Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 38(7): 1504-1518, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29853567

RESUMEN

OBJECTIVE: Cells use various mechanisms to maintain cellular cholesterol homeostasis including efflux of cholesterol from the cellular plasma membrane to cholesterol acceptors such as HDLs (high-density lipoproteins). Little is known about the transfer of cholesterol from cells into the extracellular matrix. Using a unique monoclonal antibody that detects ordered cholesterol arrays (ie, cholesterol micro[or nano]-domains), we previously identified that particles containing these cholesterol domains accumulate in the extracellular matrix during cholesterol enrichment of human monocyte-derived macrophages and are found in atherosclerotic lesions. In this study, we further investigate these deposited particles containing cholesterol microdomains and discover their unexpected morphology. APPROACH AND RESULTS: Although appearing spherical at the resolution of the conventional fluorescence microscope, super-resolution immunofluorescence and atomic force microscopy of in situ cholesterol microdomains, and immunoelectron microscopy of isolated cholesterol microdomains revealed that the microdomains are not vesicles or 3-dimensional crystals but rather appear as branching irregularly shaped deposits of varying size. These cholesterol microdomain-containing deposits are shed from the plasma membrane into the extracellular matrix. CONCLUSIONS: To date, research on cellular excretion of excess cholesterol has demonstrated cellular cholesterol efflux in the form of membranous vesicles and discoidal HDL particles released into the fluid-phase medium. Shedding of plasma membrane cholesterol microdomains provides an additional mechanism for cells such as macrophages to maintain plasma membrane cholesterol homeostasis. Furthermore, recognition that macrophages shed cholesterol microdomains into the extracellular matrix is important to our understanding of extracellular buildup of cholesterol in atherosclerosis.


Asunto(s)
Colesterol/metabolismo , Matriz Extracelular/metabolismo , Macrófagos/metabolismo , Microdominios de Membrana/metabolismo , Animales , Células Cultivadas , Matriz Extracelular/ultraestructura , Humanos , Macrófagos/ultraestructura , Masculino , Microdominios de Membrana/ultraestructura , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Microscopía de Fuerza Atómica , Microscopía Electroquímica de Rastreo , Microscopía Fluorescente
2.
BMC Genomics ; 12: 109, 2011 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-21324157

RESUMEN

BACKGROUND: Chlamydiae are obligate intracellular bacteria that multiply in a vacuolar compartment, the inclusion. Several chlamydial proteins containing a bilobal hydrophobic domain are translocated by a type III secretion (TTS) mechanism into the inclusion membrane. They form the family of Inc proteins, which is specific to this phylum. Based on their localization, Inc proteins likely play important roles in the interactions between the microbe and the host. In this paper we sought to identify and analyze, using bioinformatics tools, all putative Inc proteins in published chlamydial genomes, including an environmental species. RESULTS: Inc proteins contain at least one bilobal hydrophobic domain made of two transmembrane helices separated by a loop of less than 30 amino acids. Using bioinformatics tools we identified 537 putative Inc proteins across seven chlamydial proteomes. The amino-terminal segment of the putative Inc proteins was recognized as a functional TTS signal in 90% of the C. trachomatis and C. pneumoniae sequences tested, validating the data obtained in silico. We identified a macro domain in several putative Inc proteins, and observed that Inc proteins are enriched in segments predicted to form coiled coils. A surprisingly large proportion of the putative Inc proteins are not constitutively translocated to the inclusion membrane in culture conditions. CONCLUSIONS: The Inc proteins represent 7 to 10% of each proteome and show a great degree of sequence diversity between species. The abundance of segments with a high probability for coiled coil conformation in Inc proteins support the hypothesis that they interact with host proteins. While the large majority of Inc proteins possess a functional TTS signal, less than half may be constitutively translocated to the inclusion surface in some species. This suggests the novel finding that translocation of Inc proteins may be regulated by as-yet undetermined mechanisms.


