首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 52 毫秒
1.
2.
The hepatitis A virus cellular receptor 1 (HAVcr-1) cDNA codes for a class I integral membrane glycoprotein, termed havcr-1, of unknown natural function which serves as an African green monkey kidney (AGMK) cell receptor for HAV. The extracellular domain of havcr-1 has an N-terminal Cys-rich region that displays homology with sequences of members of the immunoglobulin superfamily, followed by a Thr/Ser/Pro (TSP)-rich region characteristic of mucin-like O-glycosylated proteins. The havcr-1 glycoprotein contains four putative N-glycosylation sites, two in the Cys-rich region and two in the TSP-rich region. To characterize havcr-1 and define region(s) involved in HAV receptor function, we expressed the TSP-rich region in Escherichia coli fused to glutathione S-transferase and generated antibodies (Ab) in rabbits (anti-GST2 Ab). Western blot analysis with anti-GST2 Ab detected 62- and 65-kDa bands in AGMK cells and 59-, 62-, and 65-kDa bands in dog cells transfected with the HAVcr-1 cDNA (cr5 cells) but not in dog cells transfected with the vector alone (DR2 cells). Treatment of AGMK and cr5 cell extracts with peptide-N-glycosidase F resulted in the collapse of the havcr-1-specific bands into a single band of 56 kDa, which indicated that different N-glycosylated forms of havcr-1 were expressed in these cells. Treatment of AGMK and cr5 cells with tunicamycin reduced binding of protective monoclonal Ab (MAb) 190/4, which suggested that N-glycans are required for binding of MAb 190/4 to havcr-1. To test this hypothesis, havcr-1 mutants lacking the N-glycosylation motif at the first site (mut1), second site (mut2), and both (mut3) sites were constructed and transfected into dog cells. Binding of MAb 190/4 and HAV to mut1 and mut3 cells was highly reduced, while binding to mut2 cells was not affected and binding to dog cells expressing an havcr-1 construct containing a deletion of the Cys-rich region (d1− cells) was undetectable. HAV-infected cr5 and mut2 cells but not mut1, mut3, d1−, and DR2 cells developed the characteristic cytoplasmic granular fluorescence of HAV-infected cells. These results indicate that the Cys-rich region of havcr-1 and its first N-glycosylation site are required for binding of protective MAb 190/4 and HAV receptor function.

Viral hepatitis is a major public health problem, with estimated annual medical costs of billions of dollars. The Center for Disease Control and Prevention estimated that in the United States alone, hepatitis A virus (HAV), the causative agent of acute hepatitis in humans, produces substantial morbidity and mortality, with an estimated 125,000 to 200,000 infections occurring each year and approximately 100 deaths from fulminant hepatitis. HAV is the only member of the hepatovirus genus of the Picornaviridae, a family of small, nonenveloped, positive-strand RNA viruses that include human pathogens such as poliovirus (PV) and rhinovirus as well as animal pathogens such as foot-and-mouth disease virus and encephalomyocarditis virus. Hepatitis A is transmitted via the oral-fecal route and can be prevented by vaccination with cell culture-adapted formalin-inactivated HAV (6, 22). The HAV RNA genome of about 7,500 nucleotides (nt) is covalently linked to the small virus-encoded VPg protein at its 5′ end (21) and has a poly(A) tail at its 3′ end. The approximately 750-nt long 5′ nontranslated region of the HAV genome codes for a long and complex internal ribosome entry site which directs the cap-independent translation of the viral message (reference ;9; and references therein). The HAV mRNA contains a single long open reading frame, which is translated into a polyprotein from which the structural proteins VP0, VP3, and VP1 and nonstructural proteins are cleaved by 3Cpro, the only HAV-encoded protease (8, 17). Sixty copies of VP0, VP3, and VP1 assemble into viral capsids, which, in association with the HAV genome, form provirions that undergo a slow RNA-dependent maturation cleavage of VP0 into VP4 and VP2 (2). VP4 of HAV is a very small protein of 21 to 23 amino acids which, in contrast to VP4s of all other picornaviruses, has not yet been found in the viral capsid (5, 12, 19).Although there have been major advances in our knowledge about human hepatitis viruses, very little is known about the mechanisms of their cell entry. Cellular receptors for human hepatitis viruses have been difficult to characterize due to poor in vitro viral growth, association of virions with serum and cell-derived materials which mask genuine virus-receptor interactions leading to cell entry, and attachment of virions to susceptible and nonsusceptible cells. We identified havcr-1 as an African green monkey kidney (AGMK) cellular receptor for HAV using protective monoclonal antibody (MAb) 190/4 as a probe (10). Ashida and Hamada recently identified a protein very similar to havcr-1 in S.la/Ve-1 cells, hybrid cells between marmoset liver and Vero cells, as an HAV receptor using the independently derived protective MAb 2H4 to screen a cDNA library (1). Nucleotide sequence analysis revealed that the HAVcr-1 cDNA codes for a novel mucin-like class I integral membrane glycoprotein, termed havcr-1, whose extracellular domain contains four putative N-glycosylation sites and two distinctive regions: an N-terminal Cys-rich region that displays homology to sequences of members of the immunoglobulin superfamily, and a mucin-like C-terminal region containing 27 repeats of the consensus PTTTTL. Our knowledge about the interaction of HAV with havcr-1 is currently limited, and the natural function(s) and ligand(s) of this receptor are unknown. In this work we characterize different species of havcr-1 migrating between 59 and 65 kDa as N-glycosylated forms of a 56-kDa band present after removal of N-glycans with peptide-N-glycosidase F (PNGase F). We also determined, using N-glycosylation and deletion mutants, that the havcr-1 Cys-rich region and its first N-glycosylation site are required for HAV receptor function. Further characterization of havcr-1 and the HAV–havcr-1 interaction will help us to understand the mechanism of cell entry of HAV and possibly to develop drugs which can prevent such interaction.  相似文献   

3.
4.
5.
6.
Unsaturated glucuronyl hydrolase (UGL) categorized into the glycoside hydrolase family 88 catalyzes the hydrolytic release of an unsaturated glucuronic acid from glycosaminoglycan disaccharides, which are produced from mammalian extracellular matrices through the β-elimination reaction of polysaccharide lyases. Here, we show enzyme characteristics of pathogenic streptococcal UGLs and structural determinants for the enzyme substrate specificity. The putative genes for UGL and phosphotransferase system for amino sugar, a component of glycosaminoglycans, are assembled into a cluster in the genome of pyogenic and hemolytic streptococci such as Streptococcus agalactiae, Streptococcus pneumoniae, and Streptococcus pyogenes, which produce extracellular hyaluronate lyase as a virulent factor. The UGLs of these three streptococci were overexpressed in Escherichia coli cells, purified, and characterized. Streptococcal UGLs degraded unsaturated hyaluronate and chondroitin disaccharides most efficiently at approximately pH 5.5 and 37 °C. Distinct from Bacillus sp. GL1 UGL, streptococcal UGLs preferred sulfated substrates. DNA microarray and Western blotting indicated that the enzyme was constitutively expressed in S. agalactiae cells, although the expression level increased in the presence of glycosaminoglycan. The crystal structure of S. agalactiae UGL (SagUGL) was determined at 1.75 Å resolution by x-ray crystallography. SagUGL adopts α66-barrel structure as a basic scaffold similar to Bacillus UGL, but the arrangement of amino acid residues in the active site differs between the two. SagUGL Arg-236 was found to be one of the residues involved in its activity for the sulfated substrate through structural comparison and site-directed mutagenesis. This is the first report on the structure and function of streptococcal UGLs.Cell surface polysaccharides play an important role in linking neighboring cells and protecting cells against physicochemical stress such as osmotic pressure or invasion by pathogens. Glycosaminoglycans such as chondroitin, hyaluronan, and heparin are highly negatively charged polysaccharides with a repeating disaccharide unit consisting of an uronic acid residue (glucuronic or iduronic acid) and an amino sugar residue (glucosamine or galactosamine) (1), and they are widely present in mammalian cells as an extracellular matrix responsible for cell-to-cell association, cell signaling, and cell growth and differentiation (2). For example, in humans, glycosaminoglycans exist in tissues such as the eye, brain, liver, skin, and blood (3). Except for hyaluronan, glycosaminoglycans such as chondroitin sulfate, dermatan sulfate, keratan sulfate, heparin sulfate, and heparan sulfate are often sulfated. Chondroitin consists of d-glucuronic acid (GlcA)2 and N-acetyl-d-galactosamine (GalNAc) with a sulfate group(s) at position 4 or 6 or both (4). Hyaluronan, is composed of GlcA and N-acetyl-d-glucosamine (GlcNAc) (5).The adhesion of pathogenic bacteria to mammalian cells is regarded as a primary mechanism of bacterial infection, followed by secondary effects of the infectious process. Polysaccharides, including the glycosaminoglycans that form part of the cell surface matrix, are typical targets for microbial pathogens that invade host cells, and many specific interactions between pathogens and these polysaccharides have been described (6). Glycosaminoglycans in the extracellular matrix are also degraded enzymatically by hydrolases and lyases (1). Generally, hydrolases cleave the glycoside bonds between the glycosyl oxygen and the anomeric carbon atom through the addition of water and play an important role in glycosaminoglycan metabolism in mammals (7). On the other hand, bacterial pathogens invading host cells degrade glycosaminoglycans through the action of lyases. Bacterial polysaccharide lyases recognize the uronic acid residue in polysaccharides, cleave the glycoside bonds through the β-elimination reaction without water addition, and produce unsaturated saccharides with the unsaturated uronic acid residue having a CC double bond at the nonreducing terminus (8).Streptococci such as group B Streptococcus agalactiae, group nonassigned Streptococcus pneumoniae, and group A Streptococcus pyogenes are typical pyogenic and hemolytic pathogens causing severe infections (e.g. pneumonia, bacteremia, sinusitis, or meningitis) (911). In S. pneumoniae, hyaluronate lyase, neuraminidases, autolysin, choline-binding protein A, and pneumococcal surface protein A are suggested to function as cell surface virulent factors (12). Hyaluronate lyase degrades the extracellular matrix component hyaluronan in mammalian cells through the β-elimination reaction and releases unsaturated disaccharide, indicating that the enzyme produced by pathogenic bacteria functions as a spreading factor (13). Because hyaluronate lyase is commonly produced by the three pyogenic and hemolytic streptococci (1416), the structure and function of their enzymes have been intensively studied (17, 18). Groups A, B, C, and G streptococci also produce hyaluronate lyase (19), suggesting that the enzyme is ubiquitously present in pathogenic streptococci. Streptococcal hyaluronate lyase can also act on sulfated and nonsulfated chondroitin (20). The metabolism of the resultant unsaturated disaccharides in streptococci, however, remains to be clarified.Unsaturated glucuronyl hydrolase (UGL), a member of the glycoside hydrolase family 88 in the CAZY data base (21), acts on unsaturated oligosaccharides having an unsaturated GlcA (ΔGlcA) with β-glycoside bond, such as ΔGlcA-GalNAc produced by chondroitin lyase and ΔGlcA-GlcNAc produced by hyaluronate lyase (22) (Fig. 1A). We have first identified the UGL-coding gene in Bacillus sp. GL1 (23) and clarified the structure and function of the enzyme by x-ray crystallography (2427). The enzyme reaction generates ΔGlcA and the leaving saccharide. ΔGlcA is spontaneously converted to 4-deoxy-1-threo-5-hexosulose-uronate (Fig. 1A) because the ringed form of ΔGlcA has not been obtained because of keto-enole equilibrium (23, 28). In contrast with general glycoside hydrolases with retention or inversion catalytic mechanism of an anomeric configuration, UGL uniquely triggers hydrolysis of vinyl ether groups in unsaturated saccharides but not of the glycoside bond (26) (Fig. 1B). This article deals with the characteristics of streptococcal UGLs by using recombinant enzymes, gene expression in S. agalactiae cells by DNA microarray, and structural determinants of S. agalactiae UGL for substrate specificity by x-ray crystallography and site-directed mutagenesis.Open in a separate windowFIGURE 1.UGL reaction. A, degradation scheme of Δ6S by UGL. B, catalytic reaction mechanism of UGL. C, structures of unsaturated oligosaccharides. ΔGellan, unsaturated gellan tetrasaccharide; ΔHA, unsaturated hyaluronan disaccharide; Δ0S, unsaturated chondroitin disaccharide; Δ2′S, unsaturated chondroitin disaccharide sulfated at C-2 position of ΔGlcA residue; Δ2′S4S, unsaturated chondroitin disaccharide sulfated at C-2 position of ΔGlcA residue and C-4 position of GalNAc residue; Δ2′S6S, unsaturated chondroitin disaccharide sulfated at C-2 position of ΔGlcA residue and C-6 position of GalNAc residue; Δ4S6S, unsaturated chondroitin disaccharide sulfated at C-4 and C-6 positions of GalNAc residue; Δ2′S4S6S, unsaturated chondroitin disaccharide sulfated at C-2 position of ΔGlcA residue and C-4 and C-6 positions of GalNAc residue.  相似文献   

7.
8.
9.
Bazdar DA  Sieg SF 《Journal of virology》2007,81(22):12670-12674
Proliferation responses of naïve CD4+ T cells to T-cell receptor and interleukin-7 (IL-7) stimulation were evaluated by using cells from human immunodeficiency virus-positive (HIV+) donors. IL-7 enhanced responses to T-cell receptor stimulation, and the magnitude of this enhancement was similar in cells from healthy controls and from HIV+ subjects. The overall response to T-cell receptor stimulation alone or in combination with IL-7, however, was diminished among viremic HIV+ donors and occurred independent of antigen-presenting cells. Frequencies of CD127+ cells were related to the magnitudes of proliferation enhancement that were mediated by IL-7. Thus, IL-7 enhances but does not fully restore the function of naïve CD4+ T cells from HIV-infected persons.Interleukin-7 (IL-7) plays an important role in T-cell homeostasis by modulating thymic output (1, 16, 22) and by enhancing the peripheral expansion and survival of both naïve and memory T-cell subsets (12, 18, 20, 25, 26, 31, 32). Under normal circumstances, the homeostatic maintenance of naïve CD4+ T cells is regulated by at least two types of signals that include T-cell receptor (TCR) engagement and IL-7 (10, 26, 30). In addition, IL-7 may play an important role in the conversion of effector T cells into long-term memory cells (12, 14).Homeostasis of T cells is dysregulated in human immunodeficiency virus (HIV) infection such that there is a marked depletion of CD4+ cells and a progressive loss of naïve CD4 and CD8+ T cells (24). Although the mechanisms for these deficiencies are not fully understood, it is possible that impairments in T-cell proliferation and responsiveness to immunomodulatory cytokines could play a role. In HIV disease, IL-7 is increased in plasma (2, 5, 11, 15, 19, 21, 23) and the alpha chain of the IL-7 receptor, CD127, is less frequently expressed among T lymphocytes (2, 5, 11, 21, 23). The ability of patient T cells to respond to IL-7 stimulation may be diminished in HIV disease but may not be related to the density of CD127 expression as it is in T cells from healthy controls (4). Moreover, the responsiveness of T cells, including naïve CD4+ lymphocytes, to TCR stimulation is diminished in HIV disease (27-29). Thus, defects in responsiveness to cytokines or TCR stimulation could contribute to the perturbations in T-cell proliferation and survival in HIV disease.In these studies, we examined the responsiveness of naïve CD4+ T cells from viremic HIV-positive (HIV+) donors (median plasma HIV RNA level, 25,200 copies/ml [range, 1,015 to 1,000,000 copies/ml]; median CD4 cell count, 429 cells/μl [range, 41 to 950 cells/μl]; median age, 38 years [range, 22 to 64 years]; n = 25) and aviremic, highly active antiretroviral therapy (HAART)-treated HIV+ donors (plasma HIV RNA level, <400 copies/ml; median CD4 cell count, 309 cells/μl [range, 74 to 918 cells/μl]; median age, 48 years [range, 37 to 55 years]; n = 12) to the combined stimulus of recombinant IL-7 (Cytheris) plus agonistic anti-CD3 antibody. Peripheral blood mononuclear cells (PBMC) were depleted of CD45RO+ cells by magnetic bead depletion (>90% purity) and were incubated in medium alone or were stimulated with anti-CD3 antibody, IL-7, or anti-CD3 antibody plus IL-7. CD4+CD45ROCD28+CD27+ cells were assessed for the expression of Ki67 2 days poststimulation by flow cytometric analyses. The addition of IL-7 to anti-CD3 antibody enhanced the induction of Ki67 expression in cells from both HIV+ and HIV-negative (HIV) donors (Fig. (Fig.11 and Fig. Fig.2).2). A diminished response to anti-CD3 antibody was observed among naïve CD4+ T cells from viremic HIV+ donors. In contrast, cells from aviremic HIV+ donors (all receiving antiretroviral therapy) had normal responses to anti-CD3 antibody compared to cells from healthy donors (Fig. (Fig.2).2). Importantly, the addition of IL-7 to the cultures significantly improved the responses to above those observed with anti-CD3 alone in HIV and HIV+ donors, regardless of viremia (Wilcoxon signed ranks test; for each comparison, P was <0.04), and the magnitude of that enhancement, although slightly diminished in cells from HIV+ donors, was not significantly different between groups of subjects when measured as either the enhancement (n-fold; not shown) or as the change in percent Ki67+ cells above the background observed for cells stimulated with anti-CD3 alone (Fig. (Fig.3).3). Although IL-7 enhanced responses to TCR stimulation in HIV subjects, the overall magnitude of the responses among cells from HIV viremic subjects did not reach the levels seen with cells from healthy donors, even in the presence of IL-7 (Fig. (Fig.2).2). It should be noted, however, that these functional readouts were not related to clinical indices of plasma HIV RNA level, CD4 cell count, or age when considered as continuous variables, suggesting that the functional perturbations in naïve CD4+ T cells are probably undermined by complexities extending beyond HIV replication (not shown). Together, these results suggest that TCR responsiveness is diminished in naïve CD4+ T cells from viremic HIV+ subjects, whereas responsiveness to IL-7 stimulation is relatively preserved.Open in a separate windowFIG. 1.IL-7 enhances the induction of Ki67 expression in naïve CD4+ T cells from healthy controls and HIV+ donors. CD45RO-depleted PBMC were incubated with anti-CD3 antibody (100 ng/ml), IL-7 (50 ng/ml), anti-CD3 antibody plus IL-7, or medium alone (RPMI with 10% fetal bovine serum). Cells were gated on CD4+CD27+CD28+ lymphocytes and examined for Ki67 expression by intracellular flow cytometry.Open in a separate windowFIG. 2.IL-7 responsiveness in cells from viremic and aviremic HIV+ donors. Plotted values represent the percentages of CD4+CD27+CD28+CD45RO T cells that expressed Ki67 after a 2-day incubation with anti-CD3 or with anti-CD3 plus IL-7. Percentages of Ki67+ cells in cultures without stimulation or with IL-7 only were subtracted from the values shown. Responses of cells from healthy controls (n = 9), HIV+ subjects with plasma HIV RNA levels of >400 copies/ml (n = 25), and HIV+ subjects on HAART with suppressed viral replication (<400 copies/ml; n = 12) are shown. Statistically significant differences between cells from controls and HIV+ donors are indicated. Analyses included Kruskal-Wallis test (P = 0.002) for multigroup comparisons and Mann-Whitney U test for comparison of two groups (*, P < 0.05).Open in a separate windowFIG. 3.IL-7 enhances responses to anti-CD3 antibody stimulation to a similar degree in cells from HIV+ and HIV donors. Naïve CD4+ T cells were incubated with IL-7, anti-CD3, anti-CD3 plus IL-7, or medium alone for 2 days. Background division (percent Ki67+ cells) in medium alone or IL-7 alone was first subtracted from the responses observed with cells stimulated with anti-CD3 alone or with anti-CD3 plus IL-7, respectively. The magnitude of IL-7 enhancement was then calculated by subtracting the percentage of naïve CD4+ cells that expressed Ki67+ after anti-CD3 antibody stimulation from the percentage of naïve CD4+ cells that expressed Ki67 after stimulation with anti-CD3 plus IL-7. n = 9, 25, and 12 for healthy controls, viremic subjects, and aviremic subjects, respectively.Previous studies indicate that the frequency of CD127+ T cells, particularly memory T-cell subsets, is reduced in patients with HIV disease (5, 11, 21, 23). This could, in part, result from the modulation of receptor expression through increased exposure to IL-7 in vivo and also may reflect accumulation of CD127 effector memory cells (21). We assessed the expression of CD127 in naïve CD4+CD45RA+CD28+CD27+ and memory CD4+CD45RO+ T cells in a subset of patients and asked if the frequencies of CD127+ cells were related to the induction of Ki67 expression by anti-CD3 or by anti-CD3 plus IL-7 among naïve CD4+ T cells. We reasoned that the ability of IL-7 to enhance responses to TCR stimulation might be limited if CD127 expression was diminished among naïve CD4+ T cells from HIV+ donors. Alternatively, a defect in functional responses also could be related to increased exposure to IL-7 in vivo, as may be reflected by the absence of CD127 receptor expression on memory T-cell subsets.In agreement with previous studies, our results suggest that CD127 expression is relatively preserved in naïve CD4+ T cells from HIV+ donors (representative histograms in Fig. Fig.4)4) (mean percentage of CD127+ cells, 87 and 83 for HIV donors [n = 5] and HIV+ donors [n = 17], respectively; P = 0.96) but is diminished in memory CD4+ T cells from HIV+ donors (mean percentage of CD127+ cells, 83 and 59 for HIV and HIV+ donors, respectively; P = 0.01). The frequencies of CD127+ naïve T cells were directly related to the frequencies of CD127+ memory T cells (Spearman''s correlations; r = 0.711, P = 0.001; n = 18) in HIV+ subjects. This result suggests that a similar mechanism modulates the expression of CD127 in these T-cell subsets, even though the loss of CD127 expression is clearly greater among the memory T cells in HIV disease. Neither CD127 expression among naïve CD4+ T cells nor CD127 expression among memory CD4+ T cells was related to the functional response of naïve CD4+ T cells to anti-CD3 (r = 0.238 and P = 0.36 for naïve CD127 expression; r = 0.293 and P = 0.25 for memory CD127 expression) or to anti-CD3 plus IL-7 (r = 0.32 and P = 0.21 for naïve CD127 expression; r = 0.31 and P = 0.22 for memory CD127 expression). There was a relationship between the percentage of CD127+ naïve T cells and the delta Ki67 expression that resulted from the addition of IL-7 to anti-CD3-treated cultures (percentage of Ki67+ cells in cultures treated with anti-CD3 plus IL-7 minus the percentage of Ki67+ cells in cultures treated with anti-CD3 alone) (Fig. (Fig.4).4). This relationship was statistically significant by Pearson''s correlation (r = 0.5, P = 0.041), the use of which was justified based on the normal distribution of the data. Spearman''s analysis, which is independent of data distribution, indicated a similar trend that was not statistically significant (r = 0.41, P = 0.1). The mean fluorescence intensity of CD127 expression on CD4+CD45RA+CD27+CD28+ T cells was not significantly related to the delta Ki67 expression induced by IL-7 but also suggested a trend consistent with a direct relationship between these indices (r = 0.45 and P = 0.07 by Pearson''s correlation; r = 0.34 and P = 0.18 by Spearman''s correlation). Despite the relative preservation of IL-7 receptor in naïve CD4+ T cells from HIV+ donors, the association between the frequencies of CD127+ cells and CD4+ T-cell proliferation responses to TCR plus IL-7 suggests that subtle IL-7 receptor perturbations might contribute to functional defects of naïve CD4+ T cells in HIV-infected persons.Open in a separate windowFIG. 4.CD127 receptor expression is related to enhancement of proliferation by IL-7. (A) Whole blood from a healthy control and an HIV-infected person was examined by flow cytometry for expression of CD127 on CD4+CD45RA+CD27+CD28+ (naïve) T cells. The gating strategy for identifying naïve cells involved an initial gate for lymphocyte forward and side scatter (SSC) characteristics (not shown) and then sequential gates for CD4 positive, CD45RA positive and, finally, CD28+CD27+ double-positive cells. (B) Plotted values indicating the relationship between the delta Ki67 expression in naïve CD4+ T cells and the percentage of CD127+ naïve T cells that was determined by using freshly isolated whole blood. The delta Ki67 expression was calculated by subtracting the percentage of naïve CD4+ cells that expressed Ki67+ after anti-CD3 antibody stimulation from the percentage of naïve CD4+ cells that expressed Ki67 after stimulation with anti-CD3 plus IL-7.To consider the possibility that antigen-presenting cells could contribute to the diminished response of T-cells to stimulation with TCR plus IL-7, we next asked if defects in TCR-plus-IL-7 stimulation could be detected in purified naïve CD4+ T-cell populations. CD4+CD45RO cells were negatively selected by magnetic bead depletion, achieving a purity of >90% as determined by flow cytometric analyses. Purified naïve CD4+ T cells were labeled with carboxy fluorescein succinimidyl ester (CFSE) tracking dye and incubated with IL-7, anti-CD3 antibody that was immobilized on a plate, anti-CD3 plus IL-7, or medium alone. The induction of proliferation was measured 7 days later by the dilution of CFSE tracking dye among CD4+CD27+ cells by calculating the division index (average number of cell divisions of all CD4+CD27+ cells) and the proliferation index (average number of divisions of CD4+CD27+ cells that had diluted tracking dye; Flow-Jo analysis software). These purified CD4+ T cells proliferated poorly in response to anti-CD3 antibody stimulation alone, providing functional evidence that the samples were free of antigen-presenting cell contamination (Fig. (Fig.5A).5A). The combined treatment of anti-CD3 and IL-7 induced cellular expansion, whereas alone, neither stimulus induced cellular proliferation during the 7-day period (Fig. (Fig.5A).5A). Responses of cells from HIV+ donors were reduced compared to those of cells from healthy donors, confirming that the defects in naïve CD4+ T-cell expansion are independent of antigen-presenting cells and not fully corrected by IL-7 (Fig. (Fig.5B5B).Open in a separate windowFIG. 5.Diminished responses to TCR plus IL-7 in purified naïve CD4+ T cells from HIV+ donors. CD4+CD45RO cells were purified from PBMC by negative selection. Cells from HIV+ donors (n = 7) and healthy controls (n = 7) were labeled with CSFE and incubated with anti-CD3 immobilized on a plate (5 μg/ml, overnight at 4°C) plus IL-7 (10 ng/ml). CFSE dye dilution was measured among the CD4+CD27+ cells. (A) Representative histograms showing the dilution of CFSE and CD27 expression among cells incubated with anti-CD3 antibody alone, IL-7 alone, or the combination of anti-CD3 plus IL-7. Placements of quadrant gates were based on an isotype control antibody stain (for CD27 expression) and on cells that had been incubated in medium alone (for CFSE dye dilution). (B) Division indices (average number of cell divisions among CD4+CD27+ cells) and proliferation indices (average number of cell divisions among CD4+CD27+ cells that had diluted tracking dye) are shown.IL-7 is a promising candidate for therapeutic and vaccine adjuvant applications in HIV disease. This cytokine may be especially beneficial in circumstances of immune reconstitution, since it normally plays an essential role in T-cell proliferation and survival. Here, we demonstrate that IL-7 efficiently enhances TCR-triggered naïve CD4+ T-cell expansion in cells from healthy individuals and from HIV+ donors. The mechanism of IL-7 activity is not discerned in these experiments but may involve effects on survival, such as the induction of Bcl-2 (9), or may involve the enhancement of IL-2 or IL-2 receptor expression (6, 8). In any case, our studies provide evidence that IL-7 should provide an effective therapy for the regulation of naïve CD4+ T-cell homeostasis and may be useful for vaccine adjuvant applications in HIV disease. The potential of this approach has been illustrated by recent human trials of IL-7 that demonstrated the expansion of naïve T cells in vivo after IL-7 administration to HIV-infected persons (13) and by animal studies, wherein IL-7 administration enhanced T-cell responses to immunization in mice (17).Notably, the depletion studies and purification methods employed here did not necessarily eliminate terminally differentiated effector memory CD4+ T cells from our cultures; however, studies of CMV-specific terminally differentiated cells suggested that these cells are primarily CD27 (3), and the use of three markers to identify naïve CD4+ T cells, including the ones used here (CD27, CD28, and CD45RO) is estimated to provide 98% assurance that the cells being examined are truly naïve (7). Thus, it is likely that terminally differentiated cells were largely removed from our analyses.Our observations provide confirmation of a significant defect in the responses of naïve CD4+ T cells to TCR triggering in HIV disease, and this defect is not fully corrected by IL-7, as shown here, or by IL-2, as we demonstrated previously (27). These deficiencies are reproduced even among naïve CD4+ T cells that are purified from professional antigen-presenting cells, indicating that the defects are intrinsic to the T cells and not a consequence of dysfunctional antigen-presenting cells. We propose that functional defects in naïve CD4+ T cells from HIV+ donors stem primarily from deficiencies in TCR signaling. Further studies that define the nature of naïve CD4+ T-cell defects in HIV disease will be required to address the underlying mechanisms.  相似文献   

10.
11.
12.
13.
14.
15.
Hepatitis A virus (HAV), an atypical member of the Picornaviridae, grows poorly in cell culture. To define determinants of HAV growth, we introduced a blasticidin (Bsd) resistance gene into the virus genome and selected variants that grew at high concentrations of Bsd. The mutants grew fast and had increased rates of RNA replication and translation but did not produce significantly higher virus yields. Nucleotide sequence analysis and reverse genetic studies revealed that a T6069G change resulting in a F42L amino acid substitution in the viral polymerase (3Dpol) was required for growth at high Bsd concentrations whereas a silent C7027T mutation enhanced the growth rate. Here, we identified a novel determinant(s) in 3Dpol that controls the kinetics of HAV growth.Hepatitis A virus (HAV) is an atypical member of the Picornaviridae that replicates poorly in cell culture and generally does not cause cytopathic effect (CPE). The HAV positive-strand RNA genome of about 7.5 kb is encapsidated in a 27- to 32-nm icosahedral shell (12). The HAV genome contains a long open reading frame (ORF) that codes for a polyprotein of approximately 250 kDa, which undergoes co- and posttranslational processing by the virus-encoded protease 3Cpro into structural (VP0, VP3, and VP1-2A) and nonstructural proteins (11, 13, 14, 18). VP0 undergoes structural cleavage into VP2 and VP4, and an unknown cellular protease cleaves the VP1-2A precursor (9, 23).HAV replicates inefficiently in cell culture and in general establishes persistent infections (3, 4, 7, 8) without causing CPE. However, some strains of HAV that replicate quickly can induce cell death (5, 19, 27). Due to the growth limitations, experimentation with HAV is difficult and the biology of this virus is poorly understood. To facilitate genetic studies, we recently introduced a blasticidin (Bsd) resistance gene at the 2A-2B junction of wild-type (wt) HAV (16). Bsd, an antibiotic that blocks translation in prokaryotes and eukaryotes and thus affects HAV translation, is inactivated by the Bsd-deaminase encoded in the Bsd resistance gene (15). Cells infected with the wt HAV construct carrying the Bsd resistance gene (HAV-Bsd) grew in the presence of Bsd. We have recently used the wt HAV-Bsd construct to select human hepatoma cell lines that support the stable growth of wt HAV (16) and to establish simple and rapid neutralization and virus titration assays (17). In this study, we developed a genetic approach to study determinants of HAV replication based on the selection of HAV-Bsd variants grown under increased concentrations of Bsd. We hypothesized that by increasing the concentration of Bsd, we would select HAV variants that grew better and allowed the survival of persistently infected cells at higher concentrations of the antibiotic. We also reasoned that we would need a robust HAV-Bsd replication system to provide enough Bsd-deaminase for cell survival. Therefore, we used attenuated HAV grown in rhesus monkey fetal kidney FRhK4 cells as an experimental system because (i) the virus grows 100-fold better in this system than wt HAV in human hepatoma cells (16), and (ii) it already contains cell culture-adapting mutations (3, 4, 7, 8) that are likely to accumulate during passage of wt HAV at high concentrations of Bsd and confound our analysis.  相似文献   

16.
Nicotinic acetylcholine receptor (nAChR) agonists, such as epibatidine and its molecular derivatives, are potential therapeutic agents for a variety of neurological disorders. In order to identify determinants for subtype-selective agonist binding, it is important to determine whether an agonist binds in a common orientation in different nAChR subtypes. To compare the mode of binding of epibatidine in a muscle and a neuronal nAChR, we photolabeled Torpedo α2βγδ and expressed human α4β2 nAChRs with [3H]epibatidine and identified by Edman degradation the photolabeled amino acids. Irradiation at 254 nm resulted in photolabeling of αTyr198 in agonist binding site Segment C of the principal (+) face in both α subunits and of γLeu109 and γTyr117 in Segment E of the complementary (−) face, with no labeling detected in the δ subunit. For affinity-purified α4β2 nAChRs, [3H]epibatidine photolabeled α4Tyr195 (equivalent to Torpedo αTyr190) in Segment C as well as β2Val111 and β2Ser113 in Segment E (equivalent to Torpedo γLeu109 and γTyr111, respectively). Consideration of the location of the photolabeled amino acids in homology models of the nAChRs based upon the acetylcholine-binding protein structure and the results of ligand docking simulations suggests that epibatidine binds in a single preferred orientation within the α-γ transmitter binding site, whereas it binds in two distinct orientations in the α4β2 nAChR.Nicotinic acetylcholine receptors (nAChRs)3 are prototypical members of the Cys loop superfamily of neurotransmitter-gated ion channels that mediate the actions of the neurotransmitter acetylcholine (1). nAChRs from vertebrate skeletal muscle and the electric organs of Torpedo rays are heteropentamers of homologous subunits with a stoichiometry of 2α:β:γ(ϵ):δ that are arranged pseudosymmetrically around central cation-selective ion channels (1, 2). There are 12 mammalian neuronal nAChR subunit genes: nine neuronal α subunits (α2–α10) and three neuronal β subunits (β2–β4). The α4β2 nAChR is the most abundant and widely distributed nAChR subtype expressed in the brain and is a major target for potential therapeutic agents for neurological diseases and conditions, including nicotine dependence and Alzheimer and Parkinson diseases (3, 4). Although the ratio of α4 to β2 subunit in vivo is uncertain, expressed receptors containing either three α4 or three β2 subunits have distinct pharmacological properties (5, 6).The agonist binding sites (ABS) of nAChRs are located within the amino-terminal extracellular domain at the interface of adjacent subunits (α-γ and α-δ in the Torpedo nAChR), and different nAChR subunit combinations form ABS with distinct physical and pharmacological properties (3, 7). Affinity labeling studies with Torpedo nAChR and site-directed mutational analyses of muscle and neuronal nAChRs identified key amino acids delineating the ABS from three noncontiguous stretches of the α subunit (Segments A-C, the principal component (+ face)) and three noncontiguous regions of the non-α subunit (Segments D–F, the complementary component (− face)) (8, 9). The three-dimensional structure of the ABS in the absence and presence of nAChR agonists or competitive antagonists has been determined for snail acetylcholine-binding proteins (AChBPs) that are soluble homopentamers homologous to the extracellular (amino-terminal) domain of a nAChR (1012). In the AChBP, four aromatic amino acids from Segments A–C that are conserved within α subunits, along with a conserved Trp in Segment D, form a core aromatic “pocket” with a dimension optimal for accommodation of a trimethylammonium group. The other amino acids in the non-α subunits closest to the aromatic pocket, which are generally not conserved among γ, δ, or neuronal β subunits, are on three antiparallel β strands. The AChBP structure was used to refine the structure of the Torpedo nAChR in the absence of agonist to 4 Å resolution (13). In this structure, there is a reorientation of Segments A–C, resulting in the absence of a well defined core aromatic binding pocket.Analysis of agonist interactions with mutant nAChRs containing fluorine-substituted core aromatic residues provides evidence that cation-π interactions, particularly with αTrp149 in Segment B, are important determinants of agonist binding affinity (14) and for the higher affinity binding of nicotine to α4β2 nAChRs compared with α2βγδ nAChRs (15). Mutational analyses and molecular docking calculations have also provided evidence that two molecules of very similar structure may actually bind to a single receptor in very different orientations, as seen for two high affinity antagonists, d-tubocurarine and its quaternary ammonium analog metocurine, binding to the AChBP and to the muscle nAChR (16, 17).Photoaffinity labeling provides an alternative means to identify amino acids contributing to a drug binding site (18, 19) and has been used to determine the orientation of drugs bound in the ABS of Torpedo nAChR (20). Epibatidine binds with very high affinity (∼10 pm) to heteromeric neuronal nAChRs (e.g. α4β2) and with nanomolar affinity to α7 and muscle-type/Torpedo nAChRs (3). Utilizing a photoreactive analogue of epibatidine (azidoepibatidine; Fig. 1) and mass spectrometry, Tomizawa et al. (21) identified photolabeled amino acids in the Aplysia AChBP (Tyr195 in Segment C and Met116 in Segment E), establishing an orientation for bound azidoepibatidine consistent with the orientation of epibatidine in an AChBP crystal structure (12).Open in a separate windowFIGURE 1.Structure of [3H]epibatidine (top) and azidoepibatidine (bottom).In this report, we use [3H]epibatidine as a photoaffinity reagent to identify the amino acids photolabeled in an expressed α4β2 nAChR and in the Torpedo α2βγδ nAChR. Comparisons of the labeled amino acids seen in the Torpedo nAChR α-γ binding site and in the α4β2 nAChR, in conjunction with the results of docking calculations for epibatidine binding to homology models of the α2βγδ and α4β2 nAChRs, suggests that epibatidine binds in a single orientation in the α-γ site but in two orientations in the α4β2 ABS.  相似文献   

17.
Hepatitis A virus (HAV) infects African green monkey kidney (AGMK) cells via the HAV cellular receptor-1 (havcr-1), a mucin-like type 1 integral-membrane glycoprotein of unknown natural function. The ectodomain of havcr-1 contains an N-terminal immunoglobulin-like cysteine-rich region (D1), which binds protective monoclonal antibody (MAb) 190/4, followed by an O-glycosylated mucin-like threonine-serine-proline-rich region that extends D1 well above the cell surface. To study the interaction of HAV with havcr-1, we constructed immunoadhesins fusing the hinge and Fc portion of human IgG1 to D1 (D1-Fc) or the ectodomain of the poliovirus receptor (PVR-Fc) and expressed them in CHO cells. These immunoadhesins were secreted to the cell culture medium and purified through protein A-agarose columns. In a solid-phase assay, HAV bound to D1-Fc in a concentration-dependent manner whereas background levels of HAV bound to PVR-Fc. Binding of HAV to D1-Fc was blocked by treatment with MAb 190/4 but not with control MAb M2, which binds to a tag epitope introduced between the D1 and Fc portions of the immunoadhesin. D1-Fc neutralized approximately 1 log unit of the HAV infectivity in AGMK cells, whereas PVR-Fc had no effect in the HAV titers. A similarly poor reduction in HAV titers was observed after treating the same stock of HAV with murine neutralizing MAbs K2-4F2, K3-4C8, and VHA 813. Neutralization of poliovirus by PVR-Fc but not by D1-Fc indicated that the virus-receptor interactions were specific. These results show that D1 is sufficient for binding and neutralization of HAV and provide further evidence that havcr-1 is a functional cellular receptor for HAV.  相似文献   

18.
19.
20.
The most common mutation in cystic fibrosis (CF) is ΔF508, which is associated with failure of the mutant cystic fibrosis transmembrane conductance regulator (CFTR) to traffic to the plasma membrane. By a still unknown mechanism, the loss of correctly trafficked ΔF508-CFTR results in an excess of the epithelial sodium channel (ENaC) on the apical plasma membrane. ENaC trafficking is known to be regulated by a signaling pathway involving the glucocorticoid receptor, the serum- and glucocorticoid-regulated kinase SGK1, and the ubiquitin E3 ligase Nedd4-2. We show here that dexamethasone rescues functional expression of ΔF508-CFTR. The half-life of ΔF508-CFTR is also dramatically enhanced. Dexamethasone-activated ΔF508-CFTR rescue is blocked either by the glucocorticoid receptor antagonist RU38486 or by the phosphatidylinositol 3-kinase inhibitor LY294002. Co-immunoprecipitation studies indicate that Nedd4-2 binds to both wild-type- and ΔF508-CFTR. These complexes are inhibited by dexamethasone treatment, and CFTR ubiquitination is concomitantly decreased. We further show that knockdown of Nedd4-2 by small interfering RNA also corrects ΔF508-CFTR trafficking. Conversely, knockdown of SGK1 by small interfering RNA completely blocks dexamethasone-activated ΔF508-CFTR rescue. These data suggest that the SGK1/Nedd4-2 signaling pathway regulates both CFTR and ENaC trafficking in CF epithelial cells.Cystic fibrosis (CF)2 is the most common life-limiting genetic disease in the United States and is due to mutations in the CFTR gene. The most common mutation, ΔF508-CFTR, results in a failure of the mutant protein to traffic properly to the apical plasma membrane of epithelial cells in the lung and other organs (1, 2). By a still unknown mechanism, the loss of correctly trafficked ΔF508-CFTR results in an excess of the epithelial sodium channel (ENaC) on the apical plasma membrane (35). In the CF lung, such high levels of ENaC activity are believed to cause dehydration of the airway, and the consequent proinflammatory condition that characterizes CF lung pathophysiology. Similar proinflammatory pathophysiology has been reported to characterize the lung of transgenic mice which overexpress β-ENaC (6). Operationally, it seems that when membrane-localized CFTR decreases in CF, ENaC activity at the plasma membrane increases; CF-related morbidity and mortality follow.In the case of ENaC trafficking, the process is known to be regulated by a glucocorticoid receptor/SGK1 signaling pathway affecting phosphorylation of the ubiquitin ligase E3 protein Nedd4-2 (7, 8). Fig. 1 illustrates how surface expression of ENaC is controlled by the serum- and glucocorticoid-inducible kinase SGK1, the upstream signal, and the ubiquitin E3 ligase Nedd4-2, the downstream signal. Under default conditions, Nedd4-2 suppresses ENaC surface expression by binding to ENaC via the interaction between the PPXY motifs of ENaC and WW domains on Nedd4-2. Nedd4-2 then catalyzes the ubiquitination of bound ENaC. This step targets ENaC for proteasomal degradation (9, 10). However, when Nedd4-2 is phosphorylated by SGK1, the default interaction between Nedd4-2 and ENaC is reduced, and ENaC is maintained at the plasma membrane (7, 8). The requirement for Nedd4-2 for destruction of ENaC is supported by the recent observation that siRNA against Nedd4-2 is sufficient to permit ENaC to be expressed at the plasma membrane (10). Importantly, both glucocorticoid receptor (GR) and phosphoinositide-3-kinase (PI 3-kinase) signaling pathways must be present for high levels of Na+ transport to occur. For example, treatment with the GR antagonist RU38486 (1113) or the PI 3-kinase inhibitor LY294002 (1416) results in a complete loss of glucocorticoid-activated ENaC activity.Open in a separate windowFIGURE 1.Schematic diagram of regulation of ENaC and CFTR by SGK1/Nedd4-2. The surface expression of ENaC is controlled by the serum/glucocorticoid inducible kinase SGK1, the upstream signal, and the neural precursor cell-expressed developmentally down-regulated isoform 2 (Nedd4-2), the downstream signal. The solid black arrows trace the signal to a point where phospho-Nedd4-2 releases ENaC, thereby saving it from default ubiquitination and proteasomal destruction. ENaC is then maintained at the plasma membrane. Glucocorticoid-activated ENaC membrane trafficking is blocked by the glucocorticoid receptor antagonist RU38486 and the PI 3-kinase inhibitor LY294002. Alternatively, silencing of endogenous Nedd4-2 by siRNA enhances ENaC trafficking to the plasma membrane. (+) indicates positive regulation, and (−) indicates negative regulation.The placement of the parenthetical (CFTR) in the SGK1/Nedd4-2 signaling pathway (Fig. 1) serves to underscore our hypothesis that CFTR itself could play an interactive or parallel role in the SGK1/Nedd4-2/ENaC-trafficking mechanism. This hypothesis seems reasonable because the regulatory effects of SGK1 and Nedd4-2 are not limited to trafficking of ENaC but also regulate several other epithelial channels and transporters (17, 18). Additionally, co-expression studies in Xenopus oocytes (19, 20) have shown that SGK1 appears to greatly enhance the functional activity of CFTR.In this report we have shown that activation of the SGK1 signaling pathway by the glucocorticoid dexamethasone results in the rescue of ΔF508-CFTR. The half-life of ΔF508-CFTR, once it reaches the plasma membrane, is also dramatically enhanced. Consistently, glucocorticoid-activated ΔF508-CFTR rescue is blocked by the GR antagonist RU38486 and by the PI 3-kinase inhibitor LY294002 as well as by knockdown of endogenous SGK1 by siRNA. We have further shown that at the downstream end of the SGK1/Nedd4-2 signaling pathway, knockdown of Nedd4-2 by siRNA also results in ΔF508-CFTR rescue. Finally, co-immunoprecipitation studies indicated that Nedd4-2 binds to both WT- and ΔF508-CFTR and that treatment with either glucocorticoid or Nedd4-2 siRNA reduces formation of Nedd4-2·CFTR complexes as well as ubiquitination of ΔF508-CFTR. Consistently, chloride transport is well correlated with the level of plasma membrane expression of ΔF508-CFTR protein. These data suggest that the glucocorticoid receptor-dependent SGK1/Nedd4-2 signaling pathway regulates both CFTR and ENaC trafficking in CF epithelial cells. We interpret these results to indicate that drugs affecting the SGK1/Nedd4-2 signaling pathway may be promising targets for cystic fibrosis therapeutic development.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号