Doenças dos Macacos/parasitologia , Plasmodium cynomolgi/patogenicidade , Plasmodium vivax/patogenicidade , Proteínas de Protozoários/genética , Receptores de Superfície Celular/genética , Sequência de Aminoácidos , Animais , Antígenos de Protozoários/química , Suscetibilidade a Doenças , Humanos , Macaca/sangue , Macaca/genética , Macaca/imunologia , Macaca fascicularis/sangue , Macaca fascicularis/genética , Macaca fascicularis/imunologia , Macaca mulatta/sangue , Macaca mulatta/genética , Macaca mulatta/imunologia , Malária/parasitologia , Malária Vivax/parasitologia , Dados de Sequência Molecular , Plasmodium cynomolgi/genética , Plasmodium vivax/genética , Proteínas de Protozoários/química , Receptores de Superfície Celular/química , Sorotipagem , Especificidade da Espécie
16.
Infect Immun ; 82(1): 243-52, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24166950

RESUMO

Resurgence is a major concern after malaria elimination. After the initiation of the elimination program on Aneityum Island in 1991, microscopy showed that Plasmodium falciparum disappeared immediately, whereas P. vivax disappeared from 1996 onward, until P. vivax cases were reported in January 2002. By conducting malariometric surveys of the entire population of Aneityum, we investigated the age distribution of individuals with parasites during this epidemic in the context of antimalarial antibody levels and parasite antigen diversity. In July 2002, P. vivax infections were detected by microscopy in 22/759 individuals: 20/298 born after the beginning of the elimination program in 1991, 2/126 born between 1982 and 1991, and none of 335 born before 1982. PCR increased the number of infections detected to 77, distributed among all age groups. Prevalences were 12.1%, 16.7%, and 6.0%, respectively (P < 0.001). In November, a similar age pattern was found, but with fewer infections: 6/746 and 39/741 individuals were found to be infected by microscopy and PCR, respectively. The frequencies of antibody responses to P. vivax were significantly higher in individuals born before 1991 than in younger age groups and were similar to those on Malakula Island, an area of endemicity. Remarkably low antigen diversity (h, 0.15) of P. vivax infections was observed on Aneityum compared with the other islands (h, 0.89 to 1.0). A P. vivax resurgence was observed among children and teenagers on Aneityum, an age distribution similar to those before elimination and on islands where P. vivax is endemic, suggesting that in the absence of significant exposure, immunity may persist, limiting infection levels in adults. The limited parasite gene pool on islands may contribute to this protection.


Assuntos
Malária Vivax/epidemiologia , Adolescente , Adulto , Distribuição por Idade , Antígenos de Protozoários/sangue , Criança , Pré-Escolar , DNA de Protozoário/análise , Feminino , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Plasmodium vivax/genética , Plasmodium vivax/isolamento & purificação , Prevalência , Análise de Sequência de DNA , Estudos Soroepidemiológicos , Vanuatu/epidemiologia , Adulto Jovem
17.
Mitochondrion ; 13(6): 630-6, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24004956

RESUMO

The geographical distribution of single nucleotide polymorphism (SNP) in the mitochondrial genome of the human malaria parasite Plasmodium falciparum was investigated. We identified 88 SNPs in 516 isolates from seven parasite populations in Africa, Southeast Asia and Oceania. Analysis of the SNPs postulated a sub-Saharan African origin and recovered a strong negative correlation between within-population SNP diversity and geographic distance from the putative African origin over Southeast Asia and Oceania. These results are consistent with those previously obtained for nuclear genome-encoded housekeeping genes, indicating that the pattern of inheritance does not substantially affect the geographical distribution of SNPs.


Assuntos
Genoma Mitocondrial , Plasmodium falciparum/genética , África Subsaariana , Animais , Haplótipos , Reação em Cadeia da Polimerase , Polimorfismo de Nucleotídeo Único
18.
Nat Genet ; 44(9): 1051-5, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22863735

RESUMO

P. cynomolgi, a malaria-causing parasite of Asian Old World monkeys, is the sister taxon of P. vivax, the most prevalent malaria-causing species in humans outside of Africa. Because P. cynomolgi shares many phenotypic, biological and genetic characteristics with P. vivax, we generated draft genome sequences for three P. cynomolgi strains and performed genomic analysis comparing them with the P. vivax genome, as well as with the genome of a third previously sequenced simian parasite, Plasmodium knowlesi. Here, we show that genomes of the monkey malaria clade can be characterized by copy-number variants (CNVs) in multigene families involved in evasion of the human immune system and invasion of host erythrocytes. We identify genome-wide SNPs, microsatellites and CNVs in the P. cynomolgi genome, providing a map of genetic variation that can be used to map parasite traits and study parasite populations. The sequencing of the P. cynomolgi genome is a critical step in developing a model system for P. vivax research and in counteracting the neglect of P. vivax.