Asunto(s)
Proteínas Bacterianas/genética , Sistemas de Secreción Bacterianos , Chlamydia/genética , Genoma Bacteriano , Proteínas de la Membrana/genética , Secuencia de Aminoácidos , Biología Computacional , Células HeLa , Humanos , Datos de Secuencia Molecular , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Transporte de Proteínas , Proteoma/genética , Alineación de Secuencia
3.
BMC Microbiol ; 11: 87, 2011 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-21527029

RESUMEN

BACKGROUND: The periplasmic High Temperature Requirement protein A (HtrA) plays important roles in bacterial protein folding and stress responses. However, the role of chlamydial HtrA (cHtrA) in chlamydial pathogenesis is not clear. RESULTS: The cHtrA was detected both inside and outside the chlamydial inclusions. The detection was specific since both polyclonal and monoclonal anti-cHtrA antibodies revealed similar intracellular labeling patterns that were only removed by absorption with cHtrA but not control fusion proteins. In a Western blot assay, the anti-cHtrA antibodies detected the endogenous cHtrA in Chlamydia-infected cells without cross-reacting with any other chlamydial or host cell antigens. Fractionation of the infected cells revealed cHtrA in the host cell cytosol fraction. The periplasmic cHtrA protein appeared to be actively secreted into host cell cytosol since no other chlamydial periplasmic proteins were detected in the host cell cytoplasm. Most chlamydial species secreted cHtrA into host cell cytosol and the secretion was not inhibitable by a type III secretion inhibitor. CONCLUSION: Since it is hypothesized that chlamydial organisms possess a proteolysis strategy to manipulate host cell signaling pathways, secretion of the serine protease cHtrA into host cell cytosol suggests that the periplasmic cHtrA may also play an important role in chlamydial interactions with host cells.


Asunto(s)
Chlamydia trachomatis/enzimología , Chlamydia trachomatis/patogenicidad , Citosol/química , Proteínas de Choque Térmico/metabolismo , Proteínas Periplasmáticas/metabolismo , Serina Proteasas/metabolismo , Factores de Virulencia/metabolismo , Western Blotting , Fraccionamiento Celular , Técnica del Anticuerpo Fluorescente Directa , Células HeLa , Humanos , Transporte de Proteínas
4.
Microbiology (Reading) ; 156(Pt 10): 3031-3040, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20522495

RESUMEN

The chlamydial protease/proteasome-like activity factor (CPAF) is secreted into the host cytosol to degrade various host factors that benefit chlamydial intracellular survival. Although the full-length CPAF is predicted to contain a putative signal peptide at its N terminus, the secretion pathway of CPAF is still unknown. Here, we have provided experimental evidence that the N-terminal sequence covering the M1-G31 region was cleaved from CPAF during chlamydial infection. The CPAF N-terminal sequence, when expressed in a phoA gene fusion construct, was able to direct the export of the mature PhoA protein across the inner membrane of wild-type Escherichia coli. However, E. coli mutants deficient in SecB failed to support the CPAF signal-peptide-directed secretion of PhoA. Since native PhoA secretion was known to be independent of SecB, this SecB dependence must be rendered by the CPAF leader peptide. Furthermore, lack of SecY function also blocked the CPAF signal-peptide-directed secretion of PhoA. Most importantly, CPAF secretion into the host cell cytosol during chlamydial infection was selectively inhibited by an inhibitor specifically targeting type I signal peptidase but not by a type III secretion-system-specific inhibitor. Together, these observations have demonstrated that the chlamydial virulence factor CPAF relies on Sec-dependent transport for crossing the chlamydial inner membrane, which has provided essential information for further delineating the pathways of CPAF action and understanding chlamydial pathogenic mechanisms.


