35 results on '"Lindsay Droit"'
Search Results
2. Tracking the prevalence and emergence of SARS CoV2 variants of concern using a regional genomic surveillance program
- Author
-
Ana Jung, Lindsay Droit, Binita Febles, Catarina Fronick, Lisa Cook, Scott A. Handley, Bijal A Parikh, and David Wang
- Abstract
SARS-CoV-2 molecular testing coupled with whole genome sequencing is instrumental for real-time genomic surveillance. Genomic surveillance is critical for monitoring the spread of variants of concern (VOC) as well as novel variant discovery. Since the beginning of the pandemic millions of SARS-CoV-2 genomes have been deposited into public sequence databases. This is the result of efforts of both national and regional diagnostic laboratories. Here we describe the results of SARS-CoV-2 genomic surveillance from February 2021 to June 2022 at a metropolitan hospital in the USA. We demonstrate that consistent daily sampling is sufficient to track the regional prevalence and emergence of VOC. Similar sampling efforts should be considered a viable option for local SARS-CoV-2 genomic surveillance at other regional laboratories.
- Published
- 2023
3. Novel crAssphage isolates exhibit conserved gene order and purifying selection of the host specificity protein
- Author
-
Bhavya Papudeshi, Alejandro A. Vega, Cole Souza, Sarah K. Giles, Vijini Mallawaarachchi, Michael J. Roach, Michelle An, Nicole Jacobson, Katelyn McNair, Maria Fernanda Mora, Karina Pastrana, Christopher Leigh, Clarice Cram, Will S. Plewa, Susanna R. Grigson, George Bouras, Przemysław Decewicz, Antoni Luque, Lindsay Droit, Scott A. Handley, Anca M. Segall, Elizabeth A. Dinsdale, and Robert A. Edwards
- Abstract
Bacteroidota are the most common bacteria in the human gut and are responsible for degrading complex polysaccharides that would otherwise remain undigested. The abundance of Bacteroides in the gut is shaped by phages such as crAssphages that infect and kill them. While close to 600 genomes have been identified computationally, only four have been successfully cultured. Here, we identify and characterize three novel crAssphage species isolated from wastewater and infecting the bacterial hostBacteroides cellulosilyticusWH2. We named the novel species,Kehishuvirus winsdale(Bc01),Kolpuevirus frurule(Bc03), andRudgehvirus redwords(Bc11) which span two different families and three genera. These phages may not have co-evolved with their respective bacterial hosts. The phages had a conserved gene arrangement with known crAssphages, but gene similarity within phages belonging to the same taxa was highly variable. Across the three species, only two structural genes encoding a hypothetical protein and a tail spike protein were similar. Evolutionary analysis revealed the tail spike protein is undergoing purifying selection and was predicted to bind to a TonB-dependent transporter on the host cell surface, suggesting a role for host specificity. This study expands the known crAssphage isolates and reveals insights into the crAssphage infection mechanism. The availability of pure cultures of multiple crAssphage infecting the same host provides an opportunity to perform controlled experiments on one of the most dominant members of the human enteric virome.
- Published
- 2023
4. Resistance of SARS-CoV-2 variants to neutralization by monoclonal and serum-derived polyclonal antibodies
- Author
-
Alfred H.J. Kim, Mahima Thapa, Jane A. O’Halloran, Emma S. Winkler, Aaron J. Schmitz, Xianwen Zhang, Herbert W. Virgin, Jackson S. Turner, Seth J. Zost, John M. Errico, Xuping Xie, Davide Corti, Lindsay Droit, Yang Liu, Naveenchandra Suryadevara, Daved H. Fremont, Stephen Tahan, Pei Yong Shi, Jianying Liu, Wooseob Kim, Michael S. Diamond, Parakkal Deepak, Adrianus C. M. Boon, James E. Crowe, Dora Pinto, James Brett Case, Laura A. VanBlargan, Ali H. Ellebedy, Pavlo Gilchuk, Rachel M. Presti, Rita E. Chen, and David Wang
- Subjects
0301 basic medicine ,biology ,medicine.drug_class ,General Medicine ,Monoclonal antibody ,Virology ,General Biochemistry, Genetics and Molecular Biology ,Neutralization ,In vitro ,law.invention ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,law ,Polyclonal antibodies ,030220 oncology & carcinogenesis ,Monoclonal ,medicine ,Vero cell ,Recombinant DNA ,biology.protein ,Antibody - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused the global COVID-19 pandemic. Rapidly spreading SARS-CoV-2 variants may jeopardize newly introduced antibody and vaccine countermeasures. Here, using monoclonal antibodies (mAbs), animal immune sera, human convalescent sera and human sera from recipients of the BNT162b2 mRNA vaccine, we report the impact on antibody neutralization of a panel of authentic SARS-CoV-2 variants including a B.1.1.7 isolate, chimeric strains with South African or Brazilian spike genes and isogenic recombinant viral variants. Many highly neutralizing mAbs engaging the receptor-binding domain or N-terminal domain and most convalescent sera and mRNA vaccine-induced immune sera showed reduced inhibitory activity against viruses containing an E484K spike mutation. As antibodies binding to spike receptor-binding domain and N-terminal domain demonstrate diminished neutralization potency in vitro against some emerging variants, updated mAb cocktails targeting highly conserved regions, enhancement of mAb potency or adjustments to the spike sequences of vaccines may be needed to prevent loss of protection in vivo.
