Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Emerg Infect Dis ; 26(2): 265-272, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31961294

RESUMO

Most human pegivirus 2 (HPgV-2) infections are associated with past or current hepatitis C virus (HCV) infection. HPgV-2 is thought to be a bloodborne virus: higher prevalence of active infection has been found in populations with a history of parenteral exposure to viruses. We evaluated longitudinally collected blood samples obtained from injection drug users (IDUs) for active and resolved HPgV-2 infections using a combination of HPgV-2-specific molecular and serologic tests. We found evidence of HPgV-2 infection in 11.2% (22/197) of past or current HCV-infected IDUs, compared with 1.9% (4/205) of an HCV-negative IDU population. Testing of available longitudinal blood samples from HPgV-2-positive participants identified 5 with chronic infection (>6 months viremia in >3 timepoints); 2 were identified among the HCV-positive IDUs and 3 among the HCV-negative IDUs. Our findings indicate that HPgV-2 can establish chronic infection and replicate in the absence of HCV.


Assuntos
Usuários de Drogas , Infecções por Flaviviridae/epidemiologia , Hepatite C , Pegivirus/isolamento & purificação , Adolescente , Adulto , California/epidemiologia , Coinfecção , Feminino , Infecções por Flaviviridae/sangue , Infecções por Flaviviridae/virologia , Humanos , Estudos Longitudinais , Masculino , Prevalência , Assunção de Riscos , Inquéritos e Questionários , Adulto Jovem
2.
J Viral Hepat ; 26(1): 30-37, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30187640

RESUMO

The prevalence of chronic hepatitis C virus (HCV) and the presence of human pegivirus 2 (HPgV-2) have not been examined in Cameroon, although HCV has been associated with HPgV-2 infections previously. Herein we aimed to characterize the burden and genetic diversity of HCV and the presence of HPgV-2 in Cameroon. Retrospective plasma specimens collected from N = 12 369 consenting subjects in South Cameroon from 2013 to 2016 were included in the study. The majority (97.1%) of participants were patients seeking health care. All specimens were screened for HCV using the Abbott RealTime HCV viral load assay and positive specimens with remaining volume were also screened for HPgV-2 antibodies on the Abbott ARCHITECT instrument, followed by molecular characterization. Overall, HCV RNA was detected in 305 (2.47%; 95% CI: 2.21%-2.75%) specimens. Notably, the prevalence of HCV RNA was 9.09% amongst participants over age 40 and 3.81% amongst males. Phylogenetic classification of N = 103 HCV sequences identified genotypes 1 (19.4%), 2 (15.5%) and 4 (65.1%) within the study cohort. Amongst HCV RNA-positive specimens, N = 28 (10.6%; 95% CI: 7.44%-14.90%) specimens also had detectable HPgV-2 antibodies. Of these, N = 2 viremic HPgV-2 infections were confirmed by sequencing and shared 93-94 median % identity with strains found on other continents. This is the first study to determine the prevalence of chronic HCV in Cameroon, and the discovery of HPgV-2 in this study cohort expands the geography of HPgV-2 to the African continent, indicating a widespread distribution exists.


Assuntos
Anticorpos Antivirais/sangue , Monitoramento Epidemiológico , Infecções por Flaviviridae/epidemiologia , Flaviviridae/isolamento & purificação , Hepatite C Crônica/epidemiologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Camarões/epidemiologia , Criança , Pré-Escolar , Coinfecção/epidemiologia , Coinfecção/virologia , Feminino , Flaviviridae/genética , Infecções por Flaviviridae/sangue , Hepacivirus/genética , Hepatite C Crônica/sangue , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Filogenia , Prevalência , RNA Viral/sangue , Estudos Retrospectivos , Adulto Jovem
3.
J Virol Methods ; 246: 117-124, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28479349

RESUMO

Zika virus is an arthropod-borne flavivirus that has rapidly developed into a world-wide concern. Discovered in 1947, the virus was relatively obscure until an outbreak occurred in 2007 in the Yap islands and spread eventually to the Americas in 2015. Only 20% of patients infected with Zika virus develop symptoms. However, there can be serious consequences of infection including birth defects in developing fetuses and links to Guillain-Barré syndrome. The swift rise in infections has necessitated the development of diagnostic tests for both the detection of viral RNA and the presence of virus-specific antibodies. Abbott has developed a dual target RT-PCR assay for the detection of Zika virus RNA within serum, plasma, whole blood, and urine using the automated m2000 system for sample extraction to result reporting. The Abbott RealTime ZIKA assay has a limit of detection of 30 copies per mL in serum, 40 copies per mL in plasma and urine, and 120 copies per mL in whole blood and demonstrates high specificity against challenges from closely related infectious agents.