Asunto(s)
Proteínas Bacterianas/metabolismo , Chlamydia trachomatis/metabolismo , Señales de Clasificación de Proteína , Factores de Virulencia/metabolismo , Fosfatasa Alcalina/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Chlamydia trachomatis/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Células HeLa , Humanos , Datos de Secuencia Molecular , Transporte de Proteínas , Factores de Virulencia/genética
5.
Microb Pathog ; 49(4): 164-73, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20510344

RESUMEN

The Chlamydia-secreted protease/proteasome-like activity factor (CPAF) is synthesized as a proenzyme (proCPAF) and requires processing for proteolytic activity. Recent structural studies have further demonstrated that CPAF is a serine protease that can undergo autoprocessing and self-activation in a concentration-dependent manner in vitro. However, it is not known how CPAF is processed and activated during chlamydial infection. In the current study, we used a mutant CPAF designated as CPAF(E558A) that is deficient in processing by itself as a substrate to search for putative CPAF activation factor(s) in Chlamydia-infected cells. CPAF(E558A) was processed by the lysates made from Chlamydia-infected cells and the processing activity correlated with the presence of endogenous active CPAF in the fractionated lysate samples. CPAF produced in the Chlamydia-infected cells is required for processing the mutant CPAF(E558A) since the processing activity was removed by depletion with anti-CPAF but not control antibodies. Furthermore, a purified and activated wild type CPAF alone was sufficient for processing CPAF(E558A) and no other chlamydial proteases are required. Finally, fusion tag-induced oligomerization can lead to autoprocessing and self-activation of the wild type CPAF in mammalian cells. These observations together have demonstrated that CPAF undergoes autoprocessing and self-activation during chlamydial infection.


Asunto(s)
Proteínas Bacterianas/metabolismo , Chlamydia trachomatis/patogenicidad , Serina Proteasas/metabolismo , Factores de Virulencia/metabolismo , Sustitución de Aminoácidos/genética , Proteínas Bacterianas/genética , Células HeLa , Humanos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación Missense , Serina Proteasas/genética , Factores de Virulencia/genética
6.
Vaccines (Basel) ; 8(4)2020 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-33066540

RESUMEN

Human respiratory syncytial virus (RSV) is a cause of lower respiratory tract infection in infants, young children, and older adults. There is no licensed vaccine and prophylactic treatment options are limited. The RSV fusion (F) glycoprotein is a target of host immunity and thus a focus for vaccine development. F-trimers are metastable and undergo significant rearrangements from the prefusion to a stable postfusion structure with neutralizing epitopes on intermediate structures. We hypothesize that vaccine strategies that recapitulate the breathable F quaternary structure, and provide accessibility of B-cells to epitopes on intermediate conformations, may collectively contribute to protective immunity, while rigid prefusion F structures restrict access to key protective epitopes. To test this hypothesis, we used the near full-length prefusogenic F as a backbone to construct three prefusion F variants with substitutions in the hydrophobic head cavity: (1) disulfide bond mutant (DS), (2) space filling hydrophobic amino acid substitutions (Cav1), and (3) DS, Cav1 double mutant (DS-Cav1). In this study, we compared the immunogenicity of prefusogenic F to prefusion F variants in two animal models. Native prefusogenic F was significantly more immunogenic, producing high titer antibodies to prefusogenic, prefusion, and postfusion F structures, while animals immunized with DS or DS-Cav1 produced antibodies to prefusion F. Importantly, prefusogenic F elicited antibodies that target neutralizing epitopes including prefusion-specific site zero (Ø) and V and conformation-independent neutralizing sites II and IV. Immunization with DS or DS-Cav1 elicited antibodies primarily to prefusion-specific sites Ø and V with little or no antibodies to other key neutralizing sites. Animals immunized with prefusogenic F also had significantly higher levels of antibodies that cross-neutralized RSV A and B subtypes, while immunization with DS or DS-Cav1 produced antibodies primarily to the A subtype. We conclude that breathable trimeric vaccines that closely mimic the native F-structure, and incorporate strategies for B-cell accessibility to protective epitopes, are important considerations for vaccine design. F structures locked in a single conformation restrict access to neutralizing epitopes that may collectively contribute to destabilizing F-trimers important for broad protection. These results also have implications for vaccine strategies targeting other type 1 integral membrane proteins.