- Published
- 2021
5. Human enteric viruses autonomously shape inflammatory bowel disease phenotype through divergent innate immunomodulation
- Author
-
Fatemeh Adiliaghdam, Hajera Amatullah, Sreehaas Digumarthi, Tahnee L. Saunders, Raza-Ur Rahman, Lai Ping Wong, Ruslan Sadreyev, Lindsay Droit, Jean Paquette, Philippe Goyette, John D. Rioux, Richard Hodin, Kathie A. Mihindukulasuriya, Scott A. Handley, and Kate L. Jeffrey
- Subjects
Inflammation ,Immunology ,General Medicine ,Inflammatory Bowel Diseases ,Article ,digestive system diseases ,Gastrointestinal Microbiome ,Immunomodulation ,Mice ,Phenotype ,Viruses ,Animals ,Humans ,Enterovirus - Abstract
Altered enteric microorganisms in concert with host genetics shape inflammatory bowel disease (IBD) phenotypes. However, insight is limited to bacteria and fungi. We found that eukaryotic viruses and bacteriophages (collectively, the virome), enriched from non-IBD, noninflamed human colon resections, actively elicited atypical anti-inflammatory innate immune programs. Conversely, ulcerative colitis or Crohn’s disease colon resection viromes provoked inflammation, which was successfully dampened by non-IBD viromes. The IBD colon tissue virome was perturbed, including an increase in the enterovirus B species of eukaryotic picornaviruses, not previously detected in fecal virome studies. Mice humanized with non-IBD colon tissue viromes were protected from intestinal inflammation, whereas IBD virome mice exhibited exacerbated inflammation in a nucleic acid sensing–dependent fashion. Furthermore, there were detrimental consequences for IBD patient–derived intestinal epithelial cells bearing loss-of-function mutations within virus sensor MDA5 when exposed to viromes. Our results demonstrate that innate recognition of IBD or non-IBD human viromes autonomously influences intestinal homeostasis and disease phenotypes. Thus, perturbations in the intestinal virome, or an altered ability to sense the virome due to genetic variation, contribute to the induction of IBD. Harnessing the virome may offer therapeutic and biomarker potential.
- Published
- 2022
6. Resilience of S309 and AZD7442 monoclonal antibody treatments against infection by SARS-CoV-2 Omicron lineage strains
- Author
-
James Brett Case, Samantha Mackin, John M. Errico, Zhenlu Chong, Emily A. Madden, Bradley Whitener, Barbara Guarino, Michael A. Schmid, Kim Rosenthal, Kuishu Ren, Ha V. Dang, Gyorgy Snell, Ana Jung, Lindsay Droit, Scott A. Handley, Peter J. Halfmann, Yoshihiro Kawaoka, James E. Crowe, Daved H. Fremont, Herbert W. Virgin, Yueh-Ming Loo, Mark T. Esser, Lisa A. Purcell, Davide Corti, and Michael S. Diamond
- Subjects
Multidisciplinary ,Membrane Glycoproteins ,SARS-CoV-2 ,General Physics and Astronomy ,Antibodies, Monoclonal ,General Chemistry ,Antibodies, Viral ,Antibodies, Neutralizing ,General Biochemistry, Genetics and Molecular Biology ,COVID-19 Drug Treatment ,Drug Combinations ,Mice ,Viral Envelope Proteins ,Neutralization Tests ,Spike Glycoprotein, Coronavirus ,Animals ,Humans - Abstract
Omicron variant strains encode large numbers of changes in the spike protein compared to historical SARS-CoV-2 isolates. Although in vitro studies have suggested that several monoclonal antibody therapies lose neutralizing activity against Omicron variants, the effects in vivo remain largely unknown. Here, we report on the protective efficacy against three SARS-CoV-2 Omicron lineage strains (BA.1, BA.1.1, and BA.2) of two monoclonal antibody therapeutics (S309 [Vir Biotechnology] monotherapy and AZD7442 [AstraZeneca] combination), which correspond to ones used to treat or prevent SARS-CoV-2 infections in humans. Despite losses in neutralization potency in cell culture, S309 or AZD7442 treatments reduced BA.1, BA.1.1, and BA.2 lung infection in susceptible mice that express human ACE2 (K18-hACE2) in prophylactic and therapeutic settings. Correlation analyses between in vitro neutralizing activity and reductions in viral burden in K18-hACE2 or human FcγR transgenic mice suggest that S309 and AZD7442 have different mechanisms of protection against Omicron variants, with S309 utilizing Fc effector function interactions and AZD7442 acting principally by direct neutralization. Our data in mice demonstrate the resilience of S309 and AZD7442 mAbs against emerging SARS-CoV-2 variant strains and provide insight into the relationship between loss of antibody neutralization potency and retained protection in vivo.
- Published
- 2022
7. Resilience of S309 and AZD7442 monoclonal antibody treatments against infection by SARS-CoV-2 Omicron lineage strains
- Author
-
James Brett Case, Samantha Mackin, John Errico, Zhenlu Chong, Emily A. Madden, Barbara Guarino, Michael A. Schmid, Kim Rosenthal, Kuishu Ren, Ana Jung, Lindsay Droit, Scott A. Handley, Peter J. Halfmann, Yoshihiro Kawaoka, James E. Crowe, Daved H. Fremont, Herbert W. Virgin, Yueh-Ming Loo, Mark T. Esser, Lisa A. Purcell, Davide Corti, and Michael S. Diamond
- Abstract
Omicron variant strains encode large numbers of changes in the spike protein compared to historical SARS-CoV-2 isolates. Although in vitro studies have suggested that several monoclonal antibody therapies lose neutralizing activity against Omicron variants1-4, the effects in vivo remain largely unknown. Here, we report on the protective efficacy against three SARS-CoV-2 Omicron lineage strains (BA.1, BA.1.1, and BA.2) of two monoclonal antibody therapeutics (S309 [Vir Biotechnology] monotherapy and AZD7442 [AstraZeneca] combination), which correspond to ones used to treat or prevent SARS-CoV-2 infections in humans. Despite losses in neutralization potency in cell culture, S309 or AZD7442 treatments reduced BA.1, BA.1.1, and BA.2 lung infection in susceptible mice that express human ACE2 (K18-hACE2). Correlation analyses between in vitro neutralizing activity and reductions in viral burden in K18-hACE2 or human Fcγ R transgenic mice suggest that S309 and AZD7442 have different mechanisms of protection against Omicron variants, with S309 utilizing Fc effector function interactions and AZD7442 acting principally by direct neutralization. Our data in mice demonstrate the resilience of S309 and AZD7442 mAbs against emerging SARS-CoV-2 variant strains and provide insight into the relationship between loss of antibody neutralization potency and retained protection in vivo.
