13 research outputs found

    Sexual dimorphism of viscerocranium-A logistic model

    Get PDF
    Introduction: Sex estimates are generally based on the evaluation of qualitative and quantitative aspects of anatomic structures, however, the latter has better reproducibility and reliability. Objective: Aiming to evaluate the viscerocranium as a tool for sexual prediction and verify the possibility of creation of a logistic regression model for sexual prediction. Materials and Methods: 167 craniums - 100 male and 67 female between 22 and 85 years old from a Brazilian university´s Biobank - were evaluated. Results: It was observed that of the measures carried out were presented as sexually dimorphic, except for the measures of the right frontozygomatic point – right zygion; left frontozygomatic point – left zygion. Besides, it was possible to create a logistic regression model Sex = [logits/Sex = -24.5 + (0.20 * Nasion - Naso spine) + (0.18 * Right zygion - Naso spine)]. Conclusion: It was concluded that the measures of the viscerocranium present themselves as a factor of sexual dimorphism and the quantitative method developed was 81.4% accurate

    Cardiovirus 2A Protein Associates with 40S but Not 80S Ribosome Subunits during Infectionâ–¿

    No full text
    Host translation shutoff induced in picornavirus-infected cells is a well-known phenomenon. The mechanisms by which separate genera of the picornavirus family achieve this shutoff differ. This study examined alterations in the cellular translational components in HeLa cells infected with encephalomyocarditis virus (EMCV), a cardiovirus. In agreement with previous reports, EMCV induced a marked decrease in host mRNA translation. The inhibition correlated with the appearance of a significantly enhanced 80S peak in cells and a concomitant decrease in polysome abundance. Characterization of the 80S material revealed that these ribosomes were virtually devoid of mRNA. Viral protein 2A was tightly associated with some of the free 40S ribosome subunits, but it was not present in the 80S pool which accumulated after infection. Expression of 2A protein in cells in the absence infection was able to modulate the cellular translational environment to increase the ratio of internal ribosome entry site-dependent translation to cap-dependent translation of a reporter construct. The results provide further evidence for a role of 2A protein in the mechanism of cardiovirus-induced host translational shutoff

    Effect of genetic background and delivery route on the preclinical properties of a live attenuated RSV vaccine.

    No full text
    A safe and effective vaccine against RSV remains an important unmet public health need. Intranasally (IN) delivered live-attenuated vaccines represent the most extensively studied approach for immunization of RSV-naïve infants and children, however, achieving an effective balance of attenuation and immunogenicity has proven challenging. Here we report pre-clinical immunogenicity and efficacy data utilizing a live-attenuated vaccine candidate, RGΔM2-2, which was obtained by deleting the M2-2 open reading frame from the genome of the MSA1 clinical isolate. Intramuscular (IM) administration of RGΔM2-2 in cotton rats induced immunity and protective efficacy that was comparable to that induced by intranasal (IN) immunization. In contrast, the protective efficacy of RGΔM2-2 delivered by the IM route to African green monkeys was substantially reduced as compared to the efficacy following IN administration, despite comparable levels of serum neutralizing antibodies. This result suggests that mucosal immunity may play an important role in RSV protection. The RGΔM2-2 vaccine also demonstrated different attenuation profiles when tested in cotton rats, non-human primates, and a human airway epithelial (HAE) cell model. The data suggest RGΔM2-2 is less attenuated than a similarly designed vaccine candidate constructed on the A2 genetic background. These findings have important implications with regard to both the design and the preclinical safety testing of live-attenuated vaccines

    Growth of RGΔM2-2 versus parental virus in Vero cells.

    No full text
    <p>(A) Vero cells were infected at an MOI of 0.001 and incubated at 37°C. Samples harvested at the times indicated were titrated on Vero cells.</p

    IM boost immunization in NHP.

    No full text
    <p>(A) AGMs were infected as indicated. (B) Complement-dependent serum neutralizing titers, PRNT60. (C) Protective efficacy as determined from BAL samples after challenge.</p

    IN vs IM immunization in NHP.

    No full text
    <p>(A) AGMs were infected as indicated. (B) Serum F-binding antibodies were determined by ELISA. (C) Complement-dependent serum neutralizing titers were measure by PRNT60. (D) Protective efficacy determined as virus titers from BAL samples after challenge.</p

    Attenuation and immunogenicity in cotton rats.

    No full text
    <p>(A) Groups of cotton rats were infected as indicated. (B) The degree of attenuation as amount of virus present in the lungs of animals in (A) was determined. ****p < 0.0001 by unpaired t test. (C) Immunogenicity study in cotton rats. Animals were immunized as indicated. (D) Serum neutralization titers were determined. (E) Protective efficacy as amount of challenge virus present in the lungs of immunized animals in (C).</p

    CX3CR1 Is Expressed in Differentiated Human Ciliated Airway Cells and Co-Localizes with Respiratory Syncytial Virus on Cilia in a G Protein-Dependent Manner

    No full text
    <div><p>Respiratory syncytial virus (RSV) is the principal cause of bronchiolitis in infants and a significant healthcare problem. The RSV Glycoprotein (G) mediates attachment of the virus to the cell membrane, which facilitates interaction of the RSV Fusion (F) protein with nucleolin, thereby triggering fusion of the viral and cellular membranes. However, a host protein ligand for G has not yet been identified. Here we show that CX3CR1 is expressed in the motile cilia of differentiated human airway epithelial (HAE) cells, and that CX3CR1 co-localizes with RSV particles. Upon infection, the distribution of CX3CR1 in these cells is significantly altered. Complete or partial deletion of RSV G results in viruses binding at least 72-fold less efficiently to cells, and reduces virus replication. Moreover, an antibody targeting an epitope near the G protein’s CX3CR1-binding motif significantly inhibits binding of the virus to airway cells. Given previously published evidence of the interaction of G with CX3CR1 in human lymphocytes, these findings suggest a role for G in the interaction of RSV with ciliated lung cells. This interpretation is consistent with past studies showing a protective benefit in immunizing against G in animal models of RSV infection, and would support targeting the CX3CR1-G protein interaction for prophylaxis or therapy. CX3CR1 expression in lung epithelial cells may also have implications for other respiratory diseases such as asthma.</p></div

    CX3CR1 in differentiated HAE cells interacts with RSV.

    No full text
    <p>Differentiated HAE cells, grown as in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0130517#pone.0130517.g001" target="_blank">Fig 1</a>, were incubated with RSV and imaged by immunofluorescence and confocal microscopy. (<b>a</b>) Binding experiment meant to visualize viral particles in association with HAE cells. Cultures were incubated 2h with RSV, then fixed and processed. RSV virions appear in green, β-tubulin is shown in red, and CX3CR1 is colored purple. Two regions of interest, representative of other RSV-bound cells in this image, are outlined with white squares. The top one is shown in expanded views in <b>b</b>, and the other one is shown in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0130517#pone.0130517.s002" target="_blank">S2 Fig</a>. Red, green, and purple images corresponding to β-tubulin, RSV F, and CX3CR1 respectively, are shown individually. Also shown is a merged image combining the fluorescence channels for RSV F, CX3CR1, and motile cilia. (<b>c</b>) Infected HAE cells were incubated for 3 days after infection and imaged using the same antibodies and fluorophores as in <b>a</b> and <b>b</b> but pseudo-colored differently: β-tubulin is in blue, RSV F in green, and CX3CR1 is shown in red. The two images are xy planes of the same sample separated along the z axis by 3.7 μm. The bottom image shows cilia and apical cell body, including some purple color indicative of colocalized tubulin and CX3CR1 immunofluorescence. The top image crosses the plane of the nuclei, below the cilia. Only infected cells (green) are surrounded by red-colored CX3CR1-positive circular features. Uninfected cells in the same sample and cells from uninfected control samples do not show these CX3CR1-containing structures (see <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0130517#pone.0130517.g001" target="_blank">Fig 1C</a>). (<b>d</b>) Confocal immunofluorescence of HAE cells grown, differentiated, and infected for 3 days with RSV strain MSA1. Both <i>en face</i> (xy) and side (xz and yz) views are shown in this image. The xy plane of the <i>en face</i> view mostly cuts through the cilia of the cells, above the cell body. Nuclei are shown in blue using DAPI and, as in panel <b>a</b>, anti-RSV F protein is in green, anti-β-tubulin in red, and anti-CX3CR1 in purple. As in panel <b>c</b>, large ovoid structures positive for CX3CR1 immunofluorescence are seen in the side views and located near the nuclei of infected cells.</p
    corecore