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.
Viruses ; 13(9)2021 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-34578379

RESUMO

A variety of immunolabeling procedures for both light and electron microscopy were used to examine the cellular origins of the host membranes supporting the SARS-CoV-2 replication complex. The endoplasmic reticulum has long been implicated as a source of membrane for the coronavirus replication organelle. Using dsRNA as a marker for sites of viral RNA synthesis, we provide additional evidence supporting ER as a prominent source of membrane. In addition, we observed a rapid fragmentation of the Golgi apparatus which is visible by 6 h and complete by 12 h post-infection. Golgi derived lipid appears to be incorporated into the replication organelle although protein markers are dispersed throughout the infected cell. The mechanism of Golgi disruption is undefined, but chemical disruption of the Golgi apparatus by brefeldin A is inhibitory to viral replication. A search for an individual SARS-CoV-2 protein responsible for this activity identified at least five viral proteins, M, S, E, Orf6, and nsp3, that induced Golgi fragmentation when expressed in eukaryotic cells. Each of these proteins, as well as nsp4, also caused visible changes to ER structure as shown by correlative light and electron microscopy (CLEM). Collectively, these results imply that specific disruption of the Golgi apparatus is a critical component of coronavirus replication.


Assuntos
Retículo Endoplasmático/virologia , Complexo de Golgi/virologia , SARS-CoV-2/fisiologia , Replicação Viral , Animais , Chlorocebus aethiops , Proteínas M de Coronavírus/fisiologia , Proteínas M de Coronavírus/ultraestrutura , Retículo Endoplasmático/ultraestrutura , Complexo de Golgi/ultraestrutura , Humanos , Membranas Intracelulares/ultraestrutura , Membranas Intracelulares/virologia , Microscopia Eletrônica , SARS-CoV-2/ultraestrutura , Células Vero , Proteínas Estruturais Virais/fisiologia , Proteínas Estruturais Virais/ultraestrutura
2.
Cell Rep ; 26(12): 3380-3390.e5, 2019 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-30893609

RESUMO

Chlamydial infection requires the formation of a membrane-bound vacuole, termed the inclusion, that undergoes extensive interactions with select host organelles. The importance of the Inc protein CT229 in the formation and maintenance of the chlamydial inclusion was recently highlighted by studies demonstrating that its absence during infection results in reduced bacterial replication, premature inclusion lysis, and host cell death. Previous reports have indicated that CT229 binds Rab GTPases; however, the physiological implications of this interaction are unknown. Here, we show that CT229 regulates host multivesicular trafficking by recruiting multiple Rab GTPases and their cognate effectors to the inclusion. We demonstrate that CT229 specifically modulates clathrin-coated vesicle trafficking and regulates the trafficking of transferrin and the mannose-6-phosphate receptor, both of which are crucial for proper chlamydial development. This study highlights CT229 as a master regulator of multiple host vesicular trafficking pathways essential for chlamydial infection.


Assuntos
Proteínas de Bactérias/metabolismo , Infecções por Chlamydia/metabolismo , Chlamydia trachomatis/metabolismo , Vesículas Revestidas por Clatrina/metabolismo , Vacúolos/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas de Bactérias/genética , Transporte Biológico Ativo , Infecções por Chlamydia/genética , Infecções por Chlamydia/patologia , Chlamydia trachomatis/genética , Vesículas Revestidas por Clatrina/genética , Vesículas Revestidas por Clatrina/microbiologia , Células HeLa , Humanos , Corpos de Inclusão/genética , Corpos de Inclusão/metabolismo , Corpos de Inclusão/microbiologia , Vacúolos/genética , Vacúolos/microbiologia , Proteínas rab de Ligação ao GTP/genética
3.
mBio ; 9(3)2018 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-29946049