- Published
- 2022
8. Select autophagy genes maintain quiescence of tissue-resident macrophages and increase susceptibility to Listeria monocytogenes
- Author
-
Dale R. Balce, Megan T. Baldridge, Herbert W. Virgin, Qun Lu, Chandni Desai, Konstantin Zaitsev, Lindsay Droit, Craig B. Wilen, Sunmin Park, Darren Kreamalmeyer, Shan Li, Maxim N. Artyomov, Ki-Wook Kim, Christina L. Stallings, Anthony Orvedahl, John D. Pfeifer, Ya-Ting Wang, Scott A. Handley, Robert C. Orchard, and W. Timothy Schaiff
- Subjects
Male ,Microbiology (medical) ,Immunology ,ATG5 ,Autophagy-Related Proteins ,Biology ,medicine.disease_cause ,Applied Microbiology and Biotechnology ,Microbiology ,Article ,Interferon-gamma ,Mice ,03 medical and health sciences ,Immune system ,Listeria monocytogenes ,Immunity ,Autophagy ,Genetics ,medicine ,Animals ,Genetic Predisposition to Disease ,Listeriosis ,Gene ,ATG16L1 ,Cell Proliferation ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,030306 microbiology ,Cell Biology ,Macrophage Activation ,Cell biology ,Mice, Inbred C57BL ,Macrophages, Peritoneal ,Beclin-1 ,Female ,Disease Susceptibility ,Homeostasis - Abstract
Innate and adaptive immune responses that prime myeloid cells, such as macrophages, protect against pathogens1,2. However, if left uncontrolled, these responses may lead to detrimental inflammation3. Macrophages, particularly those resident in tissues, must therefore remain quiescent between infections despite chronic stimulation by commensal microorganisms. The genes required for quiescence of tissue-resident macrophages are not well understood. Autophagy, an evolutionarily conserved cellular process by which cytoplasmic contents are targeted for lysosomal digestion, has homeostatic functions including maintenance of protein and organelle integrity and regulation of metabolism4. Recent research has shown that degradative autophagy, as well as various combinations of autophagy genes, regulate immunity and inflammation5–12. Here, we delineate a function of the autophagy proteins Beclin 1 and FIP200—but not of other essential autophagy components ATG5, ATG16L1 or ATG7—in mediating quiescence of tissue-resident macrophages by limiting the effects of systemic interferon-γ. The perturbation of quiescence in mice that lack Beclin 1 or FIP200 in myeloid cells results in spontaneous immune activation and resistance to Listeria monocytogenes infection. While antibiotic-treated wild-type mice display diminished macrophage responses to inflammatory stimuli, this is not observed in mice that lack Beclin 1 in myeloid cells, establishing the dominance of this gene over effects of the bacterial microbiota. Thus, select autophagy genes, but not all genes essential for degradative autophagy, have a key function in maintaining immune quiescence of tissue-resident macrophages, resulting in genetically programmed susceptibility to bacterial infection. The autophagy proteins Beclin 1 and FIP200, but not other essential autophagy components, such as ATG5, ATG16L1 or ATG7, regulate quiescence of tissue-resident macrophages, thereby modulating immune activation and resistance to Listeria monocytogenes infection.
- Published
- 2020
9. Single-cell genomics for resolution of conserved bacterial genes and mobile genetic elements of the human intestinal microbiota using flow cytometry
- Author
-
Dylan Lawrence, Danielle E. Campbell, Lawrence A. Schriefer, Rachel Rodgers, Forrest C. Walker, Marissa Turkin, Lindsay Droit, Miles Parkes, Scott A. Handley, Megan T. Baldridge, Baldridge, Megan T [0000-0002-7030-6131], and Apollo - University of Cambridge Repository
- Subjects
Microbiology (medical) ,prophage ,Bacteria ,Gastroenterology ,High-Throughput Nucleotide Sequencing ,mobile genetic elements ,RC799-869 ,Genomics ,Diseases of the digestive system. Gastroenterology ,Flow Cytometry ,Microbiology ,Gastrointestinal Microbiome ,genomic assembly ,Interspersed Repetitive Sequences ,Feces ,Infectious Diseases ,single-amplified genome ,Humans ,Single cell ,Single-Cell Analysis ,Genome, Bacterial ,Phylogeny - Abstract
As our understanding of the importance of the human microbiota in health and disease grows, so does our need to carefully resolve and delineate its genomic content. 16S rRNA gene-based analyses yield important insights into taxonomic composition, and metagenomics-based approaches reveal the functional potential of microbial communities. However, these methods generally fail to directly link genetic features, including bacterial genes and mobile genetic elements, to each other and to their source bacterial genomes. Further, they are inadequate to capture the microdiversity present within a genus, species, or strain of bacteria within these complex communities. Here, we present a method utilizing fluorescence-activated cell sorting for isolation of single bacterial cells, amplifying their genomes, screening them by 16S rRNA gene analysis, and selecting cells for genomic sequencing. We apply this method to both a cultured laboratory strain of Escherichia coli and human stool samples. Our analyses reveal the capacity of this method to provide nearly complete coverage of bacterial genomes when applied to isolates and partial genomes of bacterial species recovered from complex communities. Additionally, this method permits exploration and comparison of conserved and variable genomic features between individual cells. We generate assemblies of novel genomes within the Ruminococcaceae family and the Holdemanella genus by combining several 16S rRNA gene-matched single cells, and report novel prophages and conjugative transposons for both Bifidobacterium and Ruminococcaceae. Thus, we demonstrate an approach for flow cytometric separation and sequencing of single bacterial cells from the human microbiota, which yields a variety of critical insights into both the functional potential of individual microbes and the variation among those microbes. This method definitively links a variety of conserved and mobile genomic features, and can be extended to further resolve diverse elements present in the human microbiota.
- Published
- 2022
10. Neutralizing antibodies protect mice against Venezuelan equine encephalitis virus aerosol challenge
- Author
-
Natasha M. Kafai, Lauren E. Williamson, Elad Binshtein, Soila Sukupolvi-Petty, Christina L. Gardner, Jaclyn Liu, Samantha Mackin, Arthur S. Kim, Nurgun Kose, Robert H. Carnahan, Ana Jung, Lindsay Droit, Douglas S. Reed, Scott A. Handley, William B. Klimstra, James E. Crowe, and Michael S. Diamond
- Subjects
Aerosols ,Encephalitis Virus, Venezuelan Equine ,Mice ,Immunology ,Cryoelectron Microscopy ,Immunology and Allergy ,Animals ,Antibodies, Monoclonal ,Encephalomyelitis, Venezuelan Equine ,Viral Vaccines ,Horses ,Antibodies, Viral ,Antibodies, Neutralizing - Abstract
Venezuelan equine encephalitis virus (VEEV) remains a risk for epidemic emergence or use as an aerosolized bioweapon. To develop possible countermeasures, we isolated VEEV-specific neutralizing monoclonal antibodies (mAbs) from mice and a human immunized with attenuated VEEV strains. Functional assays and epitope mapping established that potently inhibitory anti-VEEV mAbs bind distinct antigenic sites in the A or B domains of the E2 glycoprotein and block multiple steps in the viral replication cycle including attachment, fusion, and egress. A 3.2-Å cryo-electron microscopy reconstruction of VEEV virus-like particles bound by a human Fab suggests that antibody engagement of the B domain may result in cross-linking of neighboring spikes to prevent conformational requirements for viral fusion. Prophylaxis or postexposure therapy with these mAbs protected mice against lethal aerosol challenge with VEEV. Our study defines functional and structural mechanisms of mAb protection and suggests that multiple antigenic determinants on VEEV can be targeted for vaccine or antibody-based therapeutic development.
- Published
- 2021
11. Human enteric viruses shape disease phenotype through divergent innate immunomodulation
- Author
-
Kathie A. Mihindukulasuriya, Richard A. Hodin, Jean Paquette, Lindsay Droit, Sreehaas Digumarthi, Scot A. Handley, Philippe Goyette, Lai Ping Wong, John D. Rioux, Raza-Ur Rahman, Ruslan I. Sadreyev, Hajera Amatullah, Tahnee L. Saunders, Kate L. Jeffrey, and Fatemeh Adiliaghdam
- Subjects
Innate immune system ,Inflammation ,MDA5 ,Biology ,medicine.disease ,Phenotype ,Inflammatory bowel disease ,digestive system diseases ,Virus ,Biomarker ,Immunology ,medicine ,Human virome ,medicine.symptom - Abstract
SummaryAltered enteric microorganisms in concert with host genetics shape inflammatory bowel disease (IBD) phenotypes. However, insight is limited to bacteria and fungi. We found virus like particles (VLPs) enriched from normal human colon resections, containing eukaryotic viruses and bacteriophages (collectively, the virome), actively elicited atypical anti-inflammatory innate immune programs. Conversely, IBD patient VLPs provoked inflammation, which was successfully dampened by healthy VLPs. The IBD colon tissue virome was perturbed, including enriched PicornovirusEnterovirus B,not previously observed in fecal virome studies. Mice with humanized healthy colon tissue viromes had attenuated intestinal inflammation while those with IBD-derived viromes exhibited exacerbated inflammation in a nucleic acid sensing-dependent fashion. Furthermore, there were detrimental consequences for IBD-associated MDA5 loss-of-function on patient intestinal epithelial cells exposed to healthy or IBD viromes. Our results demonstrate that innate recognition of either healthy or IBD human viromes autonomously influences disease phenotypes in IBD. Harnessing the virome may offer therapeutic and biomarker potential.One Sentence SummaryHuman viromes divergently shape host immunity and disease
- Published
- 2021
12. Reduced antibody activity against SARS-CoV-2 B.1.617.2 Delta virus in serum of mRNA-vaccinated patients receiving TNF-α inhibitors
- Author
-
Laura A. VanBlargan, Jane A. O’Halloran, Salim Chahin, Parakkal Deepak, Lindsay Droit, Juan Manuel Carreño, Rachel M. Presti, Michael A. Paley, Daniel Y. Zhu, Florian Krammer, Scott A. Handley, Rita E. Chen, Ali H. Ellebedy, Galit Alter, Douglas A. Lauffenburger, Wooseob Kim, Alfred H.J. Kim, Samantha Burdess, Dansu Yuan, Jackson S. Turner, Michael S. Diamond, Matthew J. Gorman, and Gregory F. Wu
- Subjects
education.field_of_study ,biology ,business.industry ,medicine.drug_class ,medicine.medical_treatment ,Population ,Immunosuppression ,Monoclonal antibody ,Virus ,Vaccination ,Titer ,Immunization ,Immunology ,biology.protein ,Medicine ,Antibody ,business ,education - Abstract
SUMMARYAlthough vaccines effectively prevent COVID-19 in healthy individuals, they appear less immunogenic in individuals with chronic inflammatory diseases (CID) and/or under chronic immunosuppression, and there is uncertainty of their activity against emerging variants of concern in this population. Here, we assessed a cohort of 74 CID patients treated as monotherapy with chronic immunosuppressive drugs for functional antibody responses in serum against historical and variant SARS-CoV-2 viruses after immunization with Pfizer mRNA BNT162b2 vaccine. Longitudinal analysis showed the greatest reductions in neutralizing antibodies and Fc effector function capacity in individuals treated with TNF-α inhibitors, and this pattern appeared worse against the B.1.617.2 Delta virus. Within five months of vaccination, serum neutralizing titers of the majority of CID patients fell below the presumed threshold correlate for antibody-mediated protection. Thus, further vaccine boosting or administration of long-acting prophylaxis (e.g., monoclonal antibodies) likely will be required to prevent SARS-CoV-2 infection in this susceptible population.
