Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Publication year range
1.
Front Immunol ; 13: 886611, 2022.
Article in English | MEDLINE | ID: mdl-35711419

ABSTRACT

Rhinoviruses (RV) have been shown to inhibit subsequent infection by heterologous respiratory viruses, including influenza viruses and severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2). To better understand the mechanisms whereby RV protects against pulmonary coronavirus infection, we used a native murine virus, mouse hepatitis virus strain 1 (MHV-1), that causes severe disease in the lungs of infected mice. We found that priming of the respiratory tract with RV completely prevented mortality and reduced morbidity of a lethal MHV-1 infection. Replication of MHV-1 was reduced in RV-primed mouse lungs although expression of antiviral type I interferon, IFN-ß, was more robust in mice infected with MHV-1 alone. We further showed that signaling through the type I interferon receptor was required for survival of mice given a non-lethal dose of MHV-1. RV-primed mice had reduced pulmonary inflammation and hemorrhage and influx of leukocytes, especially neutrophils, in the airways upon MHV-1 infection. Although MHV-1 replication was reduced in RV-primed mice, RV did not inhibit MHV-1 replication in coinfected lung epithelial cells in vitro. In summary, RV-mediated priming in the respiratory tract reduces viral replication, inflammation, and tissue damage, and prevents mortality of a pulmonary coronavirus infection in mice. These results contribute to our understanding of how distinct respiratory viruses interact with the host to affect disease pathogenesis, which is a critical step in understanding how respiratory viral coinfections impact human health.


Subject(s)
COVID-19 , Coinfection , Enterovirus Infections , Murine hepatitis virus , Pneumonia , Animals , Lung , Mice , Rhinovirus , SARS-CoV-2
2.
mSphere ; 6(3): e0047921, 2021 06 30.
Article in English | MEDLINE | ID: mdl-34160242

ABSTRACT

Coinfection by heterologous viruses in the respiratory tract is common and can alter disease severity compared to infection by individual virus strains. We previously found that inoculation of mice with rhinovirus (RV) 2 days before inoculation with a lethal dose of influenza A virus [A/Puerto Rico/8/34 (H1N1) (PR8)] provides complete protection against mortality. Here, we extended that finding to a second lethal respiratory virus, pneumonia virus of mice (PVM), and analyzed potential mechanisms of RV-induced protection. RV completely prevented mortality and weight loss associated with PVM infection. Major changes in host gene expression upon PVM infection were delayed compared to PR8. RV induced earlier recruitment of inflammatory cells, which were reduced at later times in RV-inoculated mice. Findings common to both virus pairs included the upregulated expression of mucin-associated genes and dampening of inflammation-related genes in mice that were inoculated with RV before lethal virus infection. However, type I interferon (IFN) signaling was required for RV-mediated protection against PR8 but not PVM. IFN signaling had minor effects on PR8 replication and contributed to controlling neutrophilic inflammation and hemorrhagic lung pathology in RV/PR8-infected mice. These findings, combined with differences in virus replication levels and disease severity, suggest that the suppression of inflammation in RV/PVM-infected mice may be due to early, IFN-independent suppression of viral replication, while that in RV/PR8-infected mice may be due to IFN-dependent modulation of immune responses. Thus, a mild upper respiratory viral infection can reduce the severity of a subsequent severe viral infection in the lungs through virus-dependent mechanisms. IMPORTANCE Respiratory viruses from diverse families cocirculate in human populations and are frequently detected within the same host. Although clinical studies suggest that infection by multiple different respiratory viruses may alter disease severity, animal models in which we can control the doses, timing, and strains of coinfecting viruses are critical to understanding how coinfection affects disease severity. Here, we compared gene expression and immune cell recruitment between two pairs of viruses (RV/PR8 and RV/PVM) inoculated sequentially in mice, both of which result in reduced severity compared to lethal infection by PR8 or PVM alone. Reduced disease severity was associated with suppression of inflammatory responses in the lungs. However, differences in disease kinetics and host and viral gene expression suggest that protection by coinfection with RV may be due to distinct molecular mechanisms. Indeed, we found that antiviral cytokine signaling was required for RV-mediated protection against lethal infection by PR8 but not PVM.


Subject(s)
Coinfection/immunology , Host-Pathogen Interactions , Interferon Type I/immunology , Picornaviridae Infections/immunology , Rhinovirus/immunology , Rhinovirus/pathogenicity , Animals , Coinfection/virology , Female , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Influenza A virus/immunology , Influenza A virus/pathogenicity , Lung/immunology , Lung/pathology , Lung/virology , Mice , Mice, Inbred BALB C , Murine pneumonia virus/immunology , Murine pneumonia virus/pathogenicity , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Pneumovirus Infections/immunology , Pneumovirus Infections/prevention & control , Severity of Illness Index , Transcriptome , Virus Replication
3.
J Virol ; 92(23)2018 12 01.
Article in English | MEDLINE | ID: mdl-30232180

ABSTRACT

Influenza viruses and rhinoviruses are responsible for a large number of acute respiratory viral infections in human populations and are detected as copathogens within hosts. Clinical and epidemiological studies suggest that coinfection by rhinovirus and influenza virus may reduce disease severity and that they may also interfere with each other's spread within a host population. To determine how coinfection by these two unrelated respiratory viruses affects pathogenesis, we established a mouse model using a minor serogroup rhinovirus (rhinovirus strain 1B [RV1B]) and mouse-adapted influenza A virus (A/Puerto Rico/8/1934 [PR8]). Infection of mice with RV1B 2 days before PR8 reduced the severity of infection by a low or medium, but not high, dose of PR8. Disease attenuation was associated with an early inflammatory response in the lungs and enhanced clearance of PR8. However, coinfection by RV1B did not reduce PR8 viral loads early in infection or inhibit replication of PR8 within respiratory epithelia or in vitro Inflammation in coinfected mice remained focal compared to diffuse inflammation and damage in the lungs of mice infected by PR8. The timing of RV1B coinfection was a critical determinant of protection, suggesting that sufficient time is needed to induce this response. Finally, disease attenuation was not unique to RV1B: dose-dependent coinfection by a murine coronavirus (mouse hepatitis virus strain 1 [MHV-1]) also reduced the severity of PR8 infection. Unlike RV1B, coinfection with MHV-1 reduced early PR8 replication, which was associated with upregulation of beta interferon (IFN-ß) expression. This model is critical for understanding the mechanisms responsible for influenza disease attenuation during coinfection by unrelated respiratory viruses.IMPORTANCE Viral infections in the respiratory tract can cause severe disease and are responsible for a majority of pediatric hospitalizations. Molecular diagnostics have revealed that approximately 20% of these patients are infected by more than one unrelated viral pathogen. To understand how viral coinfection affects disease severity, we inoculated mice with a mild viral pathogen (rhinovirus or murine coronavirus), followed 2 days later by a virulent viral pathogen (influenza A virus). This model demonstrated that rhinovirus can reduce the severity of influenza A virus, which corresponded with an early but controlled inflammatory response in the lungs and early clearance of influenza A virus. We further determined the dose and timing parameters that were important for effective disease attenuation and showed that influenza disease is also reduced by coinfection with a murine coronavirus. These findings demonstrate that coinfecting viruses can alter immune responses and pathogenesis in the respiratory tract.


Subject(s)
Coinfection/epidemiology , Disease Models, Animal , Influenza A virus/immunology , Orthomyxoviridae Infections/prevention & control , Picornaviridae Infections/virology , Pneumonia/prevention & control , Rhinovirus/immunology , Animals , Coinfection/virology , Female , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Picornaviridae Infections/complications , Picornaviridae Infections/epidemiology , Pneumonia/immunology , Pneumonia/virology , Severity of Illness Index , Time Factors , Virus Internalization , Virus Replication
4.
PLoS One ; 12(6): e0178408, 2017.
Article in English | MEDLINE | ID: mdl-28575086

ABSTRACT

The severity of respiratory viral infections is partially determined by the cellular response mounted by infected lung epithelial cells. Disease prevention and treatment is dependent on our understanding of the shared and unique responses elicited by diverse viruses, yet few studies compare host responses to viruses from different families while controlling other experimental parameters. Murine models are commonly used to study the pathogenesis of respiratory viral infections, and in vitro studies using murine cells provide mechanistic insight into the pathogenesis observed in vivo. We used microarray analysis to compare changes in gene expression of murine lung epithelial cells infected individually by three respiratory viruses causing mild (rhinovirus, RV1B), moderate (coronavirus, MHV-1), and severe (influenza A virus, PR8) disease in mice. RV1B infection caused numerous gene expression changes, but the differential effect peaked at 12 hours post-infection. PR8 altered an intermediate number of genes whose expression continued to change through 24 hours. MHV-1 had comparatively few effects on host gene expression. The viruses elicited highly overlapping responses in antiviral genes, though MHV-1 induced a lower type I interferon response than the other two viruses. Signature genes were identified for each virus and included host defense genes for PR8, tissue remodeling genes for RV1B, and transcription factors for MHV-1. Our comparative approach identified universal and specific transcriptional signatures of virus infection that can be used to distinguish shared and virus-specific mechanisms of pathogenesis in the respiratory tract.


Subject(s)
Coronavirus/pathogenicity , Gene Expression , Influenza A virus/pathogenicity , Lung/cytology , Rhinovirus/pathogenicity , Animals , Epithelial Cells/metabolism , Lung/metabolism , Mice
5.
J Pharmacol Exp Ther ; 314(3): 1079-86, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15976018

ABSTRACT

Glucans are immunomodulatory carbohydrates found in the cell walls of fungi and certain bacteria. We examined the pharmacokinetics of three water-soluble glucans (glucan phosphate, laminarin, and scleroglucan) after oral administration of 1 mg/kg doses in rats. Maximum plasma concentrations for glucan phosphate occurred at 4 h. In contrast, laminarin and scleroglucan showed two plasma peaks between 0.5 and 12 h. At 24 h, 27 +/- 3% of the glucan phosphate and 20 +/- 7% of the laminarin remained in the serum. Scleroglucan was rapidly absorbed and eliminated. The liver did not significantly contribute to the clearance of plasma glucan. Biological effects were further studied in mice. Following oral administration of 1 mg, glucans were bound and internalized by intestinal epithelial cells and gut-associated lymphoid tissue (GALT) cells. Internalization of glucan by intestinal epithelial cells was not Dectin-dependent. GALT expression of Dectin-1 and toll-like receptor (TLR) 2, but not TLR4, increased following oral administration of glucan. Oral glucan increased systemic levels of interleukin (IL)-12 (151 +/- 15%) in mice. Oral glucan administration also increased survival in mice challenged with Staphylococcus aureus or Candida albicans. These data demonstrate that orally administered water-soluble glucans translocate from the gastrointestinal (GI) tract into the systemic circulation. The glucans are bound by GI epithelial and GALT cells, and they modulate the expression of pattern recognition receptors in the GALT, increase IL-12 expression, and induce protection against infectious challenge.


Subject(s)
Glucans/pharmacology , Immunity, Innate/drug effects , Intestinal Absorption , Administration, Oral , Animals , Biological Availability , Candidiasis/immunology , Cytokines/biosynthesis , Glucans/administration & dosage , Glucans/pharmacokinetics , Lectins, C-Type , Male , Membrane Proteins/analysis , Membrane Proteins/physiology , Mice , Mice, Inbred ICR , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/physiology , Peyer's Patches/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Immunologic/analysis , Staphylococcal Infections/immunology , Toll-Like Receptor 2
6.
Actas Urol Esp ; 11(5): 481-4, 1987.
Article in Spanish | MEDLINE | ID: mdl-3453590

ABSTRACT

PIP: The use of vasectomy has increased considerably over the past few years in the province of Guadalajara, Spain. This study reviews the records of 140 men who underwent vasectomy between March 1985-March 1986. The protocol included a personal interview and a questionnaire on attitudes toward contraception, sex, marriage, sterilization, and likelihood of desiring more children in the future. Candidates for vasectomy were required to have 2 children. The operations were performed under local anesthesia on an outpatient basis. Semen analyses were done at 4 weeks after a minimum of 10 ejaculations and at 12 weeks. A new questionnaire 12 months after the operation sought information on postoperative pain, changes in libido, and satisfaction of the couple with the operation. The average of the men was 37 years and the average duration of union was 11 years. Couples had 3 children on average and 85% had primary educations. At the time of the initial consultation, 44% were using condoms, 17% pills, 10% IUDs, and 20% withdrawal. 63% had undesired children, mainly due to failure of withdrawal. No sperm were found 4 or 12 weeks in any of the cases, and no pregnancies were reported. Infections developed at the site of the incision in 10 cases and epididymitis occurred 25 cases.^ieng


Subject(s)
Vasectomy/statistics & numerical data , Adult , Humans , Male , Spain
SELECTION OF CITATIONS
SEARCH DETAIL
...