RESUMO

Strains of Rickettsia rickettsii, the tick-borne agent of Rocky Mountain spotted fever, vary considerably in virulence. Genomic comparisons of R. rickettsii strains have identified a relatively small number of genes divergent in an avirulent strain. Among these is one annotated as Rickettsia ankyrin repeat protein 2 (RARP-2). Homologs of RARP-2 are present in all strains of R. rickettsii, but the protein in the avirulent strain Iowa contains a large internal deletion relative to the virulent Sheila Smith strain. RARP-2 is secreted in a type IV secretion system-dependent manner and exposed to the host cell cytosol. RARP-2 of Sheila Smith colocalizes with multilamellar membranous structures bearing markers of the endoplasmic reticulum (ER), whereas the Iowa protein shows no colocalization with host cell organelles and evidence of proteolytic degradation is detected. Overexpression of Sheila Smith RARP-2 in R. rickettsii Iowa converts this avirulent strain's typically nonlytic or opaque plaque type to a lytic plaque phenotype similar to that of the virulent Sheila Smith strain. Mutation of a predicted proteolytic active site of Sheila Smith RARP-2 abolished the lytic plaque phenotype but did not eliminate association with host membrane. RARP-2 is thus a type IV secreted effector and released from the rickettsiae into the host cytosol to modulate host processes during infection. Overexpression of Sheila Smith RARP-2 did not, however, restore the virulence of the Iowa strain in a guinea pig model, likely due to the multifactorial nature of rickettsial virulence.IMPORTANCE Members of the genus Rickettsia are obligate intracellular bacteria that exhibit a range of virulence from harmless endosymbionts of arthropods to the etiologic agents of severe disease. Despite the growing number of available genomes, little is known regarding virulence determinants of rickettsiae. Here, we have characterized an ankyrin repeat-containing protein, RARP-2, which differs between a highly virulent and an avirulent strain of R. rickettsii, the agent of Rocky Mountain spotted fever. RARP-2 is secreted by a type IV secretion system into the cytosol of the host cell, where it interacts with and manipulates the structure of the endoplasmic reticulum. RARP-2 from the avirulent strain is truncated by the loss of seven of 10 ankyrin repeat units but, although secreted, fails to alter ER structure. Recognition of those rickettsial factors associated with virulence will facilitate understanding of regional and strain-specific variation in severity of disease.


Assuntos
Proteínas de Bactérias/metabolismo , Retículo Endoplasmático/metabolismo , Rickettsia rickettsii/metabolismo , Sistemas de Secreção Tipo IV/metabolismo , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Retículo Endoplasmático/genética , Feminino , Cobaias , Humanos , Transporte Proteico , Rickettsia rickettsii/química , Rickettsia rickettsii/genética , Rickettsia rickettsii/patogenicidade , Febre Maculosa das Montanhas Rochosas/microbiologia , Sistemas de Secreção Tipo IV/química , Sistemas de Secreção Tipo IV/genética , Virulência
4.
Cell Rep ; 19(7): 1406-1417, 2017 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-28514660

RESUMO

Chlamydia trachomatis is a human pathogen associated with significant morbidity worldwide. As obligate intracellular parasites, chlamydiae must survive within eukaryotic cells for sufficient time to complete their developmental cycle. To promote host cell survival, chlamydiae express poorly understood anti-apoptotic factors. Using recently developed genetic tools, we show that three inclusion membrane proteins (Incs) out of eleven examined are required for inclusion membrane stability and avoidance of host cell death pathways. In the absence of specific Incs, premature inclusion lysis results in recognition by autophagolysosomes, activation of intrinsic apoptosis, and premature termination of the chlamydial developmental cycle. Inhibition of autophagy or knockdown of STING prevented host cell death and activation of intrinsic apoptosis. Significantly, these findings emphasize the importance of Incs in the establishment of a replicative compartment that sequesters the pathogen from host surveillance systems.


Assuntos
Proteínas de Bactérias/metabolismo , Chlamydia trachomatis/metabolismo , Interações Hospedeiro-Patógeno , Corpos de Inclusão/metabolismo , Proteínas de Membrana/metabolismo , Autofagossomos/metabolismo , Autofagia , Morte Celular , Chlamydia trachomatis/crescimento & desenvolvimento , Células HeLa , Humanos , Mutação/genética , Biossíntese de Proteínas , Solubilidade , Vesículas Transportadoras/metabolismo
5.
mBio ; 8(3)2017 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-28465429

RESUMO

The intracellular bacterium Chlamydia trachomatis develops in a parasitic compartment called the inclusion. Posttranslationally modified microtubules encase the inclusion, controlling the positioning of Golgi complex fragments around the inclusion. The molecular mechanisms by which Chlamydia coopts the host cytoskeleton and the Golgi complex to sustain its infectious compartment are unknown. Here, using a genetically modified Chlamydia strain, we discovered that both posttranslationally modified microtubules and Golgi complex positioning around the inclusion are controlled by the chlamydial inclusion protein CT813/CTL0184/InaC and host ARF GTPases. CT813 recruits ARF1 and ARF4 to the inclusion membrane, where they induce posttranslationally modified microtubules. Similarly, both ARF isoforms are required for the repositioning of Golgi complex fragments around the inclusion. We demonstrate that CT813 directly recruits ARF GTPases on the inclusion membrane and plays a pivotal role in their activation. Together, these results reveal that Chlamydia uses CT813 to hijack ARF GTPases to couple posttranslationally modified microtubules and Golgi complex repositioning at the inclusion.IMPORTANCEChlamydia trachomatis is an important cause of morbidity and a significant economic burden in the world. However, how Chlamydia develops its intracellular compartment, the so-called inclusion, is poorly understood. Using genetically engineered Chlamydia mutants, we discovered that the effector protein CT813 recruits and activates host ADP-ribosylation factor 1 (ARF1) and ARF4 to regulate microtubules. In this context, CT813 acts as a molecular platform that induces the posttranslational modification of microtubules around the inclusion. These cages are then used to reposition the Golgi complex during infection and promote the development of the inclusion. This study provides the first evidence that ARF1 and ARF4 play critical roles in controlling posttranslationally modified microtubules around the inclusion and that Chlamydia trachomatis hijacks this novel function of ARF to reposition the Golgi complex.


Assuntos
Proteínas de Bactérias/metabolismo , Chlamydia trachomatis/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Complexo de Golgi/metabolismo , Microtúbulos/metabolismo , Fator 1 de Ribosilação do ADP/metabolismo , Fatores de Ribosilação do ADP/metabolismo , Actinas , Proteínas de Bactérias/genética , Chlamydia trachomatis/genética , Complexo de Golgi/ultraestrutura , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Corpos de Inclusão/microbiologia , Microtúbulos/genética , Processamento de Proteína Pós-Traducional
6.
Biochem Biophys Res Commun ; 462(2): 165-70, 2015 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-25944661

RESUMO

Chlamydia trachomatis actively subverts the minus-end directed microtubule motor, dynein, to traffic along microtubule tracks to the Microtubule Organizing Center (MTOC) where it remains within a membrane bound replicative vacuole for the duration of its intracellular development. Unlike most substrates of the dynein motor, disruption of the dynactin cargo-linking complex by over-expression of the p50 dynamitin subunit does not inhibit C. trachomatis transport. A requirement for chlamydial protein synthesis to initiate this process suggests that a chlamydial product supersedes a requirement for p50 dynamitin. A yeast 2-hybrid system was used to screen the chlamydia inclusion membrane protein CT850 against a HeLa cell cDNA library and identified an interaction with the dynein light chain DYNLT1 (Tctex1). This interaction was at least partially dependent upon an (R/K-R/K-X-X-R/K) motif that is characteristic of DYNLT1 binding domains. CT850 expressed ectopically in HeLa cells localized at the MTOC and this localization is similarly dependent upon the predicted DYNLT1 binding domain. Furthermore, DYNLT1 is enriched at focal concentrations of CT850 on the chlamydial inclusion membrane that are known to interact with dynein and microtubules. Depletion of DYNLT1 disrupts the characteristic association of the inclusion membrane with centrosomes. Collectively, the results suggest that CT850 interacts with DYNLT1 to promote appropriate positioning of the inclusion at the MTOC.


Assuntos
Proteínas de Bactérias/metabolismo , Chlamydia trachomatis/metabolismo , Dineínas/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Chlamydia trachomatis/genética , Dineínas/química , Dineínas/genética , Técnicas de Silenciamento de Genes , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Centro Organizador dos Microtúbulos/metabolismo , Domínios e Motivos de Interação entre Proteínas , RNA Interferente Pequeno/genética , Técnicas do Sistema de Duplo-Híbrido
7.
PLoS One ; 8(5): e63426, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23696825

RESUMO

The chlamydial inclusion membrane is extensively modified by the insertion of type III secreted effector proteins. These inclusion membrane proteins (Incs) are exposed to the cytosol and share a common structural feature of a long, bi-lobed hydrophobic domain but little or no primary amino acid sequence similarity. Based upon secondary structural predictions, over 50 putative inclusion membrane proteins have been identified in Chlamydia trachomatis. Only a limited number of biological functions have been defined and these are not shared between chlamydial species. Here we have ectopically expressed several C. trachomatis Incs in HeLa cells and find that they induce the formation of morphologically distinct membranous vesicular compartments. Formation of these vesicles requires the bi-lobed hydrophobic domain as a minimum. No markers for various cellular organelles were observed in association with these vesicles. Lipid probes were incorporated by the Inc-induced vesicles although the lipids incorporated were dependent upon the specific Inc expressed. Co-expression of Inc pairs indicated that some colocalized in the same vesicle, others partially overlapped, and others did not associate at all. Overall, it appears that Incs may have an intrinsic ability to induce membrane formation and that individual Incs can induce membranous structures with unique properties.


Assuntos
Proteínas de Bactérias/metabolismo , Estruturas da Membrana Celular/metabolismo , Chlamydia trachomatis , Proteínas de Membrana/metabolismo , Proteínas de Bactérias/química , Células HeLa , Humanos , Interações Hidrofóbicas e Hidrofílicas , Corpos de Inclusão/metabolismo , Corpos de Inclusão/ultraestrutura , Membranas Intracelulares/metabolismo , Lipídeos de Membrana , Proteínas de Membrana/química , Estrutura Terciária de Proteína , Transporte Proteico
8.
Microbiology (Reading) ; 157(Pt 3): 830-838, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21109560

RESUMO

Chlamydia trachomatis is an obligate intracellular pathogen that replicates within a parasitophorous vacuole termed an inclusion. The chlamydial inclusion is isolated from the endocytic pathway but fusogenic with Golgi-derived exocytic vesicles containing sphingomyelin and cholesterol. Sphingolipids are incorporated into the chlamydial cell wall and are considered essential for chlamydial development and viability. The mechanisms by which chlamydiae obtain eukaryotic lipids are poorly understood but require chlamydial protein synthesis and presumably modification of the inclusion membrane to initiate this interaction. A polarized cell model of chlamydial infection has demonstrated that chlamydiae preferentially intercept basolaterally directed, sphingomyelin-containing exocytic vesicles. Here we examine the localization and potential function of trans-Golgi and/or basolaterally associated soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins in chlamydia-infected cells. The trans-Golgi SNARE protein syntaxin 6 is recruited to the chlamydial inclusion in a manner that requires chlamydial protein synthesis and is conserved among all chlamydial species examined. The localization of syntaxin 6 to the chlamydial inclusion requires a tyrosine motif or plasma membrane retrieval signal (YGRL). Thus in addition to expression of at least two inclusion membrane proteins that contain SNARE-like motifs, chlamydiae also actively recruit eukaryotic SNARE-family proteins.


Assuntos
Chlamydia trachomatis/metabolismo , Corpos de Inclusão/metabolismo , Membranas Intracelulares/metabolismo , Proteínas Qa-SNARE/metabolismo , Linhagem Celular , Chlamydia trachomatis/genética , Chlamydia trachomatis/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas Qa-SNARE/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
9.
PLoS Pathog ; 6(7): e1000997, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20657821

RESUMO

The translocated actin recruiting phosphoprotein (Tarp) is conserved among all pathogenic chlamydial species. Previous reports identified single C. trachomatis Tarp actin binding and proline rich domains required for Tarp mediated actin nucleation. A peptide antiserum specific for the Tarp actin binding domain was generated and inhibited actin polymerization in vitro and C. trachomatis entry in vivo, indicating an essential role for Tarp in chlamydial pathogenesis. Sequence analysis of Tarp orthologs from additional chlamydial species and C. trachomatis serovars indicated multiple putative actin binding sites. In order to determine whether the identified actin binding domains are functionally conserved, GST-Tarp fusions from multiple chlamydial species were examined for their ability to bind and nucleate actin. Chlamydial Tarps harbored variable numbers of actin binding sites and promoted actin nucleation as determined by in vitro polymerization assays. Our findings indicate that Tarp mediated actin binding and nucleation is a conserved feature among diverse chlamydial species and this function plays a critical role in bacterial invasion of host cells.