- Published
- 2021
13. Intestinal antiviral signaling is controlled by autophagy gene
- Author
-
Sanghyun, Lee, Gowri, Kalugotla, Harshad, Ingle, Rachel, Rodgers, Chunyan, Wu, Yating, Wang, Yuhao, Li, Xia, Yang, Jin, Zhang, Nicolette R, Borella, Hongju, Deng, Lindsay, Droit, Ryan, Hill, Stefan T, Peterson, Chandni, Desai, Dylan, Lawrence, Qun, Lu, and Megan T, Baldridge
- Subjects
Antiviral Restriction Factors ,Intestines ,Mice ,Tumor Necrosis Factor-alpha ,Microbiota ,Autophagy ,Vesicular Transport Proteins ,Animals ,Autophagy-Related Proteins ,Receptors, Interferon - Abstract
Mutations in the macroautophagy/autophagy gene
- Published
- 2021
14. Divergent Enteroviruses from Macaques with Chronic Diarrhea
- Author
-
Lindsay Droit, Anne M. Paredes, Peter J. Didier, Scott A. Handley, Margaret H. Gilbert, David Wang, Rudolf P. Bohm, and Kathie A. Mihindukulasuriya
- Subjects
Immunology and Microbiology (miscellaneous) ,Chronic diarrhea ,biology ,biology.animal ,Family Picornaviridae ,Genome Sequences ,Genetics ,Primate ,Molecular Biology ,Virology ,Genome - Abstract
We report the draft genome sequences of five novel members of the family Picornaviridae that were isolated from the stool of rhesus macaques ( Macaca mulatta ) with chronic diarrhea. The strains were named NOLA-1 through NOLA-5 because the macaques were residents of the Tulane National Primate Research Center.
- Published
- 2021
15. SARS-CoV-2 E Gene Variant Alters Analytical Sensitivity Characteristics of Viral Detection Using a Commercial Reverse Transcription-PCR Assay
- Author
-
Carey-Ann D. Burnham, Meghan A. Wallace, Lindsay Droit, Bijal A. Parikh, David Wang, and Stephen Tahan
- Subjects
0301 basic medicine ,Microbiology (medical) ,030106 microbiology ,Biology ,Real-Time Polymerase Chain Reaction ,medicine.disease_cause ,Sensitivity and Specificity ,Virus ,03 medical and health sciences ,Virology ,medicine ,Humans ,Transversion ,Gene ,Mutation ,Reverse Transcriptase Polymerase Chain Reaction ,SARS-CoV-2 ,COVID-19 ,Reproducibility of Results ,Reverse Transcription ,Reverse transcriptase ,Reverse transcription polymerase chain reaction ,030104 developmental biology ,Real-time polymerase chain reaction ,RNA, Viral ,Primer (molecular biology) - Abstract
Diagnostic assays for detecting severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are essential for patient management, infection prevention, and the public health response for coronavirus disease 2019 (COVID-19). The efficacy and reliability of these assays are of paramount importance in both tracking and controlling the spread of the virus. Real-time reverse transcription-PCR (RT-PCR) assays rely on a fixed genetic sequence for primer and probe binding. Mutations can potentially alter the accuracy of these assays and lead to unpredictable analytical performance characteristics and false-negative results. Here, we identify a G-to-U transversion (nucleotide 26372) in the SARS-CoV-2 E gene in three specimens with reduced viral detection efficiency using a widely available commercial assay. Further analysis of the public GISAID repository led to the identification of 18 additional genomes with this mutation, which reflect five independent mutational events. This work supports the use of dual-target assays to reduce the number of false-negative PCR results.
- Published
- 2021
16. In vivo monoclonal antibody efficacy against SARS-CoV-2 variant strains
- Author
-
Baoling Ying, Pei Yong Shi, Spencer Stumpf, W. Blaine Stine, Charles Y. Chiu, James Brett Case, Davide Corti, Mehul S. Suthar, Sean P. J. Whelan, Laura A. VanBlargan, Lindsay Droit, Raul Andino, Adrianus C. M. Boon, Daved H. Fremont, Seth J. Zost, Miguel Garcia Knight, Zhuoming Liu, Rita E. Chen, Xuping Xie, Traci L. Bricker, Ali H. Ellebedy, John M. Errico, Michael S. Diamond, Lisa A. Purcell, Ishmael D. Aziati, Tamarand L. Darling, David Wang, Pavlo Gilchuk, Meredith E. Davis-Gardner, Scott A. Handley, Swathi Shrihari, James E. Crowe, Emma S. Winkler, and Astha Joshi
- Subjects
Male ,Antibodies, Viral ,Transgenic ,Mice ,Monoclonal ,Chlorocebus aethiops ,antibodies ,Viral ,Neutralizing ,Lung ,Multidisciplinary ,Serine Endopeptidases ,Antibodies, Monoclonal ,Spike Glycoprotein ,Infectious Diseases ,Spike Glycoprotein, Coronavirus ,Pneumonia & Influenza ,Female ,Angiotensin-Converting Enzyme 2 ,Antibody ,Post-Exposure Prophylaxis ,Infection ,Biotechnology ,Genetically modified mouse ,General Science & Technology ,medicine.drug_class ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Mice, Transgenic ,Biology ,Monoclonal antibody ,Antibodies ,Article ,Vaccine Related ,Neutralization Tests ,In vivo ,Biodefense ,medicine ,Animals ,Humans ,Potency ,variations ,Gene ,Vero Cells ,Mesocricetus ,SARS-CoV-2 ,Prevention ,COVID-19 ,Pneumonia ,biology.organism_classification ,Antibodies, Neutralizing ,Virology ,In vitro ,Coronavirus ,Emerging Infectious Diseases ,Cell culture ,Vero cell ,biology.protein ,SARS-CoV-2 variant strains ,Pre-Exposure Prophylaxis ,Immunization - Abstract
Rapidly-emerging variants jeopardize antibody-based countermeasures against SARS-CoV-2. While recent cell culture experiments have demonstrated loss of potency of several anti-spike neutralizing antibodies against SARS-CoV-2 variant strains1-3, the in vivo significance of these results remains uncertain. Here, using a panel of monoclonal antibodies (mAbs) corresponding to many in advanced clinical development by Vir Biotechnology, AbbVie, AstraZeneca, Regeneron, and Lilly we report the impact on protection in animals against authentic SARS-CoV-2 variants including WA1/2020 strains, a B.1.1.7 isolate, and chimeric strains with South African (B.1.351) or Brazilian (B.1.1.28) spike genes. Although some individual mAbs showed reduced or abrogated neutralizing activity against B.1.351 and B.1.1.28 viruses with E484K spike protein mutations in cell culture, low prophylactic doses of mAb combinations protected against infection in K18-hACE2 transgenic mice, 129S2 immunocompetent mice, and hamsters without emergence of resistance. Two exceptions were mAb LY-CoV555 monotherapy which lost all protective activity in vivo, and AbbVie 2B04/47D11, which showed partial loss of activity. When administered after infection as therapy, higher doses of mAb cocktails protected in vivo against viruses displaying a B.1.351 spike gene. Thus, many, but not all, of the antibody products with Emergency Use Authorization should retain substantial efficacy against the prevailing SARS-CoV-2 variant strains.
- Published
- 2021
17. A potently neutralizing anti-SARS-CoV-2 antibody inhibits variants of concern by binding a highly conserved epitope
- Author
-
Emma S. Winkler, Brian O. Smith, Lindsay Droit, Saravanan Raju, Adrian Creanga, Pei Yong Shi, Scott A. Handley, Rita Chen, James E. Crowe, Andrianus Boon, Bradley Whitener, Pavlo Gilchuk, Amarendra Pegu, Daved H. Fremont, Dong-Mei Wang, Zhuoming Liu, Ishmael D. Aziati, Lucas J. Adams, Laura A. VanBlargan, Whelan Spj, Michael P. Diamond, Haiyong Zhao, and James Brett Case
- Subjects
Genetically modified mouse ,biology ,medicine.drug_class ,In vivo ,biology.protein ,medicine ,Antibody ,Receptor ,Monoclonal antibody ,Inhibitory postsynaptic potential ,Virology ,Epitope ,Virus - Abstract
SUMMARYWith the emergence of SARS-CoV-2 variants with increased transmissibility and potential resistance, antibodies and vaccines with broadly inhibitory activity are needed. Here we developed a panel of neutralizing anti-SARS-CoV-2 mAbs that bind the receptor binding domain of the spike protein at distinct epitopes and block virus attachment to cells and its receptor, human angiotensin converting enzyme-2 (hACE2). While several potently neutralizing mAbs protected K18-hACE2 transgenic mice against infection caused by historical SARS-CoV-2 strains, others induced escape variantsin vivoand lost activity against emerging strains. We identified one mAb, SARS2-38, that potently neutralizes all SARS-CoV-2 variants of concern tested and protects mice against challenge by multiple SARS-CoV-2 strains. Structural analysis showed that SARS2-38 engages a conserved epitope proximal to the receptor binding motif. Thus, treatment with or induction of inhibitory antibodies that bind conserved spike epitopes may limit the loss of potency of therapies or vaccines against emerging SARS-CoV-2 variants.
- Published
- 2021
18. Resistance of SARS-CoV-2 variants to neutralization by monoclonal and serum-derived polyclonal antibodies
- Author
-
Rita E, Chen, Xianwen, Zhang, James Brett, Case, Emma S, Winkler, Yang, Liu, Laura A, VanBlargan, Jianying, Liu, John M, Errico, Xuping, Xie, Naveenchandra, Suryadevara, Pavlo, Gilchuk, Seth J, Zost, Stephen, Tahan, Lindsay, Droit, Jackson S, Turner, Wooseob, Kim, Aaron J, Schmitz, Mahima, Thapa, David, Wang, Adrianus C M, Boon, Rachel M, Presti, Jane A, O'Halloran, Alfred H J, Kim, Parakkal, Deepak, Dora, Pinto, Daved H, Fremont, James E, Crowe, Davide, Corti, Herbert W, Virgin, Ali H, Ellebedy, Pei-Yong, Shi, and Michael S, Diamond
- Subjects
South African Spike Gene ,COVID-19 Vaccines ,Chimeric Washington Strain ,SARS-CoV-2 ,Antibodies, Monoclonal ,COVID-19 ,Vaccine Efficacy ,Antibody-based Countermeasures ,Antibodies, Viral ,Antibodies, Neutralizing ,Article ,Mice ,Assay Standardization ,Neutralization Tests ,Pseudoviruses ,Cricetinae ,Chlorocebus aethiops ,Mutation ,Spike Glycoprotein, Coronavirus ,Animals ,Humans ,Vero Cells ,BNT162 Vaccine - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused the global COVID-19 pandemic infecting more than 106 million people and causing 2.3 million deaths. The rapid deployment of antibody-based countermeasures has provided hope for curtailing disease and ending the pandemic1. However, the emergence of rapidly-spreading SARS-CoV-2 variants in the United Kingdom (B.1.1.7), South Africa (B.1.351), and elsewhere with mutations in the spike protein has raised concern for escape from neutralizing antibody responses and loss of vaccine efficacy based on preliminary data with pseudoviruses2–4. Here, using monoclonal antibodies (mAbs), animal immune sera, human convalescent sera, and human sera from recipients of the Pfizer-BioNTech (BNT162b2) mRNA vaccine, we report the impact on antibody neutralization of a panel of authentic SARS-CoV-2 variants including a B.1.1.7 isolate, a chimeric Washington strain with a South African spike gene (Wash SA-B.1.351), and isogenic recombinant variants with designed mutations or deletions at positions 69–70, 417, 484, 501, and/or 614 of the spike protein. Several highly neutralizing mAbs engaging the receptor binding domain (RBD) or N-terminal domain (NTD) lost inhibitory activity against Wash SA-B.1.351 or recombinant variants with an E484K spike mutation. Most convalescent sera and virtually all mRNA vaccine-induced immune sera tested showed markedly diminished neutralizing activity against the Wash SA-B.1.351 strain or recombinant viruses containing mutations at position 484 and 501. We also noted that cell line selection used for growth of virus stocks or neutralization assays can impact the potency of antibodies against different SARS-CoV-2 variants, which has implications for assay standardization and congruence of results across laboratories. As several antibodies binding specific regions of the RBD and NTD show loss-of-neutralization potency in vitro against emerging variants, updated mAb cocktails, targeting of highly conserved regions, enhancement of mAb potency, or adjustments to the spike sequences of vaccines may be needed to prevent loss of protection in vivo.
