Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Microbes Infect ; 10(3): 260-8, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18316224

ABSTRACT

Type III secretion systems (T3SS) are present in many pathogenic gram-negative bacteria and mediate the translocation of bacterial effector proteins into host cells. Here, we report the phenotypic characterization of S. flexneri ipgB1 and ipgB2 mutants, in which the genes encoding the IpgB1 and IpgB2 effectors have been inactivated, either independently or simultaneously. Like IpgB1, we found that IpgB2 is secreted by the T3SS and its secretion requires the Spa15 chaperone. Upon infection of semi-confluent HeLa cells, the ipgB2 mutant exhibited the same invasive capacity as the wild-type strain and the ipgB1 mutant was 50% less invasive. Upon infection of polarised Caco2-cells, the ipgB2 mutant did not show a significant defect in invasion and the ipgB1 mutant was slightly more invasive than the wild-type strain. Entry of the ipgB1 ipgB2 mutant in polarized cells was reduced by 70% compared to the wild-type strain. Upon infection of the cornea in Guinea pigs, the ipgB2 mutant exhibited a wild-type phenotype, the ipgB1 mutant was hypervirulent and elicited a more pronounced proinflammatory response, while the ipgB1 ipgB2 mutant was highly attenuated. The attenuated phenotype of the ipgB1 ipgB2 mutant was confirmed using a murine pulmonary model of infection and histopathology and immunochemistry studies.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/metabolism , Dysentery, Bacillary/microbiology , Dysentery, Bacillary/pathology , Inflammation/pathology , Molecular Chaperones/metabolism , Shigella flexneri/metabolism , Shigella flexneri/pathogenicity , rac1 GTP-Binding Protein/metabolism , Amino Acid Sequence , Animals , Antigens, Bacterial , Caco-2 Cells , Female , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Point Mutation , Sequence Alignment , Virulence , rac1 GTP-Binding Protein/genetics
2.
Mol Microbiol ; 66(1): 139-50, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17725559

ABSTRACT

Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1), encoded by the var gene family, undergoes antigenic variation and plays an important role in chronic infection and severe malaria. Only a single var gene is transcribed per parasite, and epigenetic control mechanisms are fundamental in this strategy of mutually exclusive transcription. We show that subtelomeric upsB var gene promoters carried on episomes are silenced by default, and that promoter activation is sufficient to silence all other family members. However, they are active by default when placed downstream of a second active var promoter, underscoring the significance of local chromatin environment and nuclear compartmentalization in var promoter regulation. Native chromatin covering the SPE2-repeat array in upsB promoters is resistant to nuclease digestion, and insertion of these regulatory elements into a heterologous promoter causes local alterations in nucleosomal organization and promoter repression. Our findings suggest a common logic underlying the transcriptional control of all var genes, and have important implications for our understanding of the epigenetic processes involved in the regulation of this major virulence gene family.


Subject(s)
Chromatin/metabolism , Epigenesis, Genetic , Gene Expression Regulation , Plasmodium falciparum/genetics , Protozoan Proteins/biosynthesis , Animals , Blotting, Northern , Plasmids/genetics , Plasmodium falciparum/metabolism , RNA, Protozoan/biosynthesis
3.
Mol Microbiol ; 62(1): 72-83, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16942599

ABSTRACT

The malaria parasite Plasmodium falciparum undergoes antigenic variation through allelic exclusion and variant expression of surface proteins encoded by the var gene family. Regulation of var genes is under epigenetic control and involves reversible silencing and activation that requires the physical repositioning of a var locus into a transcriptionally permissive zone of the nuclear periphery. P. falciparum chromosome ends appear to aggregate into large perinuclear clusters which house both subtelomeric and chromosome central var genes. In this study we further define the composition of telomeric clusters using fluorescent in situ hybridization, and provide evidence that chromosome end clusters are formed by cross-linking protein. In addition, we demonstrate that a subtelomeric reporter gene and a var gene remain within clusters regardless of their transcriptional status. Our findings support a model whereby a highly localized structure dedicated to the activation of a single var gene can be housed within a gene dense chromosome end cluster that is otherwise transcriptionally silent.


Subject(s)
Chromosomes/genetics , Gene Silencing , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Animals , Cell Nucleus/metabolism , Chromosomes/metabolism , Electrophoresis, Gel, Two-Dimensional/methods , Gene Expression Regulation/genetics , In Situ Hybridization, Fluorescence/methods , Plasmodium falciparum/metabolism , Plasmodium falciparum/pathogenicity , Protozoan Proteins/metabolism , Telomere/genetics , Telomere/metabolism , Transcription, Genetic/genetics , Virulence/genetics
4.
Mol Biochem Parasitol ; 148(2): 117-24, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16631964

ABSTRACT

Malaria during pregnancy causes serious disease that is associated with sequestration in the placenta of Plasmodium falciparum infected erythrocytes that adhere to several host receptors, including chondroitin sulfate A (CSA). The principal CSA binding ligand associated with placental sequestration is the P. falciparum erythrocyte membrane protein 1 (PfEMP1), encoded by the var2csa gene. We disrupted the var2csa gene in two allogeneic parasites and ablated CSA binding. However, in one parasite line we were able to re-select for adhesion to bovine trachea CSA associated with transcription of two var genes, var-CS2 and varP. Parasites transcribing parts of var-CS2 and varP were present in the placentae of some infected women but the mutant parasites that transcribed var-CS2 and varP were recognized by sera from men and pregnant women independent of parity. This work raises the possibility that the PfEMP1 molecules encoded by var-CS2 and varP may be minor contributors to placental malaria but also confirms the importance of the immunodominant, conserved var2csa PfEMP1s in pregnancy associated malaria and strengthens the case for var2csa as a pregnancy-specific malaria vaccine.


Subject(s)
Antigens, Protozoan/metabolism , Chondroitin Sulfates/metabolism , Malaria, Falciparum/parasitology , Plasmodium falciparum/isolation & purification , Pregnancy Complications, Parasitic/parasitology , Animals , Antigens, Protozoan/genetics , Cattle , Cell Adhesion , Erythrocytes/parasitology , Female , Humans , Ligands , Malaria Vaccines , Male , Placenta/chemistry , Placenta/parasitology , Plasmodium falciparum/genetics , Pregnancy , Transfection
5.
Nature ; 439(7079): 1004-8, 2006 Feb 23.
Article in English | MEDLINE | ID: mdl-16382237

ABSTRACT

Mono-allelic expression of gene families is used by many organisms to mediate phenotypic variation of surface proteins. In the apicomplexan parasite Plasmodium falciparum, responsible for the severe form of malaria in humans, this is exemplified by antigenic variation of the highly polymorphic P. falciparum erythrocyte membrane protein 1 (PfEMP1). PfEMP1, encoded by the 60-member var gene family, represents a major virulence factor due to its central role in immune evasion and intravascular parasite sequestration. Mutually exclusive expression of PfEMP1 is controlled by epigenetic mechanisms involving chromatin modification and perinuclear var locus repositioning. Here we show that a var promoter mediates the nucleation and spreading of stably inherited silenced chromatin. Transcriptional activation of this promoter occurs at the nuclear periphery in association with chromosome-end clusters. Additionally, the var promoter sequence is sufficient to infiltrate a transgene into the allelic exclusion programme of var gene expression, as transcriptional activation of this transgene results in silencing of endogenous var gene transcription. These results show that a var promoter is sufficient for epigenetic silencing and mono-allelic transcription of this virulence gene family, and are fundamental for our understanding of antigenic variation in P. falciparum. Furthermore, the PfEMP1 knockdown parasites obtained in this study will be important tools to increase our understanding of P. falciparum-mediated virulence and immune evasion.


Subject(s)
Alleles , Gene Silencing , Malaria, Falciparum/parasitology , Plasmodium falciparum/genetics , Plasmodium falciparum/pathogenicity , Promoter Regions, Genetic/genetics , Virulence Factors/genetics , Animals , Animals, Genetically Modified , Antigenic Variation/genetics , Antigenic Variation/immunology , Cell Nucleus/metabolism , Epigenesis, Genetic/genetics , Genes, Protozoan/genetics , Malaria, Falciparum/immunology , Multigene Family/genetics , Plasmodium falciparum/immunology , Transcription, Genetic/genetics , Transfection , Transgenes/genetics , Virulence/genetics
6.
Cell ; 121(1): 13-24, 2005 Apr 08.
Article in English | MEDLINE | ID: mdl-15820675

ABSTRACT

The malaria parasite Plasmodium falciparum undergoes antigenic variation to evade host immune responses through switching expression of variant surface proteins encoded by the var gene family. We demonstrate that both a subtelomeric transgene and var genes are subject to reversible gene silencing. Var gene silencing involves the SIR complex as gene disruption of PfSIR2 results in activation of this gene family. We also demonstrate that perinuclear gene activation involves chromatin alterations and repositioning into a location that may be permissive for transcription. Together, this implies that locus repositioning and heterochromatic silencing play important roles in the epigenetic regulation of virulence genes in P. falciparum.


Subject(s)
Antigenic Variation/genetics , Cell Compartmentation/genetics , Chromatin Assembly and Disassembly/genetics , Gene Silencing/immunology , Heterochromatin/genetics , Malaria, Falciparum/parasitology , Plasmodium falciparum/genetics , Plasmodium falciparum/pathogenicity , Animals , Antigenic Variation/immunology , Cell Compartmentation/immunology , Cell Nucleus/genetics , Cell Nucleus/immunology , Chromosomes/genetics , Chromosomes/immunology , Genes, Protozoan/genetics , Genes, Protozoan/immunology , Heterochromatin/immunology , Humans , In Situ Hybridization, Fluorescence , Malaria, Falciparum/genetics , Microarray Analysis , Plasmids/genetics , Plasmids/immunology , Plasmodium falciparum/immunology , Telomere/genetics , Tetrahydrofolate Dehydrogenase/genetics , Virulence/genetics
SELECTION OF CITATIONS
SEARCH DETAIL