Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Histopathology ; 77(6): 915-925, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32614086

ABSTRACT

INTRODUCTION: We describe post-mortem pulmonary histopathologic findings of COVID-19 pneumonia in patients with a spectrum of disease course, from rapid demise to prolonged hospitalisation. METHODS AND RESULTS: Histopathologic findings in post-mortem lung tissue from eight patients who died from COVID-19 pneumonia were reviewed. Immunohistochemistry (IHC) and next-generation sequencing (NGS) were performed to detect virus. Diffuse alveolar damage (DAD) was seen in all cases with a spectrum of acute phase and/or organising phase. IHC with monoclonal antibodies against SARS-CoV-2 viral nucleoprotein and spike protein detected virus in areas of acute but not organising DAD, with intracellular viral antigen and RNA expression seen predominantly in patients with duration of illness less than 10 days. Major vascular findings included thrombi in medium- and large-calibre vessels, platelet microthrombi detected by CD61 IHC and fibrin microthrombi. CONCLUSIONS: Presence of SARS-CoV-2 viral RNA by NGS early in the disease course and expression of viral antigen by IHC exclusively in the acute, but not in the organising phase of DAD, suggests that the virus may play a major role in initiating the acute lung injury of DAD, but when DAD progresses to the organising phase the virus may have been cleared from the lung by the patient's immune response. These findings suggest the possibility of a major change during the disease course of COVID-19 pneumonia that may have therapeutic implications. Frequent thrombi and microthrombi may also present potential targets for therapeutic intervention.


Subject(s)
Coronavirus Infections/pathology , Pneumonia, Viral/pathology , Adult , Aged , Autopsy , Betacoronavirus , COVID-19 , Coronavirus Infections/mortality , Female , Humans , Immunohistochemistry , Male , Middle Aged , Pandemics , Pneumonia, Viral/mortality , Pneumonia, Viral/virology , RNA, Viral , SARS-CoV-2
2.
J Virol ; 92(4)2018 02 15.
Article in English | MEDLINE | ID: mdl-29167344

ABSTRACT

The fifth wave of A(H7N9) virus infection in China from 2016 to 2017 caused great concern due to the large number of individuals infected, the isolation of drug-resistant viruses, and the emergence of highly pathogenic strains. Antibodies against neuraminidase (NA) provide added benefit to hemagglutinin-specific immunity and may be important contributors to the effectiveness of A(H7N9) vaccines. We generated a panel of mouse monoclonal antibodies (MAbs) to identify antigenic domains on NA of the novel A(H7N9) virus and compared their functional properties. The loop formed in the region of residue 250 (250 loop) and the domain formed by the loops containing residues 370, 400, and 430 were identified as major antigenic regions. MAbs 1E8, 2F6, 10F4, and 11B2, which recognize these two antigenic domains, were characterized in depth. These four MAbs differ in their abilities to inhibit cleavage of small and large substrates (methyl-umbelliferyl-acetyl neuraminic acid [MU-NANA] and fetuin, respectively) in NA inhibition assays. 1E8 and 11B2 did not inhibit NA cleavage of either MU-NANA or fetuin, and 2F6 inhibited cleavage of fetuin alone, whereas 10F4 inhibited cleavage of both substrates. All four MAbs reduced the in vitro spread of viruses carrying either the wild-type N9 or N9 with antiviral-resistant mutations but to different degrees. These MAbs have different in vivo levels of effectiveness: 10F4 was the most effective in protecting mice against challenge with A(H7N9) virus, 2F6 was less effective, and 11B2 failed to protect BALB/c mice at the doses tested. Our study confirms that NA-specific antibodies can protect against A(H7N9) infection and suggests that in vitro properties can be used to rank antibodies with therapeutic potential.IMPORTANCE The novel A(H7N9) viruses that emerged in China in 2013 continue to infect humans, with a high fatality rate. The most recent outbreak resulted in a larger number of human cases than previous epidemic waves. Due to the absence of a licensed vaccine and the emergence of drug-resistant viruses, there is a need to develop alternative approaches to prevent or treat A(H7N9) infection. We have made a panel of mouse monoclonal antibodies (MAbs) specific for neuraminidase (NA) of A(H7N9) viruses; some of these MAbs are effective in inhibiting viruses that are resistant to antivirals used to treat A(H7N9) patients. Binding avidity, inhibition of NA activity, and plaque formation correlated with the effectiveness of these MAbs to protect mice against lethal A(H7N9) virus challenge. This study identifies in vitro measures that can be used to predict the in vivo efficacy of NA-specific antibodies, providing a way to select MAbs for further therapeutic development.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/therapeutic use , Neuraminidase/immunology , Orthomyxoviridae Infections/prevention & control , Viral Proteins/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , China , Disease Models, Animal , Dogs , Female , HEK293 Cells , Humans , Influenza A Virus, H7N9 Subtype , Lung/pathology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Reassortant Viruses
3.
J Pathol ; 238(1): 85-97, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26383585