- Published
- 2021
19. SARS-CoV-2 variants show resistance to neutralization by many monoclonal and serum-derived polyclonal antibodies
- Author
-
Xuping Xie, Xianwen Zhang, Davide Corti, Daved H. Fremont, Pei Yong Shi, Jianying Liu, Laura VanBlargan, Wooseob Kim, Stephen Tahan, James Brett Case, Lindsay Droit, Andrianus Boon, Parakkal Deepak, Herbert W. Virgin, John M. Errico, Michael S. Diamond, Yang Liu, Dora Pinto, James E. Crowe, Alfred H.J. Kim, Ali H. Ellebedy, Emma S. Winkler, Aaron J. Schmitz, Jackson S. Turner, Rita Chen, Mahima Thapa, Jane A. O’Halloran, Rachel M. Presti, Naveenchandra Suryadevara, Pavlo Gilchuk, and David Wang
- Subjects
biology ,medicine.drug_class ,Monoclonal antibody ,Virology ,Assay Standardization ,COVID-19 ,Antibody-based Countermeasures ,Pseudoviruses, Chimeric Washington Strain ,South African Spike Gene ,Vaccine Efficacy ,Article ,Neutralization ,Virus ,law.invention ,Polyclonal antibodies ,law ,Monoclonal ,medicine ,biology.protein ,Recombinant DNA ,Antibody ,Neutralizing antibody - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused the global COVID-19 pandemic infecting more than 106 million people and causing 2.3 million deaths. The rapid deployment of antibody-based countermeasures has provided hope for curtailing disease and ending the pandemic1. However, the emergence of rapidly-spreading SARS-CoV-2 variants in the United Kingdom (B.1.1.7), South Africa (B.1.351), and elsewhere with mutations in the spike protein has raised concern for escape from neutralizing antibody responses and loss of vaccine efficacy based on preliminary data with pseudoviruses2-4. Here, using monoclonal antibodies (mAbs), animal immune sera, human convalescent sera, and human sera from recipients of the Pfizer-BioNTech (BNT162b2) mRNA vaccine, we report the impact on antibody neutralization of a panel of authentic SARS-CoV-2 variants including a B.1.1.7 isolate, a chimeric Washington strain with a South African spike gene (Wash SA-B.1.351), and isogenic recombinant variants with designed mutations or deletions at positions 69-70, 417, 484, 501, and/or 614 of the spike protein. Several highly neutralizing mAbs engaging the receptor binding domain (RBD) or N-terminal domain (NTD) lost inhibitory activity against Wash SA-B.1.351 or recombinant variants with an E484K spike mutation. Most convalescent sera and virtually all mRNA vaccine-induced immune sera tested showed markedly diminished neutralizing activity against the Wash SA-B.1.351 strain or recombinant viruses containing mutations at position 484 and 501. We also noted that cell line selection used for growth of virus stocks or neutralization assays can impact the potency of antibodies against different SARS-CoV-2 variants, which has implications for assay standardization and congruence of results across laboratories. As several antibodies binding specific regions of the RBD and NTD show loss-of-neutralization potency in vitro against emerging variants, updated mAb cocktails, targeting of highly conserved regions, enhancement of mAb potency, or adjustments to the spike sequences of vaccines may be needed to prevent loss of protection in vivo.
- Published
- 2021
20. Intestinal antiviral signaling is controlled by autophagy gene Epg5 independent of the microbiota
- Author
-
Rachel Rodgers, Harshad Ingle, Gowri Kalugotla, Xia Yang, Qun Lu, Chandni Desai, Sanghyun Lee, Nicolette R. Borella, Lindsay Droit, Ryan Hill, Megan T. Baldridge, Stefan T. Peterson, Chunyan Wu, Yuhao Li, Hongju Deng, Dylan Lawrence, Ya-Ting Wang, and Jin Zhang
- Subjects
education.field_of_study ,biology ,Interferon-stimulated gene ,Cell Biology ,Molecular biology ,Sequestosome 1 ,TANK-binding kinase 1 ,Interferon ,biology.protein ,Autophagy Protein 5 ,STAT protein ,medicine ,Interferon gamma ,STAT1 ,education ,Molecular Biology ,medicine.drug - Abstract
Mutations in the macroautophagy/autophagy gene EPG5 are responsible for Vici syndrome, a human genetic disease characterized by combined immunodeficiency. Previously, we found that epg5−/- mice exhibit hyperinflammation in the lungs mediated by IL1B/IL-1β and TNF/TNFα, resulting in resistance to influenza. Here, we find that disruption of Epg5 results in protection against multiple enteric viruses including norovirus and rotavirus. Gene expression analysis reveals IFNL/IFN-λ responsive genes as a key alteration. Further, mice lacking Epg5 exhibit substantial alterations of the intestinal microbiota. Surprisingly, germ-free mouse studies indicate Epg5-associated inflammation of both the intestine and lung is microbiota-independent. Genetic studies support IFNL signaling as the primary mediator of resistance to enteric viruses, but not of microbial dysbiosis, in epg5−/- mice. This study unveils an important role, unexpectedly independent of the microbiota, for autophagy gene Epg5 in host organism protection by modulating intestinal IFNL responses. Abbreviations: CTNNB1: catenin (cadherin associated protein), beta 1; DAPI: 4′,6-diamidino-2-phenylindole; EPG5: ectopic P-granules autophagy protein 5 homolog (C. elegans); FT: fecal transplant; IFI44: interferon-induced protein 44; IFIT1: interferon-induced protein with tetratricopeptide repeats 1; IFNG/IFN-γ: interferon gamma; IFNL/IFN-λ: interferon lambda; IFNLR1: interferon lambda receptor 1; IL1B/IL-1β: interleukin 1 beta; ISG: interferon stimulated gene; GF: germ-free; LEfSe: linear discriminant analysis effect size; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MNoV: murine norovirus; MX2: MX dynamin-like GTPase 2; OAS1A: 2’-5’ oligoadenylate synthetase 1A; RV: rotavirus; SPF: specific-pathogen free; SQSTM1/p62: sequestosome 1; STAT1: signal transducer and activator of transcription 1; STING1: stimulator of interferon response cGAMP interactor 1; TBK1: TANK-binding kinase 1; TNF/TNFα: tumor necrosis factor
- Published
- 2021
- Full Text
- View/download PDF
21. UFMylation inhibits the proinflammatory capacity of interferon-γ-activated macrophages
- Author
-
Bria F. Dunlap, Craig B. Wilen, Christina L. Stallings, Dale R. Balce, Ya-Ting Wang, Lindsay Droit, Anthony Orvedahl, Scott A. Handley, Robert C. Orchard, Herbert W. Virgin, Michael R. McAllaster, Barry L. Hykes, and John G. Doench
- Subjects
0301 basic medicine ,Lipopolysaccharides ,Male ,Cellular immunity ,XBP1 ,Lipopolysaccharide ,Chaperone-Mediated Autophagy ,Endoplasmic Reticulum ,Proinflammatory cytokine ,Cell Line ,03 medical and health sciences ,chemistry.chemical_compound ,Interferon-gamma ,Mice ,0302 clinical medicine ,Mediator ,Immune system ,Interferon ,medicine ,Autophagy ,Animals ,Mice, Knockout ,Multidisciplinary ,Chemistry ,Macrophages ,Proteins ,Macrophage Activation ,Biological Sciences ,Endoplasmic Reticulum Stress ,Cell biology ,Mice, Inbred C57BL ,Protein Transport ,030104 developmental biology ,Tumor necrosis factor alpha ,Female ,030217 neurology & neurosurgery ,medicine.drug ,Protein Binding - Abstract
Macrophages activated with interferon-γ (IFN-γ) in combination with other proinflammatory stimuli, such as lipopolysaccharide or tumor necrosis factor-α (TNF-α), respond with transcriptional and cellular changes that enhance clearance of intracellular pathogens at the risk of damaging tissues. IFN-γ effects must therefore be carefully balanced with inhibitory mechanisms to prevent immunopathology. We performed a genome-wide CRISPR knockout screen in a macrophage cell line to identify negative regulators of IFN-γ responses. We discovered an unexpected role of the ubiquitin-fold modifier (Ufm1) conjugation system (herein UFMylation) in inhibiting responses to IFN-γ and lipopolysaccharide. Enhanced IFN-γ activation in UFMylation-deficient cells resulted in increased transcriptional responses to IFN-γ in a manner dependent on endoplasmic reticulum stress responses involving Ern1 and Xbp1. Furthermore, UFMylation in myeloid cells is required for resistance to influenza infection in mice, indicating that this pathway modulates in vivo responses to infection. These findings provide a genetic roadmap for the regulation of responses to a key mediator of cellular immunity and identify a molecular link between the UFMylation pathway and immune responses.
- Published
- 2020
22. Neutralizing Antibody and Soluble ACE2 Inhibition of a Replication-Competent VSV-SARS-CoV-2 and a Clinical Isolate of SARS-CoV-2
- Author
-
Lindsay Droit, Herbert W. Virgin, Jeffrey P. Henderson, Ma. Xenia G. Ilagan, Stephen Tahan, David Wang, Louis Marie Bloyet, Haiyan Zhao, Davide Corti, Sean P. J. Whelan, Sachdev S. Sidhu, Michael A Tartell, Zhuoming Liu, James Brett Case, Elitza S. Theel, Shane Miersch, Michael S. Diamond, Rita E. Chen, Daved H. Fremont, Siyuan Ding, Arthur S. Kim, Qiru Zeng, Paul W. Rothlauf, Gaya K. Amarasinghe, and Mart Ustav
- Subjects
viruses ,Clone (cell biology) ,Antibodies, Viral ,Virus Replication ,Neutralization ,Biosafety ,0302 clinical medicine ,Biosafety level ,Chlorocebus aethiops ,Medicine ,Neutralizing antibody ,skin and connective tissue diseases ,chemistry.chemical_classification ,0303 health sciences ,biology ,Vesicular stomatitis virus ,Monoclonal ,Spike Glycoprotein, Coronavirus ,Angiotensin-Converting Enzyme 2 ,Antibody ,Coronavirus Infections ,Green Fluorescent Proteins ,Pneumonia, Viral ,Peptidyl-Dipeptidase A ,Microbiology ,Article ,Vesicular stomatitis Indiana virus ,03 medical and health sciences ,Betacoronavirus ,Neutralization Tests