Assuntos
Actinas/antagonistas & inibidores , Chlamydia/patogenicidade , Internalização do Vírus , Proteínas de Bactérias/fisiologia , Sítios de Ligação , Infecções por Chlamydia/etiologia , Células HeLa/microbiologia , Humanos , Ligação Proteica , Virulência
10.
Biochem Biophys Res Commun ; 371(2): 339-44, 2008 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-18442471

RESUMO

The translocated actin recruiting phosphoprotein (Tarp) is injected into the cytosol shortly after Chlamydia trachomatis attachment to a target cell and subsequently phosphorylated by an unidentified tyrosine kinase. A role for Tarp phosphorylation in bacterial entry is unknown. In this study, recombinant C. trachomatis Tarp was employed to identify the host cell kinase(s) required for phosphorylation. Each tyrosine rich repeat of L2 Tarp harbors a sequence similar to a Src and Abl kinase consensus target. Furthermore, purified p60-src, Yes, Fyn, and Abl kinases were able to phosphorylate Tarp. Mutagenesis of potential tyrosines within a single tyrosine rich repeat peptide indicated that both Src and Abl kinases phosphorylate the same residues suggesting that C. trachomatis Tarp may serve as a substrate for multiple host cell kinases. Surprisingly, chemical inhibition of Src and Abl kinases prevented Tarp phosphorylation in culture and had no measurable effect on bacterial entry into host cells.


Assuntos
Proteínas de Bactérias/metabolismo , Chlamydia trachomatis/metabolismo , Fosfoproteínas/metabolismo , Quinases da Família src/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Células HeLa , Humanos , Dados de Sequência Molecular , Fosfoproteínas/genética , Fosforilação , Mutação Puntual
11.
Cell Microbiol ; 9(9): 2278-88, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17501982

RESUMO

Chlamydiae are Gram-negative obligate intracellular pathogens to which access to an intracellular environment is fundamental to their development. Chlamydial attachment to host cells induces the activation of the Rac GTPase, which is required for the localization of WAVE2 at the sites of chlamydial entry. Co-immunoprecipitation experiments demonstrated that Chlamydia trachomatis infection promoted the interaction of Rac with WAVE2 and Abi-1, but not with IRSp53. siRNA depletion of WAVE2 and Abi-1 abrogated chlamydia-induced actin recruitment and significantly reduced the uptake of the pathogen by the depleted cells. Chlamydia invasion also requires the Arp2/3 complex as demonstrated by its localization to the sites of chlamydial attachment and the reduced efficiency of chlamydial invasion in cells overexpressing the VCA domain of the neural Wiskott-Aldrich syndrome protein. Thus, C. trachomatis activates Rac and promotes its interaction with WAVE2 and Abi-1 to activate the Arp2/3 complex resulting in the induction of actin cytoskeletal rearrangements that are required for invasion.


Assuntos
Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Infecções por Chlamydia/metabolismo , Chlamydia trachomatis , Proteínas do Citoesqueleto/metabolismo , Família de Proteínas da Síndrome de Wiskott-Aldrich/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Células COS , Chlamydia trachomatis/metabolismo , Chlamydia trachomatis/patogenicidade , Chlorocebus aethiops , Proteínas do Citoesqueleto/genética , Ativação Enzimática , Células HeLa , Humanos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Família de Proteínas da Síndrome de Wiskott-Aldrich/genética , Proteínas rac de Ligação ao GTP/genética
12.
J Bacteriol ; 188(14): 5289-92, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16816202

RESUMO

The chlamydial histone-like proteins, Hc1 and Hc2, function as global regulators of chromatin structure and gene expression. Hc1 and Hc2 expression and activity are developmentally regulated. A small metabolite that disrupts Hc1 interaction with DNA also disrupts Hc2 interactions; however, the small regulatory RNA that inhibits Hc1 translation is specific to Hc1.


Assuntos
Proteínas de Bactérias/metabolismo , Chlamydia trachomatis/genética , Chlamydia trachomatis/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Histonas/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas de Bactérias/genética , Chlamydia trachomatis/crescimento & desenvolvimento , Escherichia coli/crescimento & desenvolvimento , Regulação Bacteriana da Expressão Gênica , Histonas/genética
13.
Infect Immun ; 73(7): 3860-8, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15972471

RESUMO

Chlamydiae are obligate intracellular pathogens that efficiently induce their endocytosis by susceptible eukaryotic host cells. Recently, a Chlamydia trachomatis type III secreted effector protein, Tarp, was found to be translocated and tyrosine phosphorylated at the site of entry and associated with the recruitment of actin that coincides with endocytosis. C. trachomatis Tarp possesses up to six direct repeats of approximately 50 amino acids each. The majority of the tyrosine residues are found within this repeat region. Here we have ectopically expressed distinct domains of Tarp in HeLa 229 cells and demonstrated that tyrosine phosphorylation occurs primarily within the repeat region, while recruitment of actin is mediated by the C-terminal domain of the protein. A comparison of other sequenced chlamydial genomes revealed that each contains an ortholog of Tarp, although Chlamydia muridarum, Chlamydophila caviae, and Chlamydophila pneumoniae Tarp lack the large repeat region. Immunofluorescence and immunoblotting using an antiphosphotyrosine antibody show no evidence of phosphotyrosine at the site of entry of C. muridarum, C. caviae, and C. pneumoniae, although each species similarly recruits actin. Ectopic expression of full-length C. trachomatis and C. caviae Tarp confirmed that both recruit actin but only C. trachomatis Tarp is tyrosine phosphorylated. The data indicate that the C-terminal domain of Tarp is essential for actin recruitment and that tyrosine phosphorylation may not be an absolute requirement for actin recruitment. The results further suggest the potential for additional, unknown signal transduction pathways associated specifically with C. trachomatis.


Assuntos
Actinas/metabolismo , Proteínas de Bactérias/metabolismo , Chlamydia/química , Tirosina/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/química , Dados de Sequência Molecular , Fosforilação , Sequências Repetitivas de Aminoácidos , Transdução de Sinais , Especificidade da Espécie
14.
Proc Natl Acad Sci U S A ; 101(19): 7451-6, 2004 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-15123794

RESUMO

The chlamydial developmental cycle is characterized by an intracellular replicative form, termed the reticulate body, and an extracellular form called the elementary body. Elementary bodies are characterized by a condensed chromatin, which is maintained by a histone H1-like protein, Hc1. Differentiation of elementary bodies to reticulate bodies is accompanied by dispersal of the chromatin as chlamydiae become transcriptionally active, although the mechanisms of Hc1 release from DNA have remained unknown. Dissociation of the nucleoid requires chlamydial transcription and translation with negligible loss of Hc1. A genetic screen was therefore designed to identify chlamydial genes rescuing Escherichia coli from the lethal effects of Hc1 overexpression. CT804, a gene homologous to ispE, which encodes an intermediate enzyme of the non-mevalonate methylerythritol phosphate (MEP) pathway of isoprenoid biosynthesis, was selected. E. coli coexpressing CT804 and Hc1 grew normally, although they expressed Hc1 to a level equivalent to that which condensed the chromatin of parent Hc1-expressing controls. Inhibition of the MEP pathway with fosmidomycin abolished IspE rescue of Hc1-expressing E. coli. Deproteinated extract from IspE-expressing bacteria caused dispersal of purified chlamydial nucleoids, suggesting that chlamydial histone-DNA interactions are disrupted by a small metabolite within the MEP pathway rather than by direct action of IspE. By partial reconstruction of the MEP pathway, we determined that 2-C-methylerythritol 2,4-cyclodiphosphate dissociated Hc1 from chlamydial chromatin. These results suggest that chlamydial histone-DNA interactions are disrupted upon germination by a small metabolite in the MEP pathway of isoprenoid biosynthesis.


Assuntos
Chlamydia trachomatis/metabolismo , DNA Bacteriano/metabolismo , Eritritol/análogos & derivados , Eritritol/metabolismo , Histonas/metabolismo , Terpenos/metabolismo , Chlamydia trachomatis/genética , Genes Bacterianos , Células HeLa , Humanos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...