ABSTRACT

To study bacterial co-infection following 1918 H1N1 influenza virus infection, mice were inoculated with the 1918 influenza virus, followed by Streptococcus pneumoniae (SP) 72 h later. Co-infected mice exhibited markedly more severe disease, shortened survival time and more severe lung pathology, including widespread thrombi. Transcriptional profiling revealed activation of coagulation only in co-infected mice, consistent with the extensive thrombogenesis observed. Immunohistochemistry showed extensive expression of tissue factor (F3) and prominent deposition of neutrophil elastase on endothelial and epithelial cells in co-infected mice. Lung sections of SP-positive 1918 autopsy cases showed extensive thrombi and prominent staining for F3 in alveolar macrophages, monocytes, neutrophils, endothelial and epithelial cells, in contrast to co-infection-positive 2009 pandemic H1N1 autopsy cases. This study reveals that a distinctive feature of 1918 influenza virus and SP co-infection in mice and humans is extensive expression of tissue factor and activation of the extrinsic coagulation pathway leading to widespread pulmonary thrombosis.


Subject(s)
Coinfection/complications , Influenza, Human/microbiology , Orthomyxoviridae Infections/microbiology , Pneumococcal Infections/microbiology , Pulmonary Embolism/microbiology , Animals , Blood Coagulation , Disease Models, Animal , Female , Humans , Immunohistochemistry , Influenza A Virus, H1N1 Subtype , Influenza Pandemic, 1918-1919 , Influenza, Human/complications , Influenza, Human/pathology , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/pathology , Pneumococcal Infections/complications , Pneumococcal Infections/pathology , Pulmonary Embolism/pathology , Reverse Transcriptase Polymerase Chain Reaction , Streptococcus pneumoniae
4.
J Pathol ; 229(4): 535-45, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23180419

ABSTRACT

Most biopsy and autopsy tissues are formalin-fixed and paraffin-embedded (FFPE), but this process leads to RNA degradation that limits gene expression analysis. The RNA genome of the 1918 pandemic influenza virus was previously determined in a 9-year effort by overlapping RT-PCR from post-mortem samples. Here, the full genome of the 1918 virus at 3000× coverage was determined in one high-throughput sequencing run of a library derived from total RNA of a 1918 FFPE sample after duplex-specific nuclease treatments. Bacterial sequences associated with secondary bacterial pneumonias were also detected. Host transcripts were well represented in the library. Compared to a 2009 pandemic influenza virus FFPE post-mortem library, the 1918 sample showed significant enrichment for host defence and cell death response genes, concordant with prior animal studies. This methodological approach should assist in the analysis of FFPE tissue samples isolated over the past century from a variety of diseases.