Assuntos
RNA Viral/análise , Reação em Cadeia da Polimerase em Tempo Real/métodos , Infecção por Zika virus/diagnóstico , Zika virus/isolamento & purificação , Automação Laboratorial/instrumentação , Humanos , Limite de Detecção , RNA Viral/sangue , RNA Viral/genética , RNA Viral/urina , Reação em Cadeia da Polimerase em Tempo Real/instrumentação , Sensibilidade e Especificidade , Zika virus/genética , Infecção por Zika virus/virologia
4.
J Bacteriol ; 198(7): 1123-36, 2016 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-26811319

RESUMO

UNLABELLED: The envelope of Staphylococcus aureus is comprised of peptidoglycan and its attached secondary polymers, teichoic acid, capsular polysaccharide, and protein. Peptidoglycan synthesis involves polymerization of lipid II precursors into glycan strands that are cross-linked at wall peptides. It is not clear whether peptidoglycan structure is principally determined during polymerization or whether processive enzymes affect cell wall structure and function, for example, by generating conduits for protein secretion. We show here that S. aureus lacking SagB, a membrane-associated N-acetylglucosaminidase, displays growth and cell-morphological defects caused by the exaggerated length of peptidoglycan strands. SagB cleaves polymerized glycan strands to their physiological length and modulates antibiotic resistance in methicillin-resistant S. aureus (MRSA). Deletion of sagB perturbs protein trafficking into and across the envelope, conferring defects in cell wall anchoring and secretion, as well as aberrant excretion of cytoplasmic proteins. IMPORTANCE: Staphylococcus aureus is thought to secrete proteins across the plasma membrane via the Sec pathway; however, protein transport across the cell wall envelope has heretofore not been studied. We report that S. aureus sagB mutants generate elongated peptidoglycan strands and display defects in protein secretion as well as aberrant excretion of cytoplasmic proteins. These results suggest that the thick peptidoglycan layer of staphylococci presents a barrier for protein secretion and that SagB appears to extend the Sec pathway across the cell wall envelope.


Assuntos
Proteínas de Bactérias/metabolismo , Farmacorresistência Bacteriana , Hexosaminidases/metabolismo , Polissacarídeos/metabolismo , Staphylococcus aureus/enzimologia , Sequência de Aminoácidos , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica/fisiologia , Hexosaminidases/genética , Dados de Sequência Molecular , Mutação , Polissacarídeos/química , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo
5.
Proc Natl Acad Sci U S A ; 111(4): 1574-9, 2014 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-24434550

RESUMO

Staphylococcal protein A (SpA) is anchored to the cell wall envelope of Staphylococcus aureus by sortase A, which links the threonyl (T) of its C-terminal LPXTG motif to peptidoglycan cross-bridges (i.e., Gly5). SpA binds the Fcγ domains of IgG and protects staphylococci from opsonophagocytic clearance. Moreover, SpA cross-links B-cell receptors to modify host adaptive immune responses. The mechanisms whereby SpA is released from the bacterial surface to access the host's immune system are not known. Here we demonstrate that SpA is released with murein tetrapeptide-tetraglycyl [L-Ala-D-iGln-(SpA-Gly5)L-Lys-D-Ala-Gly4] linked to its C-terminal threonyl. LytN, a cross-wall murein hydrolase, contributes to the release of SpA by removing amino sugars [i.e., N-acetylmuramic acid-N-acetylglucosamine (MurNAc-GlcNAc)] from attached peptidoglycan, whereas LytM, a pentaglycyl-endopeptidase, triggers polypeptide release from the bacterial envelope. A model is proposed whereby murein hydrolases cleave the anchor structure of released SpA to modify host immune responses.