7.
Cell Mol Immunol ; 16(2): 154-164, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29503442

RESUMEN

A recent meta-analysis revealed the contribution of the SIGLEC6 locus to the risk of developing systemic lupus erythematosus (SLE). However, no specific Siglec (sialic acid-binding immunoglobulin-like lectin) genes (Siglecs) have been implicated in the pathogenesis of SLE. Here, we performed in silico analysis of the function of three major protective alleles in the locus and found that these alleles were expression quantitative trait loci that enhanced expression of the adjacent SIGLEC12 gene. These data suggest that SIGLEC12 may protect against the development of SLE in Asian populations. Consistent with human genetic data, we identified two missense mutations in lupus-prone B6.NZMSle1/Sle2/Sle3 (Sle1-3) mice in Siglece, which is the murine Siglec with the greatest homology to human SIGLEC12. Since the mutations resulted in reduced binding of Siglec E to splenic cells, we evaluated whether Siglece-/- mice had SLE phenotypes. We found that Siglece-/- mice showed increased autoantibody production, glomerular immune complex deposition and severe renal pathology reminiscent of human SLE nephropathy. Our data demonstrate that the Siglec genes confer resistance to SLE in mice and humans.


Asunto(s)
Antígenos CD/genética , Antígenos CD/fisiología , Antígenos de Diferenciación de Linfocitos B/fisiología , Antígenos de Diferenciación Mielomonocítica/genética , Modelos Animales de Enfermedad , Lectinas/genética , Lupus Eritematoso Sistémico/prevención & control , Activación de Linfocitos/inmunología , Proteínas de la Membrana/genética , Polimorfismo de Nucleótido Simple , Alelos , Secuencia de Aminoácidos , Animales , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Femenino , Humanos , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NZB , Ratones Noqueados , Mutación , Fenotipo , Homología de Secuencia
8.
Biomolecules ; 9(12)2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31779197

RESUMEN

Lecithin:cholesterol acyltransferase (LCAT) is an enzyme secreted by the liver and circulates with high-density lipoprotein (HDL) in the blood. The enzyme esterifies plasma cholesterol and increases the capacity of HDL to carry and potentially remove cholesterol from tissues. Cholesterol accumulates within the extracellular connective tissue matrix of the cornea stroma in individuals with genetic deficiency of LCAT. LCAT can be activated by apolipoproteins (Apo) including ApoD and ApoA1. ApoA1 also mediates cellular synthesis of HDL. This study examined the expression of LCAT by epithelial cells, keratocytes, and endothelial cells, the cell types that comprise from anterior to posterior the three layers of the cornea. LCAT and ApoD were immunolocalized to all three cell types within the cornea, while ApoA1 was immunolocalized to keratocytes and endothelium but not epithelium. In situ hybridization was used to detect LCAT, ApoD, and ApoA1 mRNA to learn what cell types within the cornea synthesize these proteins. No corneal cells showed mRNA for ApoA1. Keratocytes and endothelium both showed ApoD mRNA, but epithelium did not. Epithelium and endothelium both showed LCAT mRNA, but despite the presence of LCAT protein in keratocytes, keratocytes did not show LCAT mRNA. RNA sequencing analysis of serum-cultured dedifferentiated keratocytes (commonly referred to as corneal stromal fibroblasts) revealed the presence of both LCAT and ApoD (but not ApoA1) mRNA, which was accompanied by their respective proteins detected by immunolabeling of the cultured keratocytes and Western blot analysis of keratocyte lysates. The results indicate that keratocytes in vivo show both ApoA1 and LCAT proteins, but do not synthesize these proteins. Rather, keratocytes in vivo must take up ApoA1 and LCAT from the corneal interstitial tissue fluid.


Asunto(s)
Apolipoproteína A-I/metabolismo , Apolipoproteínas D/metabolismo , Colesterol/metabolismo , Córnea/metabolismo , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Anciano , Apolipoproteína A-I/sangre , Apolipoproteína A-I/genética , Apolipoproteínas D/sangre , Apolipoproteínas D/genética , Córnea/enzimología , Córnea/patología , Córnea/ultraestructura , Distrofias Hereditarias de la Córnea/genética , Distrofias Hereditarias de la Córnea/metabolismo , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/metabolismo , Células Endoteliales/metabolismo , Células Epiteliales/metabolismo , Femenino , Fibroblastos/metabolismo , Humanos , Queratinocitos/metabolismo , Deficiencia de la Lecitina Colesterol Aciltransferasa/genética , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Lipoproteínas HDL/sangre , Masculino , Microscopía Electrónica , Persona de Mediana Edad , Fosfatidilcolina-Esterol O-Aciltransferasa/sangre , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Fosfolípidos/metabolismo , RNA-Seq , Enfermedad de Tangier/genética , Enfermedad de Tangier/metabolismo
9.
PLoS One ; 10(5): e0127909, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25996495

RESUMEN

We previously identified hypothetical protein Cpn1027 as a novel inclusion membrane protein that is unique to Chlamydia pneumoniae. In the current study, using a yeast-two hybrid screen assay, we identified host cell cytoplasmic activation/proliferation-associated protein 2 (Caprin2) as an interacting partner of Cpn1027. The interaction was confirmed and mapped to the C-termini of both Cpn1027 and Caprin2 using co-immunoprecipitation and GST pull-down assays. A RFP-Caprin2 fusion protein was recruited to the chlamydial inclusion and so was the endogenous GSK3ß, a critical component of the ß-catenin destruction complex in the Wnt signaling pathway. Cpn1027 also co-precipitated GSK3ß. Caprin2 is a key regulator of the Wnt signaling pathway by promoting the recruitment of the ß-catenin destruction complex to the cytoplasmic membrane in the presence of Wnt signaling while GSK3ß is required for priming ß-catenin for degradation in the absence of Wnt signaling. The Cpn1027 interactions with Caprin2 and GSK3ß may allow C. pneumoniae to actively sequester the ß-catenin destruction complex so that ß-catenin is maintained even in the absence of extracellular Wnt activation signals. The maintained ß-catenin can trans-activate Wnt target genes including Bcl-2, which may contribute to the chlamydial antiapoptotic activity. We found that the C. pneumoniae-infected cells were more resistant to apoptosis induction and the anti-apoptotic activity was dependent on ß-catenin. Thus, the current study suggests that the chlamydial inclusion protein Cpn1027 may be able to manipulate host Wnt signaling pathway for enhancing the chlamydial anti-apoptotic activity.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Chlamydophila pneumoniae/metabolismo , Proteínas de la Membrana/metabolismo , Vía de Señalización Wnt , Apoptosis , Proteínas Bacterianas/química , Línea Celular , Técnicas de Silenciamiento del Gen , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Proteínas de la Membrana/química , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Proteínas de Unión al ARN , beta Catenina/genética , beta Catenina/metabolismo
10.
Infect Immun ; 75(1): 497-503, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17101661

RESUMEN

The hypothetical protein encoded by Chlamydia pneumoniae open reading frame cpn0308 was detected in inclusion membranes of C. pneumoniae-infected cells using antibodies raised with Cpn0308 fusion proteins. The anti-Cpn0308 antibodies did not cross-react with IncA, a known C. pneumoniae inclusion membrane protein, although the anti-Cpn0308 antibody staining overlapped with the anti-IncA antibody labeling. The labeling of the inclusion membrane by the anti-Cpn0308 antibody was specifically blocked by the Cpn0308 but not IncA fusion proteins. The Cpn0308 antigen was detectable 24 h after infection and remained in the inclusion membrane throughout the infection course.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Chlamydophila pneumoniae/fisiología , Animales , Western Blotting , Células HeLa , Interacciones Huésped-Parásitos , Humanos , Inmunohistoquímica , Membranas Intracelulares/metabolismo , Ratones , Microscopía Confocal , Fosfoproteínas/metabolismo
11.
Microbiology (Reading) ; 153(Pt 3): 777-86, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17322198