Subject(s)
Genome, Viral/genetics , High-Throughput Nucleotide Sequencing/methods , Influenza A virus/genetics , Influenza, Human/virology , Lung/virology , Epithelial Cells/virology , Female , Formaldehyde , Gene Library , Humans , Influenza A virus/isolation & purification , Lung/microbiology , Lung/pathology , Male , Middle Aged , Molecular Sequence Annotation , Pandemics , Paraffin Embedding , RNA/genetics , RNA Stability , Sequence Analysis, RNA , Young Adult
5.
Proc Natl Acad Sci U S A ; 108(39): 16416-21, 2011 Sep 27.
Article in English | MEDLINE | ID: mdl-21930918

ABSTRACT

The 1918 to 1919 "Spanish" influenza pandemic virus killed up to 50 million people. We report here clinical, pathological, bacteriological, and virological findings in 68 fatal American influenza/pneumonia military patients dying between May and October of 1918, a period that includes ~4 mo before the 1918 pandemic was recognized, and 2 mo (September-October 1918) during which it appeared and peaked. The lung tissues of 37 of these cases were positive for influenza viral antigens or viral RNA, including four from the prepandemic period (May-August). The prepandemic and pandemic peak cases were indistinguishable clinically and pathologically. All 68 cases had histological evidence of bacterial pneumonia, and 94% showed abundant bacteria on Gram stain. Sequence analysis of the viral hemagglutinin receptor-binding domain performed on RNA from 13 cases suggested a trend from a more "avian-like" viral receptor specificity with G222 in prepandemic cases to a more "human-like" specificity associated with D222 in pandemic peak cases. Viral antigen distribution in the respiratory tree, however, was not apparently different between prepandemic and pandemic peak cases, or between infections with viruses bearing different receptor-binding polymorphisms. The 1918 pandemic virus was circulating for at least 4 mo in the United States before it was recognized epidemiologically in September 1918. The causes of the unusually high mortality in the 1918 pandemic were not explained by the pathological and virological parameters examined. These findings have important implications for understanding the origins and evolution of pandemic influenza viruses.


Subject(s)
Autopsy , Influenza, Human/mortality , Antigens, Viral/analysis , History, 20th Century , Humans , Influenza A virus/genetics , Influenza A virus/immunology , Influenza, Human/epidemiology , Influenza, Human/history , Molecular Sequence Data , RNA, Viral/analysis
6.
mBio ; 15(3): e0321823, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38349163

ABSTRACT

The 1918 influenza pandemic was the most devastating respiratory pandemic in modern human history, with 50-100 million deaths worldwide. Here, we characterized the complete genomes of influenza A virus (IAV) from two fatal cases during the fall wave of 1918 influenza A (H1N1) pandemic in the United States, one from Walter Reed Army Hospital in Washington, DC, and the other from Camp Jackson, SC. The two complete IAV genomes were obtained by combining Illumina deep sequencing data from both total RNA and influenza viral genome-enriched libraries along with Sanger sequencing data from PCR across the sequencing gaps. This study confirms the previously reported 1918 IAV genomes and increases the total number of available complete or near-complete influenza viral genomes of the 1918 pandemic from four to six. Sequence comparisons among them confirm that the genomes of the 1918 pandemic virus were highly conserved during the main wave of the pandemic with geographic separation in North America and Europe. Metagenomic analyses revealed bacterial co-infections in both cases. Interestingly, in the Washington, DC, case, evidence is presented of the first reported Rhodococcus-influenza virus co-infection. IMPORTANCE: This study applied modern molecular biotechnology and high-throughput sequencing to formalin-fixed, paraffin-embedded autopsy lung samples from two fatal cases during the fall wave of the 1918 influenza A (H1N1) pandemic in the United States. Complete influenza genomes were obtained from both cases, which increases the total number of available complete or near-complete influenza genomes of the 1918 pandemic virus from four to six. Sequence analysis confirms that the 1918 pandemic virus was highly conserved during the main wave of the pandemic with geographic separation in North America and Europe. Metagenomic analyses revealed bacterial co-infections in both cases, including the first reported evidence of Rhodococcus-influenza co-infection. Overall, this study offers a detailed view at the molecular level of the very limited samples from the most devastating influenza pandemic in modern human history.


Subject(s)
Coinfection , Influenza A Virus, H1N1 Subtype , Influenza A virus , Influenza, Human , Orthomyxoviridae Infections , Humans , Influenza A Virus, H1N1 Subtype/genetics , RNA , Coinfection/genetics , Paraffin Embedding , Lung , Influenza A virus/genetics , Genome, Viral , High-Throughput Nucleotide Sequencing , Formaldehyde , Autopsy
7.
J Infect Dis ; 201(9): 1397-403, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20350163

ABSTRACT

BACKGROUND: The overall impact of influenza virus infection in immunocompromised patients is largely unknown. Antigenic drift and genetic variations during prolonged influenza infection have been demonstrated. In this report we describe a multidrug-resistant H3N2 influenza virus isolated from an immunocompromised patient after 5 days of therapy. METHODS: Multiple nasal wash samples were collected from an infected patient, and viral isolates were characterized. Sensitivity to antiviral agents was evaluated. Fitness and transmissibility were assessed in ferrets and tissue culture. RESULTS: An in-frame 4-amino acid deletion emerged in the neuraminidase (NA) gene of an H3N2 virus after 5 days of oseltamivir therapy. No other changes in the NA or hemagglutinin genes were noted. Drug sensitivity assays revealed resistance to oseltamivir (>10-fold increase in 50% inhibitory concentration [IC(50)]) and reduction in sensitivity to zanamivir (3-7-fold increase in IC(50) or 50% effective concentration). No change in fitness or transmissibility was observed. CONCLUSIONS: An in-frame NA gene deletion was rapidly selected for in an immunocompromised patient, resulting in decreased sensitivity of the isolate to available NA inhibitors without a change in fitness or transmissibility. This finding has implications for our understanding of the emergence of antiviral resistance and treatment of patients with influenza A infection, especially those who are immunocompromised.


Subject(s)
Immunocompromised Host , Influenza A Virus, H3N2 Subtype/drug effects , Influenza, Human/virology , Adult , Animals , Drug Resistance, Multiple, Viral/genetics , Ferrets/virology , Humans , Influenza A Virus, H3N2 Subtype/genetics , Influenza, Human/drug therapy , Influenza, Human/immunology , Influenza, Human/transmission , Lymphoma, Mantle-Cell/complications , Lymphoma, Mantle-Cell/virology , Male , Molecular Sequence Data , Orthomyxoviridae Infections/transmission , Orthomyxoviridae Infections/virology , Reverse Transcriptase Polymerase Chain Reaction , Virus Replication/genetics
8.
Sci Transl Med ; 13(620): eabj7790, 2021 Nov 17.
Article in English | MEDLINE | ID: mdl-34648357

ABSTRACT

Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is characterized by respiratory distress, multiorgan dysfunction, and, in some cases, death. The pathological mechanisms underlying COVID-19 respiratory distress and the interplay with aggravating risk factors have not been fully defined. Lung autopsy samples from 18 patients with fatal COVID-19, with symptom onset-to-death times ranging from 3 to 47 days, and antemortem plasma samples from 6 of these cases were evaluated using deep sequencing of SARS-CoV-2 RNA, multiplex plasma protein measurements, and pulmonary gene expression and imaging analyses. Prominent histopathological features in this case series included progressive diffuse alveolar damage with excessive thrombosis and late-onset pulmonary tissue and vascular remodeling. Acute damage at the alveolar-capillary barrier was characterized by the loss of surfactant protein expression with injury to alveolar epithelial cells, endothelial cells, respiratory epithelial basal cells, and defective tissue repair processes. Other key findings included impaired clot fibrinolysis with increased concentrations of plasma and lung plasminogen activator inhibitor-1 and modulation of cellular senescence markers, including p21 and sirtuin-1, in both lung epithelial and endothelial cells. Together, these findings further define the molecular pathological features underlying the pulmonary response to SARS-CoV-2 infection and provide important insights into signaling pathways that may be amenable to therapeutic intervention.


