• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 1
  • 1
  • Tagged with
  • 7
  • 7
  • 7
  • 7
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

Antiviral function of LL-37 on respiratory syncytial virus

Currie, Silke Maria January 2016 (has links)
Recurrent infection with human respiratory syncytial virus (RSV) is one of the most common causes for lower respiratory tract illness (LRI) in infants, the elderly, and immunocompromised individuals. Due to lack of vaccines and therapeutic interventions, medical care of acute RSV bronchiolitis is mostly limited to supportive measures. Thus, novel treatment options to control RSV infection are desperately required. The cationic host defence peptide human cathelicidin LL-37 possesses both microbicidal and immunomodulatory properties. This essential effector of the innate immune system holds potent antiviral activity against a variety of viruses, including influenza virus, and has been proposed as a promising candidate for antiviral drug development. Previous studies revealed that lower cathelicidin levels put RSV infected infants at risk for more severe RSV disease, while infection of lung epithelial cells induced cathelicidin up-regulation. These findings suggest that LL-37 might possess antiviral activity against RSV. However, its potential antiviral function on RSV remains to be elucidated. This thesis therefore aimed to evaluate the antiviral activity of cathelicidins against RSV, by assessing its relevance in vitro and in vivo and elucidating the underlying antiviral mechanism. Firstly, the antiviral effects of human cathelicidin LL-37 against RSV were addressed in vitro. Presence of LL-37 during infection potently reduced viral titres and protected cells against virus-associated cytopathic effects. Experiments revealed that only the core region of LL-37 holds antiviral activity against RSV. Antiviral effects were also observed for the murine LL-37 orthologue mCRAMP. Administration of LL-37 at different stages in the infection cycle provided evidence that LL-37 can be used preventatively, protecting against RSV infection by directly acting on both cells and viral particles. When given therapeutically, once an infection was established, LL-37 also limited viral spread. Next, the molecular mechanism mediating the peptide’s antiviral activity was investigated. It was demonstrated that LL-37 does not affect the interferon-mediated cellular antiviral immune response to RSV. Experiments established that LL-37 does not contribute to viral clearance by inducing epithelial cell death. Further mechanistic studies revealed that the peptide directly binds to RSV particles, destabilises the integrity of the viral envelope, and prevents adsorption of RSV to epithelial cells during the entry stage of infection. Finally, the in vivo relevance of LL-37 treatment and endogenous cathelicidin expression was examined, employing both murine and human model systems. It was established that LL-37 has protective antiviral effects against RSV in vivo. In contrast to the cell culture model, only co-administration of LL-37 and RSV, but not treatment prior or post infection, protects mice from clinical signs of infection. Levels of the murine LL-37 orthologue mCRAMP were increased in RSV infected lungs, pointing towards its importance in antiviral defence. In keeping with this, mCRAMP-deficient mice were more susceptible to RSV induced disease. Equally, individuals with low nasal LL-37 baseline levels that were experimentally challenged with RSV, were more susceptible to infection. This highlights the importance of endogenous cathelicidin expression to fight and control RSV infection. Overall, these results identify LL-37 as an important antiviral agent against RSV in vitro and in vivo, and emphasise the role of endogenous cathelicidins in the defence against this pathogen. Moreover, unravelling the underlying antiviral mechanism of LL-37 against RSV adds to our understanding of how CHDP act on enveloped viruses, thus supporting the development of new antiviral treatment options.
2

osteopontin plays a pivotal role in in increasing severity of respiratory syncytial virus infection

Sampayo-Escobar, Viviana 07 July 2017 (has links)
The molecular mechanisms underlying susceptibility to severe respiratory syncytial virus (RSV) infection remain poorly understood. Herein, we report on the role of osteopontin (OPN) in regulation of RSV infection in human epithelial cells and how interleukin-1 beta (IL-1β), a cytokine secreted soon after RSV infection, when persistently expressed can induce OPN expression leading to increased viral infection. We first compared OPN expression in two human epithelial cell lines: HEK-293 and HEp-2. In contrast to HEp-2, HEK-293 expresses low levels of pro-caspase-1 resulting in decreased IL-1β expression in response to RSV infection. We found a correlation between low IL-1β levels and a delay in induction of OPN expression in RSV-infected HEK-293 cells compared to HEp-2. This phenomenon could partially explain the high susceptibility of HEp-2 cells to RSV infection versus the moderate susceptibility of HEK-293 cells. Also, HEK-293 cells expressing low levels of pro-caspase-1 exhibit decreased IL-1β expression and delayed OPN expression in response to RSV infection. HEK-293 cells incubated with human rIL-1β showed a dose-dependent increase in OPN expression upon RSV infection. Also, incubation with rOPN increased RSV viral load. Moreover, HEp-2 cells or mice infected with a mucogenic RSV strain RSV-L19F showed elevated levels of OPN in contrast to mice infected with the laboratory RSV strain rA2. This correlated with elevated levels of OPN following infection with RSV-L19F compared to rA2. Together, these results demonstrate that increased OPN expression is regulated in part by IL-1β, and the interplay between IL-1β and OPN signaling has a pivotal role in the spread of RSV infection.
3

Untersuchung von rekombinantem Vacciniavirus MVA zur Entwicklung von Impfstoffen gegen Infektionen mit Respiratorischen Synzytialviren / Evaluation and construction of recombinant modified vaccinia virus Ankara as candidate vector vaccine against infections with respiratory syncytial viruses

Süzer, Yasemin 08 January 2008 (has links) (PDF)
In dieser Arbeit wurden Vektorimpfstoffe auf der Basis rekombinanter Vacciniaviren hinsichtlich ihrer Eignung zur Immunisierung gegen Infektionen mit Respiratorischen Synzytialviren (RSV) untersucht. Hierfür standen genetisches Material und Viruspräparationen des Respiratorischen Synzytialvirus des Rindes (BRSV, Stamm Odijk) sowie des Respiratorischen Synzytialvirus des Menschen (HRSV, Subtyp A2) sowie rekombinante Vacciniaviren MVA-HRSV-F bzw. MVA-HRSV-G zur Verfügung. Rekombinante MVA-Viren, welche die Gene der BRSV-Oberflächenproteine G und F (MVA-BRSV-F, MVA-BRSV-G, MVA-BRSV-Gneu), sowie Viren in welchen die Fremdgensequenzen durch Deletion wieder entfernt sind (Revertante Viren MVA-∆BRSV-F und MVA-∆BRSV-G), wurden gentechnologisch hergestellt. Alle rekombinanten MVA-Viren wurden molekular-virologisch charakterisiert und dienten zur Gewinnung und Prüfung von Testimpfstoffen im Tiermodell. Die Untersuchungen zeigen: 1. Alle neu konstruierten rekombinanten MVA-BRSV-Viren produzierten nach Infektion von Zellkulturen die erwünschten Zielantigene, die BRSV-Glykoproteine F und G. Für das durch MVA-Expression hergestellte BRSV-F-Glykoprotein konnte außerdem die biologische Funktionalität in einem Fusionstest in infizierten HeLa-Zellen nachgewiesen werden. 2. Die Charakterisierung der Genome aller MVA-BRSV- sowie MVA-HRSV-Vektorviren bestätigte die exakte Insertion der Fremdgensequenzen im anvisierten Genombereich und zeigte die genetische Stabilität der Virusisolate nach Passagierung. 3. Bei der Untersuchung des Wachstumsverhaltens von MVA-BRSV-F und MVA-BRSV-G zeigte sich die eingeschränkte Vermehrungsfähigkeit des Virus MVA-BRSV-G. Die Konstruktion und Untersuchung der revertanten Viren MVA-∆BRSV-F und MVA-∆BRSV-G belegte die Koproduktion des G-Proteins als Ursache des verminderten Replikationsvermögens. Dieser für ein mögliches Impfvirus erhebliche Nachteil konnte durch die Verwendung eines moderateren Vacciniavirus-Promotors zur Fremdgenexpression (rekombinantes Virus MVA-BRSV-Gneu) behoben werden. 4. Die Prüfung von Testimpfstoffen auf der Grundlage der rekombinanten MVA-HRSV-Viren in einem Maus-HRSV-Infektionsmodell zeigte, dass MVA-HRSV-Impfstoffe, im Gegensatz zu Impfstoffen aus mit Formalin-inaktiviertem HRSV, Immunantworten mit einem ausgewogenen TH1/TH2-assoziierten Zytokinprofil induzierten. Eine infolge von Immunisierung verstärkte Einwanderung eosinophiler Zellen (Marker für Immunpathogenese) in die Lungen HRSV-infizierter Tiere, konnte nach MVA-Impfung nicht beziehungsweise in nur sehr geringem Ausmaß festgestellt werden (OLSZEWSKA et al. 2004). 5. Wichtige erste Daten hinsichtlich der Verträglichkeit, Immunogenität und Schutzwirkung rekombinanter Impfstoffe auf der Basis von MVA-BRSV-F und MVA-BRSV-G konnten in einem Kälber BRSV-Infektionsmodell erhoben werden. Die zweimalige Immunisierung mit MVA-Impfstoff verlief bei allen Tieren ohne feststellbare Nebenwirkungen und die Anregung Vaccinia- bzw. BRSV-F-spezifischer Antikörper bestätigte die Immunogenität der Vektorvakzinen. Schließlich belegten klinische Daten, insbesondere die fehlende Fieberreaktion bei Impflingen nach BRSV-Belastungsinfektion, die Schutzwirkung der MVA-BRSV-Impfstoffe. Insgesamt unterstützen die erzielten Ergebnisse dieser Arbeiten die weitere präklinische und klinische Untersuchung von MVA-Vektorimpfstoffen zur wirksameren und sichereren Bekämpfung von Infektionen mit Respiratorischen Synzytialviren. / This study investigated vector vaccines based on recombinant vaccinia virus MVA for their suitability to immunize against infections with respiratory syncytial viruses. Genetic material and virus stocks of bovine respiratory syncytial virus (BRSV, Strain Odijk) and human respiratory syncytial virus (HRSV, Strain A2) and recombinant vaccinia viruses MVA-HRSV-F and MVA-HRSV-G were provided and used in this study. The project work included the genetical engineering of recombinant MVA expressing gene sequences encoding the BRSV surface proteins G and F (MVA-BRSV-F, MVA-BRSV-G, MVA-BRSV-Gneu) and the secondary generation of mutant viruses in which recombinant gene sequences have been removed (revertant viruses MVA-∆BRSV-F, MVA-∆BRSV-G). All recombinant MVA were carefully characterized in in vitro experiments and served for generation of vaccine preparations being tested in animal model systems. The investigations demonstrate: 1. All recombinant MVA-BRSV viruses produced the target antigens (BRSV-F and -G proteins) upon tissue culture infections. Functional activity of BRSV-F protein was demonstrated in a cell fusion assay using virus-infected HeLa cells. 2. The characterization of the genomes of all MVA recombinant viruses confirmed the correct insertion of foreign gene sequences into the target site of the MVA genome and demonstrated the genetic stability of the vector viruses upon tissue culture passage. 3. In vitro studies on virus growth revealed a reduced replicative capacity of the recombinant virus MVA-BRSV-G. Construction and growth analysis of revertant viruses MVA-∆BRSV-F and MVA-∆BRSV-G demonstrated that over expression of BRSV-G protein caused this replication deficiency which could be avoided by using a more moderate vaccinia virus promoter for transcriptional control of recombinant gene expression (recombinant virus MVA-BRSV-Gneu). 4. Upon characterization in a mouse-HRSV challenge model candidate vaccines based on recombinant MVA-HRSV viruses, in contrast to formalin inactivated HRSV, and induced a well balanced TH1 and TH2 cytokine profile. In addition, none of the MVA-HRSV-F vaccinated animals and only two of the MVA-HRSV-G immunized mice showed low-level eosinophilia in the lungs after HRSV challenge infection (OLSZEWSKA et al. 2004). 5. Vaccination experiments in the calf-BRSV challenge model generated first relevant data on safety, immunogenicity and protective capacity of MVA-BRSV recombinant vaccines. The repeated application of MVA vaccine was well tolerated by all vaccinated animals and the induction of vaccinia- and BRSV-F-specific antibody responses confirmed the immunogenicity of the MVA vector vaccines. Moreover, clinical data (lack of fever response in vaccines) suggested the protective capacity of MVA-BRSV immunization upon BRSV challenge. The obtained results from these studies clearly support further preclinical and clinical evaluation of recombinant MVA candidate vaccines to immunize against disease caused by RSV infections in cattle and humans.
4

Untersuchung von rekombinantem Vacciniavirus MVA zur Entwicklung von Impfstoffen gegen Infektionen mit Respiratorischen Synzytialviren

Süzer, Yasemin 12 June 2007 (has links)
In dieser Arbeit wurden Vektorimpfstoffe auf der Basis rekombinanter Vacciniaviren hinsichtlich ihrer Eignung zur Immunisierung gegen Infektionen mit Respiratorischen Synzytialviren (RSV) untersucht. Hierfür standen genetisches Material und Viruspräparationen des Respiratorischen Synzytialvirus des Rindes (BRSV, Stamm Odijk) sowie des Respiratorischen Synzytialvirus des Menschen (HRSV, Subtyp A2) sowie rekombinante Vacciniaviren MVA-HRSV-F bzw. MVA-HRSV-G zur Verfügung. Rekombinante MVA-Viren, welche die Gene der BRSV-Oberflächenproteine G und F (MVA-BRSV-F, MVA-BRSV-G, MVA-BRSV-Gneu), sowie Viren in welchen die Fremdgensequenzen durch Deletion wieder entfernt sind (Revertante Viren MVA-∆BRSV-F und MVA-∆BRSV-G), wurden gentechnologisch hergestellt. Alle rekombinanten MVA-Viren wurden molekular-virologisch charakterisiert und dienten zur Gewinnung und Prüfung von Testimpfstoffen im Tiermodell. Die Untersuchungen zeigen: 1. Alle neu konstruierten rekombinanten MVA-BRSV-Viren produzierten nach Infektion von Zellkulturen die erwünschten Zielantigene, die BRSV-Glykoproteine F und G. Für das durch MVA-Expression hergestellte BRSV-F-Glykoprotein konnte außerdem die biologische Funktionalität in einem Fusionstest in infizierten HeLa-Zellen nachgewiesen werden. 2. Die Charakterisierung der Genome aller MVA-BRSV- sowie MVA-HRSV-Vektorviren bestätigte die exakte Insertion der Fremdgensequenzen im anvisierten Genombereich und zeigte die genetische Stabilität der Virusisolate nach Passagierung. 3. Bei der Untersuchung des Wachstumsverhaltens von MVA-BRSV-F und MVA-BRSV-G zeigte sich die eingeschränkte Vermehrungsfähigkeit des Virus MVA-BRSV-G. Die Konstruktion und Untersuchung der revertanten Viren MVA-∆BRSV-F und MVA-∆BRSV-G belegte die Koproduktion des G-Proteins als Ursache des verminderten Replikationsvermögens. Dieser für ein mögliches Impfvirus erhebliche Nachteil konnte durch die Verwendung eines moderateren Vacciniavirus-Promotors zur Fremdgenexpression (rekombinantes Virus MVA-BRSV-Gneu) behoben werden. 4. Die Prüfung von Testimpfstoffen auf der Grundlage der rekombinanten MVA-HRSV-Viren in einem Maus-HRSV-Infektionsmodell zeigte, dass MVA-HRSV-Impfstoffe, im Gegensatz zu Impfstoffen aus mit Formalin-inaktiviertem HRSV, Immunantworten mit einem ausgewogenen TH1/TH2-assoziierten Zytokinprofil induzierten. Eine infolge von Immunisierung verstärkte Einwanderung eosinophiler Zellen (Marker für Immunpathogenese) in die Lungen HRSV-infizierter Tiere, konnte nach MVA-Impfung nicht beziehungsweise in nur sehr geringem Ausmaß festgestellt werden (OLSZEWSKA et al. 2004). 5. Wichtige erste Daten hinsichtlich der Verträglichkeit, Immunogenität und Schutzwirkung rekombinanter Impfstoffe auf der Basis von MVA-BRSV-F und MVA-BRSV-G konnten in einem Kälber BRSV-Infektionsmodell erhoben werden. Die zweimalige Immunisierung mit MVA-Impfstoff verlief bei allen Tieren ohne feststellbare Nebenwirkungen und die Anregung Vaccinia- bzw. BRSV-F-spezifischer Antikörper bestätigte die Immunogenität der Vektorvakzinen. Schließlich belegten klinische Daten, insbesondere die fehlende Fieberreaktion bei Impflingen nach BRSV-Belastungsinfektion, die Schutzwirkung der MVA-BRSV-Impfstoffe. Insgesamt unterstützen die erzielten Ergebnisse dieser Arbeiten die weitere präklinische und klinische Untersuchung von MVA-Vektorimpfstoffen zur wirksameren und sichereren Bekämpfung von Infektionen mit Respiratorischen Synzytialviren. / This study investigated vector vaccines based on recombinant vaccinia virus MVA for their suitability to immunize against infections with respiratory syncytial viruses. Genetic material and virus stocks of bovine respiratory syncytial virus (BRSV, Strain Odijk) and human respiratory syncytial virus (HRSV, Strain A2) and recombinant vaccinia viruses MVA-HRSV-F and MVA-HRSV-G were provided and used in this study. The project work included the genetical engineering of recombinant MVA expressing gene sequences encoding the BRSV surface proteins G and F (MVA-BRSV-F, MVA-BRSV-G, MVA-BRSV-Gneu) and the secondary generation of mutant viruses in which recombinant gene sequences have been removed (revertant viruses MVA-∆BRSV-F, MVA-∆BRSV-G). All recombinant MVA were carefully characterized in in vitro experiments and served for generation of vaccine preparations being tested in animal model systems. The investigations demonstrate: 1. All recombinant MVA-BRSV viruses produced the target antigens (BRSV-F and -G proteins) upon tissue culture infections. Functional activity of BRSV-F protein was demonstrated in a cell fusion assay using virus-infected HeLa cells. 2. The characterization of the genomes of all MVA recombinant viruses confirmed the correct insertion of foreign gene sequences into the target site of the MVA genome and demonstrated the genetic stability of the vector viruses upon tissue culture passage. 3. In vitro studies on virus growth revealed a reduced replicative capacity of the recombinant virus MVA-BRSV-G. Construction and growth analysis of revertant viruses MVA-∆BRSV-F and MVA-∆BRSV-G demonstrated that over expression of BRSV-G protein caused this replication deficiency which could be avoided by using a more moderate vaccinia virus promoter for transcriptional control of recombinant gene expression (recombinant virus MVA-BRSV-Gneu). 4. Upon characterization in a mouse-HRSV challenge model candidate vaccines based on recombinant MVA-HRSV viruses, in contrast to formalin inactivated HRSV, and induced a well balanced TH1 and TH2 cytokine profile. In addition, none of the MVA-HRSV-F vaccinated animals and only two of the MVA-HRSV-G immunized mice showed low-level eosinophilia in the lungs after HRSV challenge infection (OLSZEWSKA et al. 2004). 5. Vaccination experiments in the calf-BRSV challenge model generated first relevant data on safety, immunogenicity and protective capacity of MVA-BRSV recombinant vaccines. The repeated application of MVA vaccine was well tolerated by all vaccinated animals and the induction of vaccinia- and BRSV-F-specific antibody responses confirmed the immunogenicity of the MVA vector vaccines. Moreover, clinical data (lack of fever response in vaccines) suggested the protective capacity of MVA-BRSV immunization upon BRSV challenge. The obtained results from these studies clearly support further preclinical and clinical evaluation of recombinant MVA candidate vaccines to immunize against disease caused by RSV infections in cattle and humans.
5

Development of a novel cell-based screening platform to identify inhibitors of viral interferon antagonists from clinically important viruses

Vasou, Andri January 2016 (has links)
All viruses encode for at least one viral interferon (IFN) antagonist, which is used to subvert the cellular IFN response, a powerful antiviral innate immune response. Numerous in vitro and in vivo studies have demonstrated that IFN antagonism is crucial for virus survival, suggesting that viral IFN antagonists could represent promising therapeutic targets. This study focuses on Respiratory Syncytial Virus (RSV), an important human pathogen for which there is no vaccine or virus-specific antiviral drug. RSV encodes two IFN antagonists NS1 and NS2, which play a critical role in RSV replication and pathogenicity. We developed a high-throughput screening (HTS) assay to target NS2 via our A549.pr(ISRE)GFP-RSV/NS2 cell-line, which contains a GFP gene under the control of an IFN-stimulated response element (ISRE) to monitor IFN- signalling pathway. NS2 inhibits the IFN-signalling pathway and hence GFP expression in the A549.pr(ISRE)GFP-RSV/NS2 cell-line by mediating STAT2 degradation. Using a HTS approach, we screened 16,000 compounds to identify small molecules that inhibit NS2 function and therefore relinquish the NS2 imposed block to IFN-signalling, leading to restoration of GFP expression. A total of twenty-eight hits were identified; elimination of false positives left eight hits, four of which (AV-14, -16, -18, -19) are the most promising. These four hit compounds have EC₅₀ values in the single μM range and three of them (AV-14, -16, -18) represent a chemically related series with an indole structure. We demonstrated that the hit compounds specifically inhibit the STAT2 degradation function of NS2, not the function of NS1 or unrelated viral IFN antagonists. At the current time, compounds do not restrict RSV replication in vitro, hence hit optimization is required to improve their potency. Nonetheless, these compounds could be used as chemical tools to determine the unknown mechanism by which NS2 mediates STAT2 degradation and tackle fundamental questions about RSV biology.
6

Influenza A viruses dual and multiple infections with other respiratory viruses and risk of hospitalization and mortality

Goka, Edward Anthony Chilongo January 2014 (has links)
Introduction: Epidemiological studies have indicated that 5-38% of influenza like illnesses (ILI) develop into severe disease due to, among others, factors such as; underlying chronic diseases, age, pregnancy, and viral mutations. There are suggestions that dual or multiple virus infections may affect disease severity. This study investigated the association between co-infection between influenza A viruses and other respiratory viruses and disease severity. Methodology: Datum for samples from North West England tested between January 2007 and June 2012 was analysed for patterns of co-infection between influenza A viruses and ten respiratory viruses. Risk of hospitalization to a general ward ICU or death in single versus mixed infections was assessed using multiple logistic regression models. Results: One or more viruses were identified in 37.8% (11,715/30,975) of samples, of which 10.4% (1,214) were mixed infections and 89.6% (10,501) were single infections. Among patients with influenza A(H1N1)pdm09, co-infections occurred in 4.7% (137⁄2,879) vs. 6.5% (59⁄902) in those with seasonal influenza A virus infection. In general, patients with mixed respiratory virus infections had a higher risk of admission to a general ward (OR: 1.43, 95% CI: 1.2 – 1.7, p = <0.0001) than those with a single infection. Co-infection between seasonal influenza A viruses and influenza B virus was associated with a significant increase in the risk of admission to ICU/ death (OR: 22.0, 95% CI: 2.21 – 219.8 p = 0.008). RSV/seasonal influenza A viruses co-infection also associated with increased risk but this was not statistically significant. For the pandemic influenza A(H1N1)pdm09 virus, RSV and AdV co-infection increased risk of hospitalization to a general ward, whereas Flu B increased risk of admission to ICU/ death, but none of these were statistically significant. Considering only single infections, RSV and hPIV1-3 increased risk of admission to a general ward (OR: 1.49, 95% CI: 1.28 – 1.73, p = <0.0001 and OR: 1.34, 95% CI: 1.003 – 1.8, p = 0.05) and admission to ICU/ death (OR: 1.5, 95% CI: 1.20 – 2.0, p = <0.0001 and OR: 1.60, 95% CI: 1.02 – 2.40, p = 0.04). Conclusion: Co-infection is a significant predictor of disease outcome; there is insufficient public health data on this subject as not all samples sent for investigation of respiratory virus infection are tested for all respiratory viruses. Integration of testing for respiratory viruses’ co-infections into routine clinical practice and R&D on integrated drugs and vaccines for influenza A&B, RSV, and AdV, and development of multi-target diagnostic tests is encouraged.
7

CLINICAL SEVERITY OF RHINOVIRUS/ENTEROVIRUS COMPARED TO OTHER RESPIRATORY VIRUSES IN CHILDREN

Asner, Andrea Sandra 10 1900 (has links)
<p><strong>Background</strong>: Human rhinovirus/enterovirus (HRV/ENT) infections are commonly identified in children with acute respiratory infections (ARIs), but data on their clinical severity remains limited. We compared the clinical severity of HRV/ENT to respiratory syncytial virus (RSV), influenza A/B (FLU) and other common respiratory virus in children.</p> <p><strong>Methods</strong>: Retrospective study of children with ARIs and confirmed single positive viral infections on mid-turbinate swabs by molecular assays. Outcome measures included hospital admission and, for inpatients, a composite end-point consisting of intensive care admission, hospitalization greater than 5 days, oxygen requirements or death.</p> <p><strong>Results</strong>: A total of 116 HRV/ENT, 102 RSV, 99 FLU and 64 other common respiratory viruses were identified. Children with single HRV/ENT infections presented with significantly higher rates of underlying immunosuppressive conditions compared to those with RSV (37.9% vs 13.6%; p</p> <p><strong>Conclusions</strong>: Children with HRV/ENT had a more severe clinical course than those with RSV and FLUA/B infections and often had significant comorbidities. These findings emphasize the importance of considering HRV/ENT infection in children presenting with severe acute respiratory tract infections.</p> / Master of Science (MSc)

Page generated in 0.1113 seconds