Assuntos
Parede Celular/metabolismo , Proteína Estafilocócica A/metabolismo , Staphylococcus aureus/metabolismo , Sequência de Aminoácidos , Dados de Sequência Molecular , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Proteína Estafilocócica A/química , Proteína Estafilocócica A/isolamento & purificação
6.
J Bacteriol ; 195(20): 4650-9, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23935043

RESUMO

The LytR-CpsA-Psr (LCP) proteins are thought to transfer bactoprenol-linked biosynthetic intermediates of wall teichoic acid (WTA) to the peptidoglycan of Gram-positive bacteria. In Bacillus subtilis, mutants lacking all three LCP enzymes do not deposit WTA in the envelope, while Staphylococcus aureus Δlcp mutants display impaired growth and reduced levels of envelope phosphate. We show here that the S. aureus Δlcp mutant synthesized WTA yet released ribitol phosphate polymers into the extracellular medium. Further, Δlcp mutant staphylococci no longer restricted the deposition of LysM-type murein hydrolases to cell division sites, which was associated with defects in cell shape and increased autolysis. Mutations in S. aureus WTA synthesis genes (tagB, tarF, or tarJ2) inhibit growth, which is attributed to the depletion of bactoprenol, an essential component of peptidoglycan synthesis (lipid II). The growth defect of S. aureus tagB and tarFJ mutants was alleviated by inhibition of WTA synthesis with tunicamycin, whereas the growth defect of the Δlcp mutant was not relieved by tunicamycin treatment or by mutation of tagO, whose product catalyzes the first committed step of WTA synthesis. Further, sortase A-mediated anchoring of proteins to peptidoglycan, which also involves bactoprenol and lipid II, was not impaired in the Δlcp mutant. We propose a model whereby the S. aureus Δlcp mutant, defective in tethering WTA to the cell wall, cleaves WTA synthesis intermediates, releasing ribitol phosphate into the medium and recycling bactoprenol for peptidoglycan synthesis.


Assuntos
Parede Celular/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Staphylococcus aureus/enzimologia , Ácidos Teicoicos/metabolismo , Divisão Celular , Membrana Celular , Parede Celular/química , Meios de Cultura/química , Família Multigênica , Mutação , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Ácidos Teicoicos/química
7.
J Biol Chem ; 287(13): 10460-10471, 2012 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-22303016

RESUMO

Cells of eukaryotic or prokaryotic origin express proteins with LysM domains that associate with the cell wall envelope of bacteria. The molecular properties that enable LysM domains to interact with microbial cell walls are not yet established. Staphylococcus aureus, a spherical microbe, secretes two murein hydrolases with LysM domains, Sle1 and LytN. We show here that the LysM domains of Sle1 and LytN direct murein hydrolases to the staphylococcal envelope in the vicinity of the cross-wall, the mid-cell compartment for peptidoglycan synthesis. LysM domains associate with the repeating disaccharide ß-N-acetylmuramic acid, (1→4)-ß-N-acetylglucosamine of staphylococcal peptidoglycan. Modification of N-acetylmuramic acid with wall teichoic acid, a ribitol-phosphate polymer tethered to murein linkage units, prevents the LysM domain from binding to peptidoglycan. The localization of LytN and Sle1 to the cross-wall is abolished in staphylococcal tagO mutants, which are defective for wall teichoic acid synthesis. We propose a model whereby the LysM domain ensures septal localization of LytN and Sle1 followed by processive cleavage of peptidoglycan, thereby exposing new LysM binding sites in the cross-wall and separating bacterial cells.


Assuntos
Proteínas de Bactérias/metabolismo , Parede Celular/metabolismo , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Peptidoglicano/metabolismo , Staphylococcus aureus/enzimologia , Proteínas de Bactérias/genética , Parede Celular/genética , Mutação , N-Acetil-Muramil-L-Alanina Amidase/genética , Peptidoglicano/genética , Estrutura Terciária de Proteína , Staphylococcus aureus/genética
8.
J Biol Chem ; 286(37): 32593-605, 2011 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-21784864

RESUMO

Cell cycle progression for the spherical microbe Staphylococcus aureus requires the coordinated synthesis and remodeling of peptidoglycan. The majority of these rearrangements takes place at the mid-cell, in a compartment designated the cross-wall. Secreted polypeptides endowed with a YSIRK-G/S signal peptide are directly delivered to the cross-wall compartment. One such YSIRK-containing protein is the murein hydrolase LytN. lytN mutations precipitate structural damage to the cross-wall and interfere with staphylococcal growth. Overexpression of lytN also affects growth and triggers rupture of the cross-wall. The lytN phenotype can be reversed by the controlled expression of lytN but not by adding purified LytN to staphylococcal cultures. LytN harbors LysM and CHAP domains, the latter of which functions as both an N-acetylmuramoyl-L-alanine amidase and D-alanyl-glycine endopeptidase. Thus, LytN secretion into the cross-wall promotes peptidoglycan separation and completion of the staphylococcal cell cycle.


Assuntos
Parede Celular/enzimologia , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Staphylococcus aureus/enzimologia , Staphylococcus aureus/crescimento & desenvolvimento , Parede Celular/genética , Mutação , N-Acetil-Muramil-L-Alanina Amidase/genética , Peptidoglicano/genética , Peptidoglicano/metabolismo , Estrutura Terciária de Proteína , Staphylococcus aureus/genética
9.
Mol Microbiol ; 78(1): 238-52, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20923422

RESUMO

The human pathogen Staphylococcus aureus requires cell wall anchored surface proteins to cause disease. During cell division, surface proteins with YSIRK signal peptides are secreted into the cross-wall, a layer of newly synthesized peptidoglycan between separating daughter cells. The molecular determinants for the trafficking of surface proteins are, however, still unknown. We screened mutants with non-redundant transposon insertions by fluorescence-activated cell sorting for reduced deposition of protein A (SpA) into the staphylococcal envelope. Three mutants, each of which harboured transposon insertions in genes for transmembrane proteins, displayed greatly reduced envelope abundance of SpA and surface proteins with YSIRK signal peptides. Characterization of the corresponding mutations identified three transmembrane proteins with abortive infectivity (ABI) domains, elements first described in lactococci for their role in phage exclusion. Mutations in genes for ABI domain proteins, designated spdA, spdB and spdC (surface protein display), diminish the expression of surface proteins with YSIRK signal peptides, but not of precursor proteins with conventional signal peptides. spdA, spdB and spdC mutants display an increase in the thickness of cross-walls and in the relative abundance of staphylococci with cross-walls, suggesting that spd mutations may represent a possible link between staphylococcal cell division and protein secretion.


Assuntos
Divisão Celular , Proteínas de Membrana/metabolismo , Proteína Estafilocócica A/metabolismo , Staphylococcus aureus/citologia , Motivos de Aminoácidos , Genes Bacterianos , Teste de Complementação Genética , Mutação INDEL , Proteínas de Membrana/genética , Mutagênese Insercional , Mutação , Sinais Direcionadores de Proteínas , Transporte Proteico , Proteína Estafilocócica A/genética , Staphylococcus aureus/genética
10.
Inflamm Bowel Dis ; 16(4): 620-9, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19714757

RESUMO

BACKGROUND: Visilizumab is a humanized IgG(2) monoclonal anti-CD3 antibody. We evaluated its safety and dose response in severe intravenous steroid-refractory ulcerative colitis (UC). METHODS: In all, 104 patients were treated. In Stage I, 73 patients were randomly assigned to receive intravenous visilizumab 5, 7.5, 10, or 12.5 microg/kg/day for 2 consecutive days. In Stage II, 33 patients received visilizumab at the optimal clinical dose (OCD) of 5 microg/kg/day for 2 days. Symptomatic response and remission were defined by the modified Truelove-Witts severity index. Clinical response and remission were defined by the Mayo score. RESULTS: The rates of symptomatic response at day 15 in the 5, 7.5, 10, or 12.5 microg/kg dose groups were 71%, 70%, 50%, and 61%, respectively, in Stage I and in 54% in Stage II. The symptomatic remission rates were 35%, 5%, 22%, and 11% in Stage I and 18% in Stage II. The rates of clinical response at day 30 in the 5, 7.5, 10, or 12.5 microg/kg dose groups were 71%, 65%, 50%, and 67%, respectively, in Stage I and 55% in Stage II. The clinical remission rates were 6%, 5%, 0%, and 11% in Stage I and 6% in Stage II. All patients experienced adverse events. Serious adverse events included abdominal abscess, cytomegalovirus infection, atrial fibrillation, herpes zoster, and esophageal candidiasis. CONCLUSIONS: Treatment with visilizumab induced symptomatic response and clinical response. Results with 5 microg/kg/day were similar to those observed with higher doses.


Assuntos
Corticosteroides/farmacologia , Anticorpos Monoclonais/uso terapêutico , Complexo CD3/imunologia , Colite Ulcerativa/tratamento farmacológico , Resistência a Medicamentos , Adolescente , Adulto , Idoso , Anticorpos Monoclonais Humanizados , Colite Ulcerativa/patologia , Relação Dose-Resposta a Droga , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Taxa de Sobrevida , Resultado do Tratamento , Adulto Jovem
11.
DNA Cell Biol ; 28(6): 277-84, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19348590

RESUMO

Apicomplexa is a phylum within the kingdom Protista that contains some of the most significant threats to public health. One of the members of this phylum, Toxoplasma gondii, is amenable to molecular genetic analyses allowing for the identification of factors critical for colonization and disease. A pathway found to be important for T. gondii pathogenesis is the Ran network of nuclear trafficking. Bioinformatics analysis of apicomplexan genomes shows that while Ran is well conserved, the key regulators of Ran--Regulator of Chromosome Condensation 1 and Ran GTPase activating protein--are either highly divergent or absent. Likewise, several import and export receptor molecules that are crucial for nuclear transport are either not present or have experienced genetic drift such that they are no longer recognizable by bioinformatics tools. In this minireview we describe the basics of nuclear trafficking and compare components within apicomplexans to defined systems in humans and yeast. A detailed analysis of the nuclear trafficking network in these eukaryotes is required to understand how this potentially unique cellular biological pathway contributes to host-parasite interactions.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Apicomplexa/citologia , Proteínas de Protozoários/fisiologia , Animais , Proteínas de Ciclo Celular/fisiologia , Núcleo Celular/metabolismo , Cryptosporidium parvum/citologia , Proteínas Ativadoras de GTPase/fisiologia , Guanosina Trifosfato/fisiologia , Humanos , Carioferinas/fisiologia , Modelos Biológicos , Proteínas Nucleares/fisiologia , Plasmodium falciparum/citologia , Especificidade da Espécie , Toxoplasma/citologia , beta Carioferinas/fisiologia , Proteína ran de Ligação ao GTP/fisiologia
12.
Am J Gastroenterol ; 104(4): 868-76, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19240707

RESUMO

OBJECTIVES: Monoclonal antibodies to CD3 and CD4 T-cell receptors are evolving for Crohn's disease (CD) and ulcerative colitis. Their administration is often associated with a cytokine release syndrome (CRS). METHODS: We evaluated data from two prospective clinical trials (NCT00267709 and NCT00267722) of visilizumab (HuM291), a novel humanized anti-CD3 antibody, in 34 patients with CD who received 10 microg/kg intravenously on 2 consecutive days. Serum hepatic tests including bilirubin, alkaline phosphatase (AP), gamma-glutamyltransferase (GGT), alanine aminotransferase (ALT) and aspartate aminotransferase (AST), visilizumab concentrations, and a panel of 16 cytokines were measured pre- and postadministration of visilizumab. RESULTS: Patients experienced CRS symptoms at a median of 45 min postinfusion. The cytokine profile was characterized by interferon-inducible protein-10 (IP-10), interleukin-10 (IL-10), tumor necrosis factor-alpha (TNF-alpha), interferon-gamma, monocyte chemotactic protein 1 (MCP-1), interleukin-8 (IL-8), interleukin-6 (IL-6), interleukin-2 (IL-2), and interleukin 1 receptor antagonist (IL-1Ra), which were elevated between 6 (IL-1Ra) and 870 (IP-10) times their baseline concentrations. TNF-alpha and IL-2 peaked at the first day 1 h post infusion, whereas all others peaked at 6 h post infusion. Eighty-six percent of patients experienced an elevation above the upper limit of normal in hepatic enzymes (GGT 73%, AST 73%, ALT 64%, and AP 42% of patients), but not bilirubin, within 6 h postinfusion. CONCLUSIONS: Transient elevation of hepatic enzymes occurred frequently in patients with CD treated with visilizumab and was associated with CRS. CD patients could be predisposed due to an aberrant expression of adhesion molecules in the liver that promotes CRS upon engagement of the T-cell receptor and may relate to extraintestinal disease manifestations such as primary sclerosing cholangitis.


Assuntos
Anticorpos Monoclonais/efeitos adversos , Doença Hepática Induzida por Substâncias e Drogas , Doença de Crohn/tratamento farmacológico , Adulto , Idoso , Alanina Transaminase/sangue , Fosfatase Alcalina/sangue , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais Humanizados , Aspartato Aminotransferases/sangue , Bilirrubina/sangue , Complexo CD3/imunologia , Doença de Crohn/sangue , Citocinas/sangue , Relação Dose-Resposta a Droga , Feminino , Seguimentos , Humanos , Injeções Intravenosas , Hepatopatias/sangue , Testes de Função Hepática , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Adulto Jovem , gama-Glutamiltransferase/sangue
13.
Mol Microbiol ; 70(2): 410-20, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18761691

RESUMO

Protozoan parasites represent major public health challenges. Many aspects of their cell biology are distinct from their animal hosts, providing potential therapeutic targets. Toxoplasma gondii is a protozoan parasite that contains a divergent regulator of chromosome condensation 1 (TgRCC1) that is required for virulence and efficient nuclear trafficking. RCC1 proteins function as a guanine exchange factor for Ras-related nuclear protein (Ran), an abundant GTPase responsible for the majority of nucleocytoplasmic transport. Here we show that while there are dramatic differences from well-conserved RCC1 proteins, TgRCC1 associates with chromatin, interacts with Ran and complements a mammalian temperature-sensitive RCC1 mutant cell line. During the investigation of TgRCC1, we observed several unprecedented phenotypes for TgRan, despite a high level of sequence conservation. The cellular distribution of TgRan is found throughout the parasite cell, whereas Ran in late branching eukaryotes is predominantly nuclear. Additionally, T. gondii tolerates at least low-level expression of dominant lethal Ran mutants. Wild type parasites expressing dominant negative TgRan grew similarly to wild type in standard tissue culture conditions, but were attenuated in serum-starved host cells and mice. These growth characteristics paralleled the TgRCC1 mutant and highlight the importance of the nuclear transport pathway for virulence of eukaryotic pathogens.


Assuntos
Transporte Ativo do Núcleo Celular , Proteínas de Protozoários/metabolismo , Toxoplasma/fisiologia , Toxoplasma/patogenicidade , Proteína ran de Ligação ao GTP/metabolismo , Animais , Núcleo Celular/química , Cromatina/metabolismo , Citoplasma/química , Feminino , Teste de Complementação Genética , Camundongos , Ligação Proteica , Mapeamento de Interação de Proteínas , Análise de Sobrevida , Toxoplasmose/parasitologia , Virulência
14.
Proc Natl Acad Sci U S A ; 104(24): 10181-6, 2007 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-17535896

RESUMO

Eukaryotic parasites are a leading cause of morbidity and mortality worldwide, yet little is known about the genetic basis of their virulence. Here, we present a forward genetic screen to study pathogenesis in the protozoan parasite Toxoplasma gondii. By using modified signature-tagged mutagenesis, the growth of 6,300 T. gondii insertional mutants was compared in cell culture and murine infection to identify genes required specifically in vivo. One of the 39 avirulent mutants is disrupted in a divergent ortholog of the regulator of chromosome condensation 1 (RCC1), which is critical for nuclear trafficking in model systems. Although this RCC1 mutant grows similar to wild type in standard tissue culture conditions, it is growth-impaired under nutrient limitation. Genetic complementation of mutant parasites with the T. gondii RCC1 gene fully restores both virulence in mice and growth under low-nutrient conditions. Further analysis shows that there is a significant defect in nuclear trafficking in the RCC1 mutant. These findings suggest that the rate of nuclear transport is a critical factor affecting growth in low-nutrient conditions in vivo and in vitro. Additionally, we observed that although RCC1 proteins are highly conserved in organisms from humans to yeast, no protozoan parasite encodes a characteristic RCC1. This protein divergence may represent a unique mechanism of nucleocytoplasmic transport. This study illustrates the power of this forward genetics approach to identify atypical virulence mechanisms.


Assuntos
Núcleo Celular/metabolismo , Genes de Protozoários , Proteínas de Protozoários/metabolismo , Toxoplasma/genética , Animais , Células Cultivadas , Cromossomos , DNA de Protozoário/genética , Fibroblastos/parasitologia , Teste de Complementação Genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos CBA , Modelos Biológicos , Dados de Sequência Molecular , Mutagênese Insercional , Mutação , Transporte Proteico , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Pele/citologia , Toxoplasmose , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...