Subject(s)
COVID-19 , Cellular Senescence , Fibrinolysis , Humans , Lung , SARS-CoV-2
9.
Nat Biotechnol ; 24(5): 555-7, 2006 May.
Article in English | MEDLINE | ID: mdl-16617336

ABSTRACT

We describe an ultrasensitive immunoassay for detecting biotoxins that uses liposomes with encapsulated DNA reporters, and ganglioside receptors embedded in the bilayer, as a detection reagent. After immobilization of the target biotoxin by a capture antibody and co-binding of the detection reagent, the liposomes are ruptured to release the reporters, which are quantified by real-time PCR. Assays for cholera and botulinum toxins are several orders of magnitude more sensitive than current detection methods.


Subject(s)
Immunoassay/methods , Liposomes/chemistry , Polymerase Chain Reaction/methods , Toxins, Biological/analysis , Botulinum Toxins/analysis , Cholera Toxin/analysis , DNA/chemistry , Gangliosides/chemistry , Lipid Bilayers/chemistry , Polymerase Chain Reaction/instrumentation , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity
10.
Virology ; 524: 182-191, 2018 11.
Article in English | MEDLINE | ID: mdl-30212665

ABSTRACT

Influenza virus infections in humans and animals are major public health concerns. In the current study, a set of universal influenza enrichment probes was developed to increase the sensitivity of sequence-based virus detection and characterization for all influenza viruses. This universal influenza enrichment probe set contains 46,953 120nt RNA biotin-labeled probes designed based on all available influenza viral sequences and it can be used to enrich for influenza sequences without prior knowledge of type or subtype. Marked enrichment was demonstrated in influenza A/H1N1, influenza B, and H1-to-H16 hemagglutinin plasmids spiked into human DNA and in cultured influenza A/H2N1 virus. Furthermore, enrichment effects and mixed influenza A virus infections were revealed in wild bird cloacal swab samples. Therefore, this universal influenza virus enrichment probe system can capture and enrich influenza viral sequences selectively and effectively in different samples, especially ones with degraded RNA or containing low amount of influenza RNA.


Subject(s)
DNA Probes/genetics , Influenza A virus/isolation & purification , Influenza in Birds/epidemiology , Animals , Birds , Cloaca/virology , Epidemiological Monitoring , High-Throughput Nucleotide Sequencing/veterinary , Influenza A virus/genetics , Influenza in Birds/virology , RNA, Viral/analysis , RNA, Viral/isolation & purification , Sequence Analysis, DNA/veterinary
11.
Diagn Cytopathol ; 35(8): 498-502, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17636483

ABSTRACT

We tested whether methylation profiles generated by real-time methylation-specific PCR (MSP) can be useful in differentiating benign, reactive mesothelial cell proliferation (RM) from malignant mesothelioma (MM). Forty-two of the 63 cases (67%) yielded informative results for RARbeta2, GPC3, CDKN2A (p16), TERT, and CCND2 (cyclinD2) gene methylation. DNA methylation of any gene was observed in much higher frequency in MM cases than RM cases (63% vs. 33%, P < 0.05). Individual gene methylation was higher in the MM than the RM cases for most of the genes; however, this was not statistically significant (RARbeta2: 58% vs. 33%, P > 0.05; GPC3: 36% vs. 27%, P > 0.05; CDKN2A: 4% vs. 0%; TERT: 4% vs. 0%), while CCND2 methylation was not detected in any case. Although preliminary, we demonstrate that real-time MSP can be applied to archival specimens and gene methylation profiling may have potential to be a useful ancillary tool to help distinguish MM from RM.


Subject(s)
DNA Methylation , Epithelium/pathology , Mesothelioma/diagnosis , Mesothelioma/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , DNA, Neoplasm/genetics , Female , Humans , Male , Middle Aged , Pilot Projects
12.
Antiviral Res ; 129: 120-129, 2016 May.
Article in English | MEDLINE | ID: mdl-26923881

ABSTRACT

BACKGROUND: Influenza results in up to 500,000 deaths annually. Seasonal influenza vaccines have an estimated 60% effectiveness, but provide little or no protection against novel subtypes, and may be less protective in high-risk groups. Neuraminidase inhibitors are recommended for the treatment of severe influenza infection, but are not proven to reduce mortality in severe disease. Preclinical models of severe influenza infection that closely correlate to human disease are needed to assess efficacy of new vaccines and therapeutics. METHODS: We developed a nonhuman primate model of influenza and bacterial co-infection that recapitulates severe pneumonia in humans. Animals were infected with influenza A virus via intra-bronchial or small-particle aerosol inoculation, methicillin-resistant Staphylococcus aureus, or co-infected with influenza and methicillin-resistant S. aureus combined. We assessed the severity of disease in animals over the course of our study using tools available to evaluate critically ill human patients including high-resolution computed tomography imaging of the lungs, arterial blood gas analyses, and bronchoalveolar lavage. RESULTS: Using an intra-bronchial route of inoculation we successfully induced severe pneumonia following influenza infection alone and following influenza and bacterial co-infection. Peak illness was observed at day 6 post-influenza infection, manifested by bilateral pulmonary infiltrates and hypoxemia. The timing of radiographic and physiologic manifestations of disease in our model closely match those observed in severe human influenza infection. DISCUSSION: This was the first nonhuman primate study of influenza and bacterial co-infection where high-resolution computed tomography scanning of the lungs was used to quantitatively assess pneumonia over the course of illness and where hypoxemia was correlated with pneumonia severity. With additional validation this model may serve as a pathway for regulatory approval of vaccines and therapeutics for the prevention and treatment of severe influenza pneumonia.


Subject(s)
Coinfection , Influenza A virus , Models, Animal , Orthomyxoviridae Infections/complications , Pneumonia, Staphylococcal/complications , Pneumonia, Viral/complications , Animals , Humans , Influenza A virus/pathogenicity , Influenza Vaccines , Influenza, Human/complications , Influenza, Human/microbiology , Lung/microbiology , Lung/pathology , Lung/virology , Macaca mulatta , Male , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Positron Emission Tomography Computed Tomography
14.
Curr Protoc Microbiol ; 37: 1E.8.1-16, 2015 May 01.
Article in English | MEDLINE | ID: mdl-26344216

ABSTRACT

The vast majority of surgical biopsy and post-mortem tissue samples are formalin-fixed and paraffin-embedded (FFPE), but this process leads to RNA degradation that limits gene expression analysis. As an example, the viral RNA genome of the 1918 pandemic influenza A virus was previously determined in a 9-year effort by overlapping RT-PCR from post-mortem samples. Using the protocols described here, the full genome of the 1918 virus was determined at high coverage in one high-throughput sequencing run of a cDNA library derived from total RNA of a 1918 FFPE sample after duplex-specific nuclease treatments. This basic methodological approach should assist in the analysis of FFPE tissue samples isolated over the past century from a variety of infectious diseases.


Subject(s)
Gene Library , High-Throughput Nucleotide Sequencing , Influenza A Virus, H1N1 Subtype/genetics , Pathology, Molecular/methods , RNA, Viral/isolation & purification , Humans , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza Pandemic, 1918-1919 , RNA, Viral/genetics
16.
Acta Cytol ; 46(6): 1069-74, 2002.
Article in English | MEDLINE | ID: mdl-12462084

ABSTRACT

OBJECTIVE: To investigate the role of reverse transcriptase polymerase chain reaction (RT-PCR) in determining telomerase catalytic subunit (hTERT) mRNA to assist with the diagnosis of colonic adenocarcinoma (CCA) in colonic brush cytology specimens. STUDY DESIGN: Twenty-seven colonic brushes of CCA were obtained. Initial cytologic diagnoses included CCA, suspicious for CCA (SFC), atypical (ATY) and unsatisfactory. The cytologic specimens were reviewed. A homogeneous RT-PCR assay for hTERT mRNA was performed on 27 colonic brushes of CCA and 24 controls (10 negative brushes, 6 resected CCA and 8 benign colonic mucosa from resected specimens). A RT-PCR assay for beta 2-microglobulin was used as an internal control for mRNA quality. RESULTS: On review, the initial cytopathologic diagnosis of CCA was confirmed in all 7 cases. In addition, 7 of 19 initially interpreted as SFC and ATY seemed to demonstrate unequivocal cytomorphologic features of malignancy. Telomerase mRNA was more often expressed in CCA than in negative controls in both brush (30%) and resected specimens (57%) (P < .05). Seven cases with hTERT mRNA expression were initially diagnosed as CCA (three), SFC (three) and ATY (one). CONCLUSION: CCA may be underdiagnosed in brush cytopathology. The expression of hTERT mRNA may be determined by RT-PCR in brush specimens and may eventually prove to be a useful diagnostic adjunct in the interpretation of inconclusive cases of CCA. Sparse cellularity in brush specimens may result in a relatively low rate of hTERT detection in colonic brushes of CCA; inflammatory changes may contribute to a higher-than-expected rate of hTERT expression in benign colonic epithelium.


Subject(s)
Adenocarcinoma/pathology , Colonic Neoplasms/pathology , Telomerase/genetics , Biomarkers, Tumor , Biopsy , DNA-Binding Proteins , Gene Expression Regulation, Neoplastic , Humans , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , beta 2-Microglobulin/genetics
17.
Virology ; 417(1): 98-105, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21658737

ABSTRACT

From August 2 to October 11, 2006, clusters of low pathogenicity (LP) North American lineage H5N1 and H7N3 avian influenza A viruses (AIV), and other subtypes, were recovered from free-flying, resident, wild mallards used as sentinels at one site. The antigenic subtypes, pathogenicity potential, and Sanger sequencing of the isolates determined the H5N1 and H7N3 isolates were only recovered from samples collected on 8/2/2006 and 9/8/2006, respectively. However, subsequent efforts using next-generation sequencing (NGS) and additional Sanger sequencing found partial H7 segments in other HA-NA virus combinations on 8/2/2006, 9/8/2006 and 10/11/2006. It is well established that over larger geographic areas and years AIVs form transient genomic constellations; this sequential sampling data revealed that over a short period of time the dynamics of AIVs can be active and newer sequencing platforms increase recognition of mixed infections. Both findings provide further insight into the natural history of AIVs in natural reservoirs.


Subject(s)
Ducks , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H7N3 Subtype/genetics , Influenza in Birds/virology , Animals , Animals, Wild , Feces/virology , Influenza A Virus, H5N1 Subtype/classification , Influenza A Virus, H5N1 Subtype/isolation & purification , Influenza A Virus, H7N3 Subtype/classification , Influenza A Virus, H7N3 Subtype/isolation & purification , Influenza in Birds/epidemiology , Molecular Sequence Data , North America/epidemiology , Phylogeny , Sentinel Surveillance
18.
Influenza Other Respir Viruses ; 5(3): 198-205, 2011 May.
Article in English | MEDLINE | ID: mdl-21477139

ABSTRACT

BACKGROUND: Zoonotic infections with H1N1 influenza viruses that evolved initially from the 1918 virus (1918) and adapted to swine threatened a pandemic in 1976 (1976 swH1N1) and a novel reassortant H1N1 virus caused a pandemic in 2009-2010 (2009 pH1N1). Epidemiological and laboratory animal studies show that protection from severe 2009 pH1N1 infection is conferred by vaccination or prior infection with 1976 swH1N1 or 1918. OBJECTIVES: Our aim was to demonstrate cross-protection by immunization with 2009 pH1N1 or 1976 swH1N1 vaccines following a lethal challenge with 1918. Further, the mechanisms of cross-protective antibody responses were evaluated. METHODS: Mice were immunized with 1976 swH1N1, 2009 pH1N1, 2009 seasonal trivalent, or 1918 vaccines and challenged with 1918. Cross-reactive antibody responses were assessed and protection monitored by survival, weight loss, and pathology in mice. RESULTS AND CONCLUSIONS: Vaccination with the 1976 swH1N1 or 2009 pH1N1 vaccines protected mice from a lethal challenge with 1918, and these mice lost no weight and had significantly reduced viral load and pathology in the lungs. Protection was likely due to cross-reactive antibodies detected by microneutralization assay. Our data suggest that the general population may be protected from a future 1918-like pandemic because of prior infection or immunization with 1976 swH1N1 or 2009 pH1N1. Also, influenza protection studies generally focus on cross-reactive hemagglutination-inhibiting antibodies; while hemagglutinin is the primary surface antigen, this fails to account for other influenza viral antigens. Neutralizing antibody may be a better correlate of human protection against pathogenic influenza strains and should be considered for vaccine efficacy.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Orthomyxoviridae Infections/veterinary , Swine Diseases/virology , Animals , China/epidemiology , Cross Protection , Female , Humans , Immunization , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza Vaccines/administration & dosage , Influenza, Human/epidemiology , Influenza, Human/virology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/virology , Pandemics , Swine , Swine Diseases/epidemiology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
19.
mBio ; 1(1)2010 May 18.
Article in English | MEDLINE | ID: mdl-20689744

ABSTRACT

The swine-origin H1N1 influenza A virus emerged in early 2009 and caused the first influenza pandemic in 41 years. The virus has spread efficiently to both the Northern and the Southern Hemispheres and has been associated with over 16,000 deaths. Given the virus's recent zoonotic origin, there is concern that the virus could acquire signature mutations associated with the enhanced pathogenicity of previous pandemic viruses or H5N1 viruses with pandemic potential. We tested the hypothesis that mutations in the polymerase PB2 gene at residues 627 and 701 would enhance virulence but found that influenza viruses containing these mutations in the context of the pandemic virus polymerase complex are attenuated in cell culture and mice.

20.
Arch Pathol Lab Med ; 134(2): 235-43, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20121613

ABSTRACT

CONTEXT: In March 2009, a novel swine-origin influenza A/H1N1 virus was identified. After global spread, the World Health Organization in June declared the first influenza pandemic in 41 years. OBJECTIVE: To describe the clinicopathologic characteristics of 34 people who died following confirmed A/H1N1 infection with emphasis on the pulmonary pathology findings. DESIGN: We reviewed medical records, autopsy reports, microbiologic studies, and microscopic slides of 34 people who died between May 15 and July 9, 2009, and were investigated either by the New York City Office of Chief Medical Examiner (32 deaths) or through the consultation service of a coauthor (2 deaths). RESULTS: Most of the 34 decedents (62%) were between 25 and 49 years old (median, 41.5 years). Tracheitis, bronchiolitis, and diffuse alveolar damage were noted in most cases. Influenza viral antigen was observed most commonly in the epithelium of the tracheobronchial tree but also in alveolar epithelial cells and macrophages. Most cases were reverse transcription-polymerase chain reaction positive for influenza. Histologic and microbiologic autopsy evidence of bacterial pneumonia was detected in 55% of cases. Underlying medical conditions including cardiorespiratory diseases and immunosuppression were present in 91% of cases. Obesity (body mass index, >30) was noted in 72% of adult and adolescent cases. CONCLUSIONS: The pulmonary pathologic findings in fatal disease caused by the novel pandemic influenza virus are similar to findings identified in past pandemics. Superimposed bacterial infections of the respiratory tract were common. Preexisting obesity, cardiorespiratory diseases, and other comorbidities also were prominent findings among the decedents.


Subject(s)
Disease Outbreaks , Influenza A Virus, H1N1 Subtype , Influenza, Human/pathology , Lung/pathology , Adolescent , Adult , Aged , Child , Child, Preschool , Comorbidity , Female , Humans , Infant , Infant, Newborn , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza, Human/diagnostic imaging , Male , Middle Aged , Pneumonia, Bacterial/microbiology , Radiography , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL