Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Microbiol ; 75(4): 957-71, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20487290

RESUMO

Plasmodium parasites possess a single pyruvate dehydrogenase (PDH) enzyme complex that is localized to the plastid-like organelle known as the apicoplast. Unlike most eukaryotes, Plasmodium parasites lack a mitochondrial PDH. The PDH complex catalyses the conversion of pyruvate to acetyl-CoA, an important precursor for the tricarboxylic acid cycle and type II fatty acid synthesis (FAS II). In this study, using a rodent malaria model, we show that the PDH E1 alpha and E3 subunits colocalize with the FAS II enzyme FabI in the apicoplast of liver stages but are not significantly expressed in blood stages. Deletion of the E1 alpha or E3 subunit genes of Plasmodium yoelii PDH caused no defect in blood stage development, mosquito stage development or early liver stage development. However, the gene deletions completely blocked the ability of the e1 alpha(-) and e3(-) parasites to form exo-erythrocytic merozoites during late liver stage development, thus preventing the initiation of a blood stage infection. This phenotype is similar to that observed for deletions of genes involved in FAS II elongation. The data strongly support the hypothesis that the sole role of PDH is to provide acetyl-CoA for FAS II.


Assuntos
Di-Hidrolipoamida Desidrogenase/metabolismo , Plasmodium yoelii/enzimologia , Piruvato Desidrogenase (Lipoamida)/metabolismo , Animais , Di-Hidrolipoamida Desidrogenase/genética , Eritrócitos/parasitologia , Feminino , Fígado/parasitologia , Malária/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Organismos Geneticamente Modificados , Plasmodium yoelii/genética , Plasmodium yoelii/crescimento & desenvolvimento , Plasmodium yoelii/metabolismo , Plastídeos/metabolismo , Piruvato Desidrogenase (Lipoamida)/genética
2.
Trends Parasitol ; 25(12): 545-50, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19819758

RESUMO

Fatty acids are essential components of membranes, and are also involved in cell signalling. Plasmodium, the parasite that causes malaria, scavenges fatty acids from its hosts. However, Plasmodium also possesses enzymes for a prokaryotic-like de novo fatty acid synthesis pathway, which resides in the apicoplast. Recent research has demonstrated that Plasmodium parasites depend on de novo fatty acid synthesis only for liver-stage development. This finding demonstrates that basic anabolic functions of Plasmodium parasites are not necessary for the growth and replication of every life cycle stage. We discuss the role of fatty acid metabolism in Plasmodium and why we believe that de novo fatty acid synthesis is only required for parasite late liver-stage development.


Assuntos
Ácidos Graxos/biossíntese , Plasmodium/fisiologia , Animais , Culicidae/parasitologia , Ácidos Graxos/metabolismo , Regulação Enzimológica da Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Estágios do Ciclo de Vida/fisiologia , Fígado/parasitologia , Malária/parasitologia , Plasmodium/enzimologia , Plasmodium/genética , Plasmodium/metabolismo
3.
BMC Genomics ; 10: 270, 2009 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-19534804

RESUMO

BACKGROUND: Plasmodium sporozoites migrate to the liver where they traverse several hepatocytes before invading the one inside which they will develop and multiply into thousands of merozoites. Although this constitutes an essential step of malaria infection, the requirements of Plasmodium parasites in liver cells and how they use the host cell for their own survival and development are poorly understood. RESULTS: To gain new insights into the molecular host-parasite interactions that take place during malaria liver infection, we have used high-throughput microarray technology to determine the transcriptional profile of P. berghei-infected hepatoma cells. The data analysis shows differential expression patterns for 1064 host genes starting at 6 h and up to 24 h post infection, with the largest proportion correlating specifically with the early stages of the infection process. A considerable proportion of those genes were also found to be modulated in liver cells collected from P. yoelii-infected mice 24 and 40 h after infection, strengthening the data obtained with the in vitro model and highlighting genes and pathways involved in the host response to rodent Plasmodium parasites. CONCLUSION: Our data reveal that host cell infection by Plasmodium sporozoites leads to a coordinated and sequential set of biological events, ranging from the initial stage of stress response up to the engagement of host metabolic processes and the maintenance of cell viability throughout infection.