Assuntos
Genoma de Protozoário , Haplorrinos/parasitologia , Doenças dos Macacos/parasitologia , Plasmodium cynomolgi/genética , Plasmodium vivax/genética , Animais , Sequência de Bases , Análise por Conglomerados , Genes de Protozoários , Genoma de Protozoário/genética , Malária/genética , Malária/parasitologia , Modelos Genéticos , Dados de Sequência Molecular , Doenças dos Macacos/classificação , Doenças dos Macacos/genética , Filogenia , Plasmodium cynomolgi/classificação , Plasmodium vivax/classificação , Análise de Sequência de DNA
19.
Gene ; 502(1): 36-9, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22555021

RESUMO

Parasites of the genus Plasmodium infect all classes of amniotes (mammals, birds and reptiles) and display host specificity in their infections. It is therefore generally believed that Plasmodium parasites co-evolved intimately with their hosts. Here, we report that based on an evolutionary analysis using 22 genes in the nuclear genome, extant lineages of Plasmodium parasites originated roughly in the Oligocene epoch after the emergence of their hosts. This timing on the age of the common ancestor of extant Plasmodium parasites suggest the importance of host switches and lends support to the evolutionary scenario of a "malaria big bang" that was proposed based on the evolutionary analysis using the mitochondrial genome.


Assuntos
Extinção Biológica , Especiação Genética , Plasmodium/genética , Animais , Ciclo do Ácido Cítrico/genética , Complexo de Proteínas da Cadeia de Transporte de Elétrons/genética , Evolução Molecular , Genes de Protozoários , Genoma Mitocondrial , Glicólise/genética , Interações Hospedeiro-Parasita/genética , Humanos , Funções Verossimilhança , Modelos Genéticos , Filogenia , Análise de Sequência de DNA
20.
Vaccine ; 29(26): 4308-15, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21514344

RESUMO

Pfs230, surface protein of gametocyte/gamete of the human malaria parasite, Plasmodium falciparum, is a prime candidate of malaria transmission-blocking vaccine. Plasmodium vivax has an ortholog of Pfs230 (Pvs230), however, there has been no study in any aspects on Pvs230 to date. To investigate whether Pvs230 can be a vivax malaria transmission-blocking vaccine, we performed evolutionary and population genetic analysis of the Pvs230 gene (pvs230: PVX_003905). Our analysis of Pvs230 and its orthologs in eight Plasmodium species revealed two distinctive parts: an interspecies variable part (IVP) containing species-specific oligopeptide repeats at the N-terminus and a 7.5kb interspecies conserved part (ICP) containing 14 cysteine-rich domains. Pvs230 was closely related to its orthologs, Pks230 and Pcys230, in monkey malaria parasites. Analysis of 113 pvs230 sequences obtained from worldwide, showed that nucleotide diversity is remarkably low in the non-repeat 8-kb region of pvs230 (θπ=0.00118) with 77 polymorphic nucleotide sites, 40 of which results in amino acid replacements. A signature of purifying selection but not of balancing selection was seen on pvs230. Functional and/or structural constraints may limit the level of polymorphism in pvs230. The observed limited polymorphism in pvs230 should ground for utilization of Pvs230 as an effective transmission-blocking vaccine.


Assuntos
Sequência de Aminoácidos , Antígenos de Protozoários/imunologia , Sequência Conservada , Vacinas Antimaláricas/imunologia , Malária Vivax/prevenção & controle , Malária Vivax/transmissão , Plasmodium vivax/imunologia , Proteínas de Protozoários/imunologia , Animais , Antígenos de Protozoários/química , Antígenos de Protozoários/genética , DNA de Protozoário/análise , Humanos , Vacinas Antimaláricas/química , Vacinas Antimaláricas/genética , Malária Vivax/parasitologia , Dados de Sequência Molecular , Filogenia , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Alinhamento de Sequência , Análise de Sequência de DNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA