Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Emerg Infect Dis ; 28(8): 1551-1558, 2022 08.
Article in English | MEDLINE | ID: mdl-35705189

ABSTRACT

A COVID-19 outbreak occurred among Cameron Peak Fire responders in Colorado, USA, during August 2020-January 2021. The Cameron Peak Fire was the largest recorded wildfire in Colorado history, lasting August-December 2020. At least 6,123 responders were involved, including 1,260 firefighters in 63 crews who mobilized to the fire camps. A total of 79 COVID-19 cases were identified among responders, and 273 close contacts were quarantined. State and local public health investigated the outbreak and coordinated with wildfire management teams to prevent disease spread. We performed whole-genome sequencing and applied social network analysis to visualize clusters and transmission dynamics. Phylogenetic analysis identified 8 lineages among sequenced specimens, implying multiple introductions. Social network analysis identified spread between and within crews. Strategies such as implementing symptom screening and testing of arriving responders, educating responders about overlapping symptoms of smoke inhalation and COVID-19, improving physical distancing of crews, and encouraging vaccinations are recommended.


Subject(s)
COVID-19 , Firefighters , Wildfires , COVID-19/epidemiology , Colorado/epidemiology , Disease Outbreaks , Humans , Phylogeny
2.
Clin Infect Dis ; 73(Suppl 1): S65-S73, 2021 07 15.
Article in English | MEDLINE | ID: mdl-33912930

ABSTRACT

BACKGROUND: Nasopharyngeal specimens (NPS) are commonly used for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) testing but can be uncomfortable for patients. Self-collected saliva specimens (SS) or anterior nasal specimens (ANS) for SARS-CoV-2 detection are less invasive, but the sensitivity of these specimen types has not been thoroughly evaluated. METHODS: During September-November 2020, 730 adults undergoing SARS-CoV-2 testing at community testing events and homeless shelters in Denver provided self-collected SS and ANS before NPS collection and answered a short survey about symptoms and specimen preference. Specimens were tested for SARS-CoV-2 by means of real-time reverse-transcription polymerase chain reaction (rRT-PCR); viral culture was performed on a subset of specimens positive by rRT-PCR. The sensitivity of SS and ANS for SARS-CoV-2 detection by rRT-PCR was measured against that of NPS. Subgroup analyses included test outcomes by symptom status and culture results. RESULTS: Sensitivity for SARS-CoV-2 detection by rRT-PCR appeared higher for SS than for ANS (85% vs 80%) and higher among symptomatic participants than among those without symptoms (94% vs 29% for SS; 87% vs 50% for ANS). Among participants with culture-positive SARS-CoV-2 by any specimen type, the sensitivities of SS and ANS by rRT-PCR were 94% and 100%, respectively. SS and ANS were equally preferred by participants; most would undergo NPS collection again despite this method's being the least preferred. CONCLUSIONS: SS were slightly more sensitive than ANS for SARS-CoV-2 detection with rRT-PCR. With both SS and ANS, SARS-CoV-2 was reliably detected among participants with symptoms. Self-collected SS and ANS offer practical advantages, are preferred by patients, and might be most useful for testing people with coronavirus disease 2019 symptoms.


Subject(s)
COVID-19 , SARS-CoV-2 , Adult , COVID-19 Testing , Delivery of Health Care , Humans , Nasopharynx , Saliva , Specimen Handling
3.
Sex Transm Dis ; 46(3): 191-195, 2019 03.
Article in English | MEDLINE | ID: mdl-30363029

ABSTRACT

BACKGROUND: Rapid syphilis tests (RST) may shorten time to syphilis diagnosis and treatment while enhancing access to testing in outreach settings. There are limited data on the performance of RST in outreach settings in the US. METHODS: We offered RST (Syphilis Health Check) at 6 outreach sites to men who reported having sex with men and no prior history of syphilis. Clients accepting RST were also tested with laboratory-based rapid plasma reagin (RPR) and reflex Treponema pallidum particle agglutination (TPPA) assay when RPR or RST were positive. Clients with positive RST were immediately referred to a sexually transmitted infection clinic. Those declining RST were screened with RPR and reflex TPPA only. The validity of the RST-based algorithm was compared with the RPR-based algorithm among participants receiving both. Time to treatment for those accepting RST was compared with those declining RST and to a historical control group screened in outreach settings with RPR and reflex TPPA before the availability of RST. RESULTS: Rapid syphilis test was accepted by 690 (64%) of 1081 eligible clients. Compared with RPR-based algorithm, RST sensitivity was 90%; specificity, 98.5%; positive predictive value, 47.4%; and negative predictive value, 98.5. The single false-negative case by RST was determined to be a late latent case by RPR/TPPA. Median time to treatment was 1 day (range, 0-6 days) for 9 of 690 accepting RST, compared to 9 days (range, 7-13 days) for 3 of 391 declining RST, and 9 days (range, 6-21 days) for 25 of 1229 historical controls (P < 0.0001). CONCLUSION: Compared with an RPR-based algorithm, RST identified all early syphilis cases. Although RST had high specificity and negative predictive value, the low positive predictive value resulted in additional assessments in a sexually transmitted infection clinic for some patients. However, RST use in outreach settings significantly decreased time to treatment for new syphilis cases.


Subject(s)
Homosexuality, Male , Sexual and Gender Minorities , Syphilis Serodiagnosis/methods , Syphilis/diagnosis , Time-to-Treatment , Treponema pallidum/immunology , Adolescent , Adult , Child , Follow-Up Studies , Humans , Male , Middle Aged , Pilot Projects , Reagins/blood , Sensitivity and Specificity , Syphilis/microbiology , Young Adult
4.
Public Health Rep ; 133(1): 93-99, 2018.
Article in English | MEDLINE | ID: mdl-29258383

ABSTRACT

OBJECTIVES: Public health laboratories (PHLs) provide essential services in the diagnosis and surveillance of diseases of public health concern, such as tuberculosis. Maintaining access to high-quality laboratory testing is critical to continued disease detection and decline of tuberculosis cases in the United States. We investigated the practical experience of sharing tuberculosis testing services between PHLs through the Shared Services Project. METHODS: The Shared Services Project was a 9-month-long project funded through the Association of Public Health Laboratories and the Centers for Disease Control and Prevention during 2012-2013 as a one-time funding opportunity to consortiums of PHLs that proposed collaborative approaches to sharing tuberculosis laboratory services. Submitting PHLs maintained testing while simultaneously sending specimens to reference laboratories to compare turnaround times. RESULTS: During the 9-month project period, 107 Mycobacterium tuberculosis complex submissions for growth-based drug susceptibility testing and molecular detection of drug resistance testing occurred among the 3 consortiums. The median transit time for all submissions was 1.0 day. Overall, median drug susceptibility testing turnaround time (date of receipt in submitting laboratory to result) for parallel testing performed in house by submitting laboratories was 31.0 days; it was 43.0 days for reference laboratories. The median turnaround time for molecular detection of drug resistance results was 1.0 day (mean = 2.8; range, 0-14) from specimen receipt at the reference laboratories. CONCLUSIONS: The shared services model holds promise for specialized tuberculosis testing. Sharing of services requires a balance among quality, timeliness, efficiency, communication, and fiscal costs.


Subject(s)
Centers for Disease Control and Prevention, U.S./organization & administration , Laboratories/organization & administration , Public Health Practice , Tuberculosis/diagnosis , Bacteriological Techniques , Centers for Disease Control and Prevention, U.S./economics , Cooperative Behavior , Humans , Laboratories/economics , Public Health Surveillance/methods , United States
5.
Diagn Microbiol Infect Dis ; 86(2): 144-7, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27489119

ABSTRACT

Four commercial transport systems for the recovery of Neisseria gonorrhoeae were evaluated in support of the need to obtain culture isolates for the detection of antimicrobial resistance. Bacterial recovery from the InTray GC system was superior with minimal loss of viability in contrast to non-nutritive transport systems.


Subject(s)
Bacteriological Techniques/methods , Neisseria gonorrhoeae/isolation & purification , Specimen Handling/methods , Gonorrhea/diagnosis , Humans , Male , Microbial Viability , Neisseria gonorrhoeae/physiology
6.
J Virol ; 89(23): 11935-44, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26378172

ABSTRACT

UNLABELLED: Human alveolar epithelial cells (AECs) and alveolar macrophages (AMs) are the first lines of lung defense. Here, we report that AECs are the direct targets for H1N1 viruses that have circulated since the 2009 pandemic (H1N1pdm09). AMs are less susceptible to H1N1pdm09 virus, but they produce significantly more inflammatory cytokines than AECs from the same donor. AECs form an intact epithelial barrier that is destroyed by H1N1pdm09 infection. However, there is significant variation in the cellular permissiveness to H1N1pdm09 infection among different donors. AECs from obese donors appear to be more susceptible to H1N1pdm09 infection, whereas gender, smoking history, and age do not appear to affect AEC susceptibility. There is also a difference in response to different strains of H1N1pdm09 viruses. Compared to A/California04/09 (CA04), A/New York/1682/09 (NY1682) is more infectious and causes more epithelial barrier injury, although it stimulates less cytokine production. We further determined that a single amino acid residue substitution in NY1682 hemagglutinin is responsible for the difference in infectivity. In conclusion, this is the first study of host susceptibility of human lung primary cells and the integrity of the alveolar epithelial barrier to influenza. Further elucidation of the mechanism of increased susceptibility of AECs from obese subjects may facilitate the development of novel protection strategies against influenza virus infection. IMPORTANCE: Disease susceptibility of influenza is determined by host and viral factors. Human alveolar epithelial cells (AECs) form the key line of lung defenses against pathogens. Using primary AECs from different donors, we provided cellular level evidence that obesity might be a risk factor for increased susceptibility to influenza. We also compared the infections of two closely related 2009 pandemic H1N1 strains in AECs from the same donor and identified a key viral factor that affected host susceptibility, the dominance of which may be correlated with disease epidemiology. In addition, primary human AECs can serve as a convenient and powerful model to investigate the mechanism of influenza-induced lung injury and determine the effect of genetic and epigenetic factors on host susceptibility to pandemic influenza virus infection.


Subject(s)
Epithelial Cells/virology , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/physiopathology , Lung/cytology , Macrophages/metabolism , Obesity/complications , Pulmonary Alveoli/cytology , Adiposity , Cytokines/biosynthesis , Disease Susceptibility , Flow Cytometry , Hemagglutinins/genetics , Host-Pathogen Interactions , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza, Human/complications , Influenza, Human/virology , Lung/virology , Macrophages/virology , Species Specificity , Statistics, Nonparametric
7.
PLoS One ; 8(7): e70129, 2013.
Article in English | MEDLINE | ID: mdl-23894604

ABSTRACT

Because they are the natural target for respiratory pathogens, primary human respiratory epithelial cells provide the ideal in vitro system for isolation and study of human respiratory viruses, which display a high degree of cell, tissue, and host specificity. Human coronavirus HKU1, first discovered in 2005, has a worldwide prevalence and is associated with both upper and lower respiratory tract disease in both children and adults. Research on HCoV-HKU1 has been difficult because of its inability to be cultured on continuous cell lines and only recently it was isolated from clinical specimens using primary human, ciliated airway epithelial cells. Here we demonstrate that HCoV-HKU1 can infect and be serially propagated in primary human alveolar type II cells at the air-liquid interface. We were not able to infect alveolar type I-like cells or alveolar macrophages. Type II alveolar cells infected with HCoV-HKU1 demonstrated formation of large syncytium. At 72 hours post inoculation, HCoV-HKU1 infection of type II cells induced increased levels of mRNAs encoding IL29,CXCL10, CCL5, and IL-6 with no significant increases in the levels of IFNß. These studies demonstrate that type II cells are a target cell for HCoV-HKU1 infection in the lower respiratory tract, that type II alveolar cells are immune-competent in response to infection exhibiting a type III interferon and proinflammatory chemokine response, and that cell to cell spread may be a major factor for spread of infection. Furthermore, these studies demonstrate that human alveolar cells can be used to isolate and study novel human respiratory viruses that cause lower respiratory tract disease.


Subject(s)
Coronavirus Infections/immunology , Coronavirus/immunology , Immunity, Innate/physiology , Adolescent , Adult , Cells, Cultured , Child, Preschool , Coronavirus/pathogenicity , Coronavirus Infections/virology , Epithelial Cells/immunology , Epithelial Cells/virology , Female , Humans , Infant , Interleukin-6/metabolism , Macrophages, Alveolar/cytology , Macrophages, Alveolar/metabolism , Male , RNA, Viral , Respiratory Tract Infections/immunology , Respiratory Tract Infections/metabolism , Young Adult
8.
Am J Respir Cell Mol Biol ; 48(6): 742-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23418343

ABSTRACT

Severe acute respiratory syndrome (SARS)-coronavirus (CoV) produces a devastating primary viral pneumonia with diffuse alveolar damage and a marked increase in circulating cytokines. One of the major cell types to be infected is the alveolar type II cell. However, the innate immune response of primary human alveolar epithelial cells infected with SARS-CoV has not been defined. Our objectives included developing a culture system permissive for SARS-CoV infection in primary human type II cells and defining their innate immune response. Culturing primary human alveolar type II cells at an air-liquid interface (A/L) improved their differentiation and greatly increased their susceptibility to infection, allowing us to define their primary interferon and chemokine responses. Viral antigens were detected in the cytoplasm of infected type II cells, electron micrographs demonstrated secretory vesicles filled with virions, virus RNA concentrations increased with time, and infectious virions were released by exocytosis from the apical surface of polarized type II cells. A marked increase was evident in the mRNA concentrations of interferon-ß and interferon-λ (IL-29) and in a large number of proinflammatory cytokines and chemokines. A surprising finding involved the variability of expression of angiotensin-converting enzyme-2, the SARS-CoV receptor, in type II cells from different donors. In conclusion, the cultivation of alveolar type II cells at an air-liquid interface provides primary cultures in which to study the pulmonary innate immune responses to infection with SARS-CoV, and to explore possible therapeutic approaches to modulating these innate immune responses.


Subject(s)
Epithelial Cells/immunology , Immunity, Innate , Severe Acute Respiratory Syndrome/immunology , Severe acute respiratory syndrome-related coronavirus/immunology , Angiotensin-Converting Enzyme 2 , Antigens, Viral/immunology , Antigens, Viral/metabolism , Cell Differentiation , Cytoplasm/immunology , Cytoplasm/ultrastructure , Cytoplasm/virology , Epithelial Cells/virology , Humans , Interferon-beta/immunology , Interferon-beta/metabolism , Interferons , Interleukins/immunology , Interleukins/metabolism , Peptidyl-Dipeptidase A/immunology , Peptidyl-Dipeptidase A/metabolism , Primary Cell Culture , Pulmonary Alveoli/cytology , Pulmonary Alveoli/immunology , Pulmonary Alveoli/virology , RNA, Messenger/metabolism , Receptors, Virus/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/immunology , Respiratory Mucosa/virology , Time Factors , Virus Release
9.
PLoS One ; 7(3): e29879, 2012.
Article in English | MEDLINE | ID: mdl-22396727

ABSTRACT

Alveolar macrophages (AM) are one of the key cell types for initiating inflammatory and immune responses to influenza virus in the lung. However, the genome-wide changes in response to influenza infection in AM have not been defined. We performed gene profiling of human AM in response to H1N1 influenza A virus PR/8 using Affymetrix HG-U133 Plus 2.0 chips and verified the changes at both mRNA and protein levels by real-time RT-PCR and ELISA. We confirmed the response with a contemporary H3N2 influenza virus A/New York/238/2005 (NY/238). To understand the local cellular response, we also evaluated the impact of paracrine factors on virus-induced chemokine and cytokine secretion. In addition, we investigated the changes in the expression of macrophage receptors and uptake of pathogens after PR/8 infection. Although macrophages fail to release a large amount of infectious virus, we observed a robust induction of type I and type III interferons and several cytokines and chemokines following influenza infection. CXCL9, 10, and 11 were the most highly induced chemokines by influenza infection. UV-inactivation abolished virus-induced cytokine and chemokine response, with the exception of CXCL10. The contemporary influenza virus NY/238 infection of AM induced a similar response as PR/8. Inhibition of TNF and/or IL-1ß activity significantly decreased the secretion of the proinflammatory chemokines CCL5 and CXCL8 by over 50%. PR/8 infection also significantly decreased mRNA levels of macrophage receptors including C-type lectin domain family 7 member A (CLEC7A), macrophage scavenger receptor 1 (MSR1), and CD36, and reduced uptake of zymosan. In conclusion, influenza infection induced an extensive proinflammatory response in human AM. Targeting local components of innate immune response might provide a strategy for controlling influenza A infection-induced proinflammatory response in vivo.


Subject(s)
Gene Expression Regulation , Influenza A Virus, H1N1 Subtype/genetics , Influenza, Human/immunology , Macrophages/virology , Pulmonary Alveoli/virology , Chemokines/metabolism , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Humans , Immunity, Innate , Kinetics , Lectins, C-Type/biosynthesis , Macrophages/immunology , Oligonucleotide Array Sequence Analysis , Phagocytosis , Pulmonary Alveoli/immunology , Real-Time Polymerase Chain Reaction/methods
10.
J Gen Virol ; 93(Pt 3): 494-503, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22090214

ABSTRACT

Human coronavirus strain 229E (HCoV-229E) commonly causes upper respiratory tract infections. However, lower respiratory tract infections can occur in some individuals, indicating that cells in the distal lung are susceptible to HCoV-229E. This study determined the virus susceptibility of primary cultures of human alveolar epithelial cells and alveolar macrophages (AMs). Fluorescent antibody staining indicated that HCoV-229E could readily infect AMs, but no evidence was found for infection in differentiated alveolar epithelial type II cells and only a very low level of infection in type II cells transitioning to the type I-like cell phenotype. However, a human bronchial epithelial cell line (16HBE) was readily infected. The innate immune response of AMs to HCoV-229E infection was evaluated for cytokine production and interferon (IFN) gene expression. AMs secreted significant amounts of tumour necrosis factor alpha (TNF-α), regulated on activation normal T-cell expressed and secreted (RANTES/CCL5) and macrophage inflammatory protein 1ß (MIP-1ß/CCL4) in response to HCoV-229E infection, but these cells exhibited no detectable increase in IFN-ß or interleukin-29 in mRNA levels. AMs from smokers had reduced secretion of TNF-α compared with non-smokers in response to HCoV-229E infection. Surfactant protein A (SP-A) and SP-D are part of the innate immune system in the distal lung. Both surfactant proteins bound to HCoV-229E, and pre-treatment of HCoV-229E with SP-A or SP-D inhibited infection of 16HBE cells. In contrast, there was a modest reduction in infection in AMs by SP-A, but not by SP-D. In summary, AMs are an important target for HCoV-229E, and they can mount a pro-inflammatory innate immune response to infection.


Subject(s)
Coronavirus 229E, Human/pathogenicity , Macrophages, Alveolar/virology , Cells, Cultured , Cytokines/biosynthesis , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/virology , Fluorescent Antibody Technique, Direct , Gene Expression , Gene Expression Profiling , Humans , Macrophages, Alveolar/immunology , Viral Plaque Assay
11.
J Virol ; 85(24): 13195-203, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21937653

ABSTRACT

The first influenza pandemic of the 21st century was caused by novel H1N1 viruses that emerged in early 2009. An Asp-to-Gly change at position 222 of the receptor-binding protein hemagglutinin (HA) correlates with more-severe infections in humans. The amino acid at position 222 of HA contributes to receptor-binding specificity with Asp (typically found in human influenza viruses) and Gly (typically found in avian and classic H1N1 swine influenza viruses), conferring binding to human- and avian-type receptors, respectively. Here, we asked whether binding to avian-type receptors enhances influenza virus pathogenicity. We tested two 2009 pandemic H1N1 viruses possessing HA-222G (isolated from severe cases) and two viruses that possessed HA-222D. In glycan arrays, viruses possessing HA-222D preferentially bound to human-type receptors, while those encoding HA-222G bound to both avian- and human-type receptors. This difference in receptor binding correlated with efficient infection of viruses possessing HA-222G, compared to those possessing HA-222D, in human lung tissue, including alveolar type II pneumocytes, which express avian-type receptors. In a nonhuman primate model, infection with one of the viruses possessing HA-222G caused lung damage more severe than did infection with a virus encoding HA-222D, although these pathological differences were not observed for the other virus pair with either HA-222G or HA-222D. These data demonstrate that the acquisition of avian-type receptor-binding specificity may result in more-efficient infection of human alveolar type II pneumocytes and thus more-severe lung damage. Collectively, these findings suggest a new mechanism by which influenza viruses may become more pathogenic in mammals, including humans.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A Virus, H1N1 Subtype/pathogenicity , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Receptors, Virus/metabolism , Virus Internalization , Animals , Cell Line , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Lung/pathology , Lung/virology , Macaca , Receptors, Virus/genetics
12.
J Virol ; 85(14): 6844-55, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21543489

ABSTRACT

Highly pathogenic influenza H5N1 virus continues to pose a threat to public health. Although the mechanisms underlying the pathogenesis of the H5N1 virus have not been fully defined, it has been suggested that cytokine dysregulation plays an important role. As the human respiratory epithelium is the primary target cell for influenza viruses, elucidating the viral tropism and innate immune responses of influenza H5N1 virus in the alveolar epithelium may help us to understand the pathogenesis of the severe pneumonia associated with H5N1 disease. Here we used primary cultures of differentiated human alveolar type II cells, alveolar type I-like cells, and alveolar macrophages isolated from the same individual to investigate viral replication competence and host innate immune responses to influenza H5N1 (A/HK/483/97) and H1N1 (A/HK/54/98) virus infection. The viral replication kinetics and cytokine and chemokine responses were compared by quantitative PCR (qPCR) and enzyme-linked immunosorbent assay (ELISA). We demonstrated that influenza H1N1 and H5N1 viruses replicated productively in type II cells and type I-like cells although with different kinetics. The H5N1 virus replicated productively in alveolar macrophages, whereas the H1N1 virus led to an abortive infection. The H5N1 virus was a more potent inducer of proinflammatory cytokines and chemokines than the H1N1 virus in all cell types. However, higher levels of cytokine expression were observed for peripheral blood monocyte-derived macrophages than for alveolar macrophages in response to H5N1 virus infection. Our findings provide important insights into the viral tropisms and host responses of different cell types found in the lung and are relevant to an understanding of the pathogenesis of severe human influenza disease.


Subject(s)
Immunity, Innate , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H5N1 Subtype/physiology , Macrophages, Alveolar/virology , Pulmonary Alveoli/virology , Virus Replication , Cell Differentiation , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/virology , Humans , Macrophages, Alveolar/immunology , Polymerase Chain Reaction , Pulmonary Alveoli/cytology , Pulmonary Alveoli/immunology
13.
Am J Respir Cell Mol Biol ; 45(3): 582-91, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21239608

ABSTRACT

Alveolar Type II (ATII) cells are important targets for seasonal and pandemic influenza. To investigate the influenza-induced innate immune response in those cells, we measured the global gene expression profile of highly differentiated ATII cells infected with the influenza A virus at a multiplicity of infection of 0.5 at 4 hours and 24 hours after inoculation. Infection with influenza stimulated a significant increase in the mRNA concentrations of many host defense-related genes, including pattern/pathogen recognition receptors, IFN, and IFN-induced genes, chemokines, and suppressors of cytokine signaling. We verified these changes by quantitative real-time RT-PCR. At the protein level, we detected a robust virus-induced secretion of the three glutamic acid-leucine-arginine (ELR)-negative chemokines CXCL9, CXCL10, and CXCL11, according to ELISA. The ultraviolet inactivation of virus abolished the chemokine and cytokine response. Viral infection did not appear to alter the differentiation of ATII cells, as measured by cellular mRNA and concentrations of surfactant proteins. However, viral infection significantly reduced the secretion of surfactant protein (SP)-A and SP-D. In addition, influenza A virus triggered a time-dependent activation of phosphatidylinositol 3-kinase signaling in ATII cells. The inhibition of this pathway significantly decreased the release of infectious virus and the chemokine response, but did not alter virus-induced cell death. This study provides insights into influenza-induced innate immunity in differentiated human ATII cells, and demonstrates that the alveolar epithelium is a critical part of the initial innate immune response to influenza.


Subject(s)
Immunity, Innate , Influenza A virus/metabolism , Pulmonary Alveoli/cytology , Adult , Aged , Aged, 80 and over , Cell Differentiation , Chemokines/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Male , Middle Aged , Phosphatidylinositol 3-Kinases/metabolism , Smoking , Surface-Active Agents/metabolism
14.
J Gen Virol ; 90(Pt 12): 2956-2964, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19741068

ABSTRACT

The rat coronavirus sialodacryoadenitis virus (SDAV) causes respiratory infection and provides a system for investigating respiratory coronaviruses in a natural host. A viral suspension in the form of a microspray aerosol was delivered by intratracheal instillation into the distal lung of 6-8-week-old Fischer 344 rats. SDAV inoculation produced a 7 % body weight loss over a 5 day period that was followed by recovery over the next 7 days. SDAV caused focal lesions in the lung, which were most severe on day 4 post-inoculation (p.i.). Immunofluorescent staining showed that four cell types supported SDAV virus replication in the lower respiratory tract, namely Clara cells, ciliated cells in the bronchial airway and alveolar type I and type II cells in the lung parenchyma. In bronchial alveolar lavage fluid (BALF) a neutrophil influx increased the population of neutrophils to 45 % compared with 6 % of the cells in control samples on day 2 after mock inoculation. Virus infection induced an increase in surfactant protein SP-D levels in BALF of infected rats on days 4 and 8 p.i. that subsided by day 12. The concentrations of chemokines MCP-1, LIX and CINC-1 in BALF increased on day 4 p.i., but returned to control levels by day 8. Intratracheal instillation of rats with SDAV coronavirus caused an acute, self-limited infection that is a useful model for studying the early events of the innate immune response to respiratory coronavirus infections in lungs of the natural virus host.


Subject(s)
Coronavirus Infections , Coronavirus, Rat/pathogenicity , Epithelial Cells/virology , Lung/virology , Pulmonary Alveoli/virology , Virus Replication , Animals , Coronavirus Infections/immunology , Coronavirus Infections/physiopathology , Coronavirus Infections/virology , Coronavirus, Rat/physiology , Cytokines/metabolism , Immunity, Innate , Lung/cytology , Male , Pulmonary Alveoli/cytology , Pulmonary Surfactants/metabolism , Rats , Rats, Inbred F344 , Weight Loss
15.
BMC Cancer ; 8: 260, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18793421

ABSTRACT

BACKGROUND: Dysregulated expression and splicing of cell adhesion marker CD44 is found in many types of cancer. In prostate cancer (PC) specifically, the standard isoform (CD44s) has been found to be downregulated compared with benign tissue whereas predominant variant isoform CD44v7-10 is upregulated. Mitogen-activated protein kinase pathways and paracrine calcitonin are two common factors linked to dysregulated expression and splicing of CD44 in cancer. Calcitonin has been found to increase proliferation and invasion in PC acting through the protein kinase A pathway. METHODS: In androgen-independent PC with known high CD44v7-10 expression, CD44 total and CD44v7-10 RNA or protein were assessed in response to exogenous and endogenous calcitonin and to inhibitors of protein kinase A, MEK, JNK, or p38 kinase. Benign cells and calcitonin receptor-negative PC cells were also tested. RESULTS: MEK or p38 but not JNK reduced CD44 total RNA by 40%-65% in cancer and benign cells. Inhibition of protein kinase A reduced CD44 total and v7-10 protein expression. In calcitonin receptor-positive cells only, calcitonin increased CD44 variant RNA and protein by 3 h and persisting to 48 h, apparently dependent on an uninhibited p38 pathway. Cells with constitutive CT expression showed an increase in CD44v7-10 mRNA but a decrease in CD44 total RNA. CONCLUSION: The MEK pathway increases CD44 RNA, while calcitonin, acting through the protein kinase A and p38 pathway, facilitates variant splicing. These findings could be used in the formulation of therapeutic methods for PC targeting CD44 alternate splicing.


Subject(s)
Antigens, Neoplasm/metabolism , Calcitonin/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Hyaluronan Receptors/metabolism , Mitogen-Activated Protein Kinases/physiology , Prostatic Neoplasms/metabolism , Alternative Splicing , Calcitonin/pharmacology , Cell Line, Tumor , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , Male , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Prostatic Neoplasms/pathology , Protein Isoforms/metabolism , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/metabolism
16.
J Virol ; 82(6): 2883-94, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18199653

ABSTRACT

The severe acute respiratory syndrome coronavirus (SARS-CoV) spike glycoprotein (S) is a class I viral fusion protein that binds to its receptor glycoprotein, human angiotensin converting enzyme 2 (hACE2), and mediates virus entry and cell-cell fusion. The juxtamembrane domain (JMD) of S is an aromatic amino acid-rich region proximal to the transmembrane domain that is highly conserved in all coronaviruses. Alanine substitutions for one or two of the six aromatic residues in the JMD did not alter the surface expression of the SARS-CoV S proteins with a deletion of the C-terminal 19 amino acids (S Delta19) or reduce binding to soluble human ACE2 (hACE2). However, hACE2-dependent entry of trypsin-treated retrovirus pseudotyped viruses expressing JMD mutant S Delta19 proteins was greatly reduced. Single alanine substitutions for aromatic residues reduced entry to 10 to 60% of the wild-type level. The greatest reduction was caused by residues nearest the transmembrane domain. Four double alanine substitutions reduced entry to 5 to 10% of the wild-type level. Rapid hACE2-dependent S-mediated cell-cell fusion was reduced to 60 to 70% of the wild-type level for all single alanine substitutions and the Y1188A/Y1191A protein. S Delta19 proteins with other double alanine substitutions reduced cell-cell fusion further, from 40% to less than 20% of wild-type levels. The aromatic amino acids in the JMD of the SARS-CoV S glycoprotein play critical roles in receptor-dependent virus-cell and cell-cell fusion. Because the JMD is so highly conserved in all coronavirus S proteins, it is a potential target for development of drugs that may inhibit virus entry and/or cell-cell fusion mediated by S proteins of all coronaviruses.


Subject(s)
Amino Acids, Aromatic/physiology , Cell Fusion , Membrane Glycoproteins/physiology , Membrane Proteins/physiology , Receptors, Virus/physiology , Severe acute respiratory syndrome-related coronavirus/physiology , Viral Envelope Proteins/physiology , Amino Acid Sequence , Amino Acids, Aromatic/chemistry , Base Sequence , Cell Line , DNA Primers , Humans , Membrane Glycoproteins/chemistry , Membrane Proteins/chemistry , Molecular Sequence Data , Receptors, Virus/chemistry , Sequence Homology, Amino Acid , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/chemistry
17.
J Virol ; 82(2): 755-63, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18003729

ABSTRACT

The MHV-JHM strain of the murine coronavirus mouse hepatitis virus is much more neurovirulent than the MHV-A59 strain, although both strains use murine CEACAM1a (mCEACAM1a) as the receptor to infect murine cells. We previously showed that Ceacam1a(-/-) mice are completely resistant to MHV-A59 infection (E. Hemmila et al., J. Virol. 78:10156-10165, 2004). In vitro, MHV-JHM, but not MHV-A59, can spread from infected murine cells to cells that lack mCEACAM1a, a phenomenon called receptor-independent spread. To determine whether MHV-JHM could infect and spread in the brain independent of mCEACAM1a, we inoculated Ceacam1a(-/-) mice. Although Ceacam1a(-/-) mice were completely resistant to i.c. inoculation with 10(6) PFU of recombinant wild-type MHV-A59 (RA59) virus, these mice were killed by recombinant MHV-JHM (RJHM) and a chimeric virus containing the spike of MHV-JHM in the MHV-A59 genome (SJHM/RA59). Immunohistochemistry showed that RJHM and SJHM/RA59 infected all neural cell types and induced severe microgliosis in both Ceacam1a(-/-) and wild-type mice. For RJHM, the 50% lethal dose (LD(50)) is <10(1.3) in wild-type mice and 10(3.1) in Ceacam1a(-/-) mice. For SJHM/RA59, the LD(50) is <10(1.3) in wild-type mice and 10(3.6) in Ceacam1a(-/-) mice. This study shows that infection and spread of MHV-JHM in the brain are dependent upon the viral spike glycoprotein. RJHM can initiate infection in the brains of Ceacam1a(-/-) mice, but expression of mCEACAM1a increases susceptibility to infection. The spread of infection in the brain is mCEACAM1a independent. Thus, the ability of the MHV-JHM spike to mediate mCEACAM1a-independent spread in the brain is likely an important factor in the severe neurovirulence of MHV-JHM in wild-type mice.


Subject(s)
Carcinoembryonic Antigen/genetics , Central Nervous System/virology , Coronavirus Infections/virology , Membrane Glycoproteins/physiology , Murine hepatitis virus/growth & development , Viral Envelope Proteins/physiology , Virus Internalization , Animals , Brain/pathology , Brain/virology , Gene Deletion , Lethal Dose 50 , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Murine hepatitis virus/physiology , Spike Glycoprotein, Coronavirus , Survival Analysis , Viral Envelope Proteins/genetics , Virulence Factors/genetics , Virulence Factors/physiology
19.
Transgenic Res ; 13(5): 411-25, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15587266

ABSTRACT

The Class II transposable element, piggyBac, was used to transform the yellow fever mosquito, Aedes aegypti. In two transformed lines only 15-30% of progeny inherited the transgene, with these individuals displaying mosaic expression of the EGFP marker gene. Southern analyses, gene amplification of genomic DNA, and plasmid rescue experiments provided evidence that these lines contained a high copy number of piggyBac transformation constructs and that much of this DNA consisted of both donor and helper plasmids. A detailed analysis of one line showed that the majority of piggyBac sequences were unit-length donor or helper plasmids arranged in a large tandem array that could be lost en masse in a single generation. Despite the presence of a transposase source and many intact donor elements, no conservative (cut and paste) transposition of piggyBac was observed in these lines. These results reveal one possible outcome of uncontrolled and/or unexpected recombination in this mosquito, and support the conclusion that further investigation is necessary before transposable elements such as piggyBac can be used as genetic drive mechanisms to move pathogen-resistance genes into mosquito populations.


Subject(s)
Aedes/genetics , Animals, Genetically Modified , DNA Transposable Elements/genetics , Genes, Insect , Transformation, Genetic , Animals , Baculoviridae/genetics , Drosophila melanogaster/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Situ Hybridization, Fluorescence , Mutagenesis, Insertional , Promoter Regions, Genetic
20.
Insect Biochem Mol Biol ; 34(7): 607-13, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15242701

ABSTRACT

Diseases caused by arthropod-borne viruses are significant public health problems, and novel methods are needed to control pathogen transmission. We hypothesize that genetic manipulation of Aedes aegypti mosquitoes can profoundly and permanently reduce vector competence and subsequent transmission of dengue viruses (DENV) to human hosts. We have identified RNA interference (RNAi) as a potential anti-viral, intracellular pathway in the vector that can be triggered by expression of virus-specific, double stranded RNAs (dsRNAs) to reduce vector competence to DENV. We identified DENV-derived RNA segments using recombinant Sindbis viruses to trigger RNAi, that when expressed in mosquitoes ablate homologous DENV replication and transmission. We also demonstrated that heritable expression of DENV-derived dsRNA in cultured mosquito cells can silence virus replication. We now have developed a number of transgenic mosquito lines that transcribe the effector dsRNA from constitutive promoters such as immediate early 1 (baculovirus) and polyubiquitin (Drosophila melanogaster). We have detected DENV-specific small interfering RNAs, the hallmark of RNAi, in at least one of these lines. Surprisingly, none of these lines expressed dsRNA in relevant tissues (e.g., midguts) that will ultimately affect transmission. A major challenge now is to express the effector dsRNA from tissue-specific promoters to allow RNAi to silence virus replication at critical sites in the vector such as midguts and salivary glands. If successful, this strategy has the advantage of harnessing a naturally occurring vector response to block DENV infection in a mosquito vector and profoundly affect virus transmission.


Subject(s)
Aedes/genetics , Aedes/virology , Dengue Virus/genetics , RNA Interference , Animals , Animals, Genetically Modified , Antiviral Agents , Carboxypeptidases/genetics , Carboxypeptidases/metabolism , Dengue Virus/pathogenicity , Dengue Virus/physiology , RNA, Double-Stranded/genetics , RNA, Small Interfering/genetics , RNA, Viral/genetics , Recombinant Proteins/metabolism , Virus Replication/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...