Assuntos
Perfilação da Expressão Gênica , Interações Hospedeiro-Parasita/genética , Fígado/metabolismo , Malária/genética , Animais , Linhagem Celular Tumoral , Hepatócitos/metabolismo , Hepatócitos/parasitologia , Fígado/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sequência com Séries de Oligonucleotídeos , Plasmodium berghei , Esporozoítos , Fatores de Tempo
4.
PLoS One ; 4(2): e4480, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19214236

RESUMO

Immunization with radiation attenuated Plasmodium sporozoites (RAS) elicits sterile protective immunity against sporozoite challenge in murine models and in humans. Similarly to RAS, the genetically attenuated sporozoites (GAPs) named uis3(-), uis4(-) and P36p(-) have arrested growth during the liver stage development, and generate a powerful protective immune response in mice. We compared the protective mechanisms in P. yoelii RAS, uis3(-) and uis4(-) in BALB/c mice. In RAS and GAPs, sterile immunity is only achieved after one or more booster injections. There were no differences in the immune responses to the circumsporozoite protein (CSP) generated by RAS and GAPs. To evaluate the role of non-CSP T-cell antigens we immunized antibody deficient, CSP-transgenic BALB/c mice, that are T cell tolerant to CSP, with P. yoelii RAS or with uis3(-) or uis4(-) GAPs, and challenged them with wild type sporozoites. In every instance the parasite liver stage burden was approximately 3 logs higher in antibody deficient CSP transgenic mice as compared to antibody deficient mice alone. We conclude that CSP is a powerful protective antigen in both RAS and GAPs viz., uis3(-) and uis4(-) and that the protective mechanisms are similar independently of the method of sporozoite attenuation.


Assuntos
Plasmodium , Esporozoítos , Animais , Antígenos de Protozoários/imunologia , Humanos , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Plasmodium/genética , Plasmodium/imunologia , Plasmodium/efeitos da radiação , Vacinas Protozoárias/imunologia , Esporozoítos/imunologia , Esporozoítos/efeitos da radiação
5.
Cell Microbiol ; 11(3): 506-20, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19068099

RESUMO

Intracellular malaria parasites require lipids for growth and replication. They possess a prokaryotic type II fatty acid synthesis (FAS II) pathway that localizes to the apicoplast plastid organelle and is assumed to be necessary for pathogenic blood stage replication. However, the importance of FAS II throughout the complex parasite life cycle remains unknown. We show in a rodent malaria model that FAS II enzymes localize to the sporozoite and liver stage apicoplast. Targeted deletion of FabB/F, a critical enzyme in fatty acid synthesis, did not affect parasite blood stage replication, mosquito stage development and initial infection in the liver. This was confirmed by knockout of FabZ, another critical FAS II enzyme. However, FAS II-deficient Plasmodium yoelii liver stages failed to form exo-erythrocytic merozoites, the invasive stage that first initiates blood stage infection. Furthermore, deletion of FabI in the human malaria parasite Plasmodium falciparum did not show a reduction in asexual blood stage replication in vitro. Malaria parasites therefore depend on the intrinsic FAS II pathway only at one specific life cycle transition point, from liver to blood.


Assuntos
Sangue/parasitologia , Ácidos Graxos/biossíntese , Fígado/parasitologia , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/metabolismo , Plasmodium yoelii/crescimento & desenvolvimento , Plasmodium yoelii/metabolismo , Animais , Enzimas/genética , Enzimas/metabolismo , Feminino , Deleção de Genes , Técnicas de Inativação de Genes , Humanos , Malária/parasitologia , Redes e Vias Metabólicas , Camundongos , Camundongos Endogâmicos BALB C , Organelas/química , Plasmodium falciparum/química , Plasmodium yoelii/química , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo
6.
Mol Cell Biol ; 28(20): 6196-207, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18710954

RESUMO

The malaria parasite sporozoite transmission stage develops and differentiates within parasite oocysts on the Anopheles mosquito midgut. Successful inoculation of the parasite into a mammalian host is critically dependent on the sporozoite's ability to first infect the mosquito salivary glands. Remarkable changes in tissue infection competence are observed as the sporozoites transit from the midgut oocysts to the salivary glands. Our microarray analysis shows that compared to oocyst sporozoites, salivary gland sporozoites upregulate expression of at least 124 unique genes. Conversely, oocyst sporozoites show upregulation of at least 47 genes (upregulated in oocyst sporozoites [UOS genes]) before they infect the salivary glands. Targeted gene deletion of UOS3, encoding a putative transmembrane protein with a thrombospondin repeat that localizes to the sporozoite secretory organelles, rendered oocyst sporozoites unable to infect the mosquito salivary glands but maintained the parasites' liver infection competence. This phenotype demonstrates the significance of differential UOS expression. Thus, the UIS-UOS gene classification provides a framework to elucidate the infectivity and transmission success of Plasmodium sporozoites on a whole-genome scale. Genes identified herein might represent targets for vector-based transmission blocking strategies (UOS genes), as well as strategies that prevent mammalian host infection (UIS genes).


Assuntos
Anopheles/parasitologia , Insetos Vetores/parasitologia , Malária/genética , Malária/parasitologia , Mamíferos/parasitologia , Esporozoítos/metabolismo , Transcrição Gênica , Animais , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Marcação de Genes , Genes de Protozoários , Hemolinfa/citologia , Hemolinfa/metabolismo , Interações Hospedeiro-Parasita , Injeções Intravenosas , Camundongos , Oocistos/citologia , Oocistos/metabolismo , Parasitos/citologia , Parasitos/genética , Parasitos/patogenicidade , Plasmodium falciparum/genética , Plasmodium yoelii/genética , Transporte Proteico , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Glândulas Salivares/parasitologia , Esporozoítos/citologia
7.
Bioinformatics ; 24(13): i383-9, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18586738

RESUMO

MOTIVATION: The sequencing of the Plasmodium yoelii genome, a model rodent malaria parasite, has greatly facilitated research for the development of new drug and vaccine candidates against malaria. Unfortunately, only preliminary gene models were annotated on the partially sequenced genome, mostly by in silico gene prediction, and there has been no major improvement of the annotation since 2002. RESULTS: Here we report on a systematic assessment of the accuracy of the genome annotation based on a detailed analysis of a comprehensive set of cDNA sequences and proteomics data. We found that the coverage of the current annotation tends to be biased toward genes expressed in the blood stages of the parasite life cycle. Based on our proteomic analysis, we estimate that about 15% of the liver stage proteome data we have generated is absent from the current annotation. Through comparative analysis we identified and manually curated a further 510 P. yoelii genes which have clear orthologs in the P. falciparum genome, but were not present or incorrectly annotated in the current annotation. This study suggests that improvements of the current P. yoelii genome annotation should focus on genes expressed in stages other than blood stages. Comparative analysis will be critically helpful for this re-annotation. The addition of newly annotated genes will facilitate the use of P. yoelii as a model system for studying human malaria. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Algoritmos , Mapeamento Cromossômico/métodos , Genoma de Protozoário/genética , Plasmodium yoelii/genética , Alinhamento de Sequência/métodos , Análise de Sequência de DNA/métodos , Animais , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
8.
Proc Natl Acad Sci U S A ; 105(1): 305-10, 2008 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-18172196

RESUMO

For 50 years since their discovery, the malaria parasite liver stages (LS) have been difficult to analyze, impeding their utilization as a critical target for antiinfection vaccines and drugs. We have undertaken a comprehensive transcriptome analysis in combination with a proteomic survey of LS. Green fluorescent protein-tagged Plasmodium yoelii (PyGFP) was used to efficiently isolate LS-infected hepatocytes from the rodent host. Genome-wide LS gene expression was profiled and compared with other parasite life cycle stages. The analysis revealed approximately 2,000 genes active during LS development, and proteomic analysis identified 816 proteins. A subset of proteins appeared to be expressed in LS only. The data revealed exported parasite proteins and LS metabolic pathways including expression of FASII pathway enzymes. The FASII inhibitor hexachlorophene and the antibiotics, tetracycline and rifampicin, that target the apicoplast inhibited LS development, identifying FASII and other pathways localized in the apicoplast as potential drug targets to prevent malaria infection.


Assuntos
Fígado/parasitologia , Malária/parasitologia , Proteômica/métodos , Transcrição Gênica , Animais , Desenho de Fármacos , Ácidos Graxos/metabolismo , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/química , Hepatócitos/parasitologia , Humanos , Fases de Leitura Aberta , Plasmodium yoelii/metabolismo , Proteoma
9.
J Infect Dis ; 196(4): 608-16, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17624848

RESUMO

Irradiation-attenuated sporozoite vaccinations confer sterile protection against malaria infection in animal models and humans. Persistent, nonreplicating parasite forms in the liver are presumably necessary for the maintenance of sterile immunity. A novel vaccine approach uses genetically attenuated parasites (GAPs) that undergo arrested development during liver infection. The fate of GAPs after immunization, their persistence in vaccinated animals, and the immune mechanisms that mediate protection are unknown. To examine the developmental defects of genetically attenuated liver stages in vivo, we created deletions of the UIS3 and UIS4 loci in the Plasmodium yoelii rodent malaria model (Pyuis3[-] and Pyuis4[-]). The low 50% infectious dose of P. yoelii in BALB/c mice provides the most sensitive infectivity model. We show that P. yoelii GAPs reach the liver, invade hepatocytes, and develop a parasitophorous vacuole but do not significantly persist 40 h after infection. A single dose of Pyuis4(-) sporozoites conferred complete protection, but full protection by Pyuis3(-) sporozoites required at least 2 immunizations. CD8(+) T cells were essential for protection, but CD4(+) T cells were not. Our results show that genetically distinct GAPs confer different degrees of protective efficacy and that live vaccine persistence in the liver is not necessary to sustain long-lasting protection. These findings have important implications for the development of a P. falciparum GAP malaria vaccine.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Fígado/imunologia , Fígado/parasitologia , Vacinas Antimaláricas/genética , Malária/prevenção & controle , Plasmodium yoelii , Esporozoítos/imunologia , Vacinação , Animais , Feminino , Deleção de Genes , Hepatócitos/parasitologia , Hepatócitos/patologia , Esquemas de Imunização , Injeções Intravenosas , Malária/imunologia , Malária/parasitologia , Vacinas Antimaláricas/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Plasmodium yoelii/genética , Plasmodium yoelii/crescimento & desenvolvimento , Plasmodium yoelii/imunologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacúolos/parasitologia , Vacúolos/patologia
10.
Mycopathologia ; 163(2): 97-104, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17286166

RESUMO

The plasmid, pAF28, a genomic clone from Aspergillus flavus NRRL 6541, has been used as a hybridization probe to fingerprint A. flavus strains isolated in corn and peanut fields. The insert of pAF28 contains a 4.5 kb region which encodes a truncated retrotransposon (AfRTL-1). In search for a full-length and intact copy of retrotransposon, we exploited a novel PCR cloning strategy by amplifying a 3.4 kb region from the genomic DNA of A. flavus NRRL 6541. The fragment was cloned into pCR 4-TOPO. Sequence analysis confirmed that this region encoded putative domains of partial reverse transcriptase, RNase H, and integrase of the predicted retrotransposon. The two flanking long terminal repeats (LTRs) and the sequence between them comprise a putative full-length LTR retrotransposon of 7799 bp in length. This intact retrotransposon sequence is named AFLAV (A. flavus Retrotransposon). The order of the predicted catalytic domains in the polyprotein (Pol) placed AFLAV in the Tf1/sushi subgroup of the Ty3/gypsy retrotransposon family. Primers derived from AFLAV sequence were used to screen this retrotransposon in other strains of A. flavus. More than fifty strains of A. flavus isolated from different geological origins were surveyed and the results show that many strains have extensive deletions in the regions encoding the capsid (Gag) and Pol.


Assuntos
Aspergillus flavus/genética , DNA Fúngico/genética , Retroelementos/genética , Arachis/microbiologia , Sequência de Bases , Southern Blotting , DNA Fúngico/química , Dados de Sequência Molecular , Plasmídeos/genética , Reação em Cadeia da Polimerase/métodos , Análise de Sequência de DNA , Zea mays/microbiologia
11.
Int J Parasitol ; 36(12): 1283-93, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16890231

RESUMO

The liver stages of Plasmodium, the causative agent of malaria, are the least explored forms in the parasite's life cycle despite their recognition as key vaccine and drug targets. In vivo experimental access to liver stages of human malaria parasites is practically prohibited and therefore rodent model malaria parasites have been used for in vivo studies. However, even in rodent models progress in the analysis of liver stages has been limited, mainly due to their low abundance and associated difficulties in visualisation and isolation. Here, we present green fluorescent protein (GFP)-tagged Plasmodium yoelii rodent malaria parasite liver infections in BALB/c mice as an excellent quantitative model for the live visualisation and isolation of the so far elusive liver stages. We believe P. yoelii GFP-tagged liver stages allow, for the first time, the efficient quantitative isolation of intact early and late liver stage-infected hepatocyte units by fluorescence activated cell sorting. GFP-tagged liver stages are also well suited for intravital imaging, allowing us for the first time to visualise them in real time. We identify previously unrecognised features of liver stages including vigorous parasite movement and expulsion of 'extrusomes'. Intravital imaging thus reveals new, important information on the malaria parasite's transition from tissue to blood stage.


Assuntos
Hepatopatias Parasitárias/parasitologia , Malária/parasitologia , Plasmodium yoelii/isolamento & purificação , Animais , Animais Geneticamente Modificados , Linhagem Celular , Modelos Animais de Doenças , Feminino , Citometria de Fluxo/métodos , Genótipo , Proteínas de Fluorescência Verde , Hepatócitos/parasitologia , Fígado/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência/métodos , Fenótipo , Plasmodium berghei/genética , Plasmodium berghei/isolamento & purificação , Plasmodium yoelii/genética , Transfecção/métodos
12.
Plant Physiol ; 135(1): 377-83, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-14671009

RESUMO

A major goal of phytoremediation is to transform fast-growing plants with genes from plant species that hyperaccumulate toxic trace elements. We overexpressed the gene encoding selenocysteine methyltransferase (SMT) from the selenium (Se) hyperaccumulator Astragalus bisulcatus in Arabidopsis and Indian mustard (Brassica juncea). SMT detoxifies selenocysteine by methylating it to methylselenocysteine, a nonprotein amino acid, thereby diminishing the toxic misincorporation of Se into protein. Our Indian mustard transgenic plants accumulated more Se in the form of methylselenocysteine than the wild type. SMT transgenic seedlings tolerated Se, particularly selenite, significantly better than the wild type, producing 3- to 7-fold greater biomass and 3-fold longer root lengths. Moreover, SMT plants had significantly increased Se accumulation and volatilization. This is the first study, to our knowledge, in which a fast-growing plant was genetically engineered to overexpress a gene from a hyperaccumulator in order to increase phytoremediation potential.


Assuntos
Adaptação Fisiológica/genética , Arabidopsis/enzimologia , Cisteína/análogos & derivados , Metiltransferases/genética , Mostardeira/enzimologia , Selênio/metabolismo , Adaptação Fisiológica/fisiologia , Arabidopsis/genética , Cisteína/metabolismo , Regulação Enzimológica da Expressão Gênica , Regulação da Expressão Gênica de Plantas , Metiltransferases/metabolismo , Dados de Sequência Molecular , Mostardeira/genética , Compostos Organosselênicos/metabolismo , Plantas Geneticamente Modificadas , Selenocisteína/análogos & derivados , Volatilização
13.
Chem Senses ; 28(7): 621-9, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14578124

RESUMO

Carbonic anhydrase (CA) is physiologically important in the reversible hydration reaction of CO(2); it is expressed in a number of isoforms (CA I-XIV) with varying degrees of enzymatic activity. In nasal chemesthesis, CA inhibition decreases the electrophysiologic response to CO(2), a common irritant test compound. CA enzymatic activity has been demonstrated in the human nasal mucosa using enzyme histochemical methods, but no systematic study of nasal mucosal CA isoenzyme gene expression has been published. We examined CA gene expression in superficial nasal mucosal scrapings from 15 subjects (6 females; 6 allergic rhinitics; age range, 21-56 years). Both non-quantitative and quantitative reverse transcription polymerase chain reaction (RT-PCR) were performed using primers for each gene coding for the 11 catalytically active CA isoenzymes and the housekeeping gene GADPH. Amplification products of GADPH and 10 of the 11 CA genes were detected in the specimens (CA VA was not detected). Relative expression of the CA genes was quantified using real-time PCR. Averaged across subjects, the relative abundance of the CA isoenzyme transcripts is as follows: CA XII > CA II > CA VB > CA IV > CA IX > CA III > CA XIV > CA I > CA VI > CA VII. Limited qualitative validation of gene expression was obtained by immunohistochemistry for CA I, CA II and CA IV. We also observed inter-individual variability in the expression of CA isoenzymes in human nasal mucosa, potentially contributing to differences in nasal chemosensitivity to CO(2) between individuals


Assuntos
Anidrases Carbônicas/genética , Expressão Gênica , Isoenzimas/genética , Mucosa Nasal/enzimologia , Adolescente , Adulto , Idoso , Feminino , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade
14.
Mycol Res ; 107(Pt 7): 841-6, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12967211

RESUMO

A genomic clone of the aflatoxin-producing fungus Aspergillus flavus, designated pAF28, has been used as a probe for Southern blot fingerprinting of fungal strains. A large number of A. flavus strains isolated from corn fields and tree-nut orchards can be distinguished because the DNA fingerprint patterns are highly polymorphic. We have completed the sequencing of a 6355 bp insert in pAF28. The sequence features motifs and open reading frames characteristic of transposable elements of the gypsy class. We have named this new element AfRTL-1, for A. flavus retrotransposon-like DNA.


Assuntos
Aspergillus flavus/classificação , Elementos de DNA Transponíveis/genética , Genoma Fúngico , Sequência de Aminoácidos , Aspergillus flavus/genética , Clonagem Molecular , Dados de Sequência Molecular , Alinhamento de Sequência , Análise de Sequência de DNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...