RESUMEN

The hypothetical protein Cpn1027 was detected in the inclusion membrane of Chlamydia pneumoniae-infected cells with antibodies raised with Cpn1027 fusion proteins in an indirect immunofluorescence assay. The inclusion membrane staining by the anti-Cpn1027 antibodies co-localized with the staining of an antibody recognizing a known inclusion membrane protein designated IncA and these membrane stainings were blocked by the corresponding but not irrelevant fusion proteins. Although Cpn1027 was not predicted to be an inclusion membrane protein, it contained a bi-lobed hydrophobic domain region at its N-terminus, a signature secondary structural motif possessed by most chlamydial inclusion membrane proteins. The Cpn1027 protein was detected as early as 12 h after C. pneumoniae infection and remained in the inclusion membrane throughout the rest of the infection cycle. Cytosolic expression of Cpn1027 via a transgene failed to affect the subsequent chlamydial infection. The anti-Cpn1027 polyclonal antisera failed to detect any significant signals in cells infected with chlamydial species other than C. pneumoniae, which is consistent with the sequence analysis result that no significant homologues of Cpn1027 were found in any other species. These experiments together have demonstrated that Cpn1027 is a newly identified inclusion membrane protein unique to C. pneumoniae.


Asunto(s)
Proteínas Bacterianas/metabolismo , Chlamydophila pneumoniae/química , Cuerpos de Inclusión/química , Proteínas de la Membrana/metabolismo , Proteínas Bacterianas/química , Chlamydophila pneumoniae/metabolismo , Proteínas de la Membrana/química , Microscopía Confocal , Microscopía Fluorescente , Estructura Secundaria de Proteína
12.
Microb Pathog ; 42(2-3): 111-6, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17236746

RESUMEN

Cpn0585, encoded by a hypothetical open reading frame in Chlamydia pneumoniae genome, was detected in the inclusion membrane during C. pneumoniae infection using both polyclonal and monoclonal antibodies raised with Cpn0585 fusion protein. The anti-Cpn0585 antibodies specifically recognized the endogenous Cpn0585 without cross-reacting with IncA (a known inclusion membrane protein of C. pneumoniae) or other control antigens. A homologue of Cpn0585 in the C. caviae species (encoded by the ORF CCA00156) was also localized in the inclusion membrane of the C. caviae-infected cells. The Cpn0585 protein became detectable 24h while CCA00156 as early as 8h after infection. Once expressed, both proteins remained in the inclusion membrane throughout the rest of infection course.


Asunto(s)
Proteínas Bacterianas/metabolismo , Membrana Celular/metabolismo , Infecciones por Chlamydophila/metabolismo , Chlamydophila pneumoniae , Cuerpos de Inclusión/metabolismo , Proteínas Bacterianas/genética , Infecciones por Chlamydophila/microbiología , Infecciones por Chlamydophila/fisiopatología , Chlamydophila pneumoniae/genética , Células HeLa/metabolismo , Células HeLa/microbiología , Humanos , Operón , Especificidad de la Especie , Factores de Tiempo
13.
Infect Immun ; 74(11): 6479-86, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17057097

RESUMEN

Using antibodies raised with chlamydial fusion proteins, we have localized a protein encoded by the hypothetical open reading frame Cpn0797 in the cytoplasm of Chlamydia pneumoniae-infected host cells. The anti-Cpn0797 antibodies specifically recognized Cpn0797 protein without cross-reacting with either CPAFcp or Cpn0796, the only two proteins known to be secreted into the host cell cytosol by C. pneumoniae organisms. Thus, Cpn0797 represents the third C. pneumoniae protein secreted into the host cell cytosol experimentally identified so far.


Asunto(s)
Antígenos Bacterianos/metabolismo , Chlamydophila pneumoniae/inmunología , Citoplasma/metabolismo , Citoplasma/microbiología , Sistemas de Lectura Abierta , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/aislamiento & purificación , Chlamydophila pneumoniae/patogenicidad , Citoplasma/inmunología , Citosol/inmunología , Citosol/metabolismo , Citosol/microbiología , Células HeLa , Humanos , Sistemas de Lectura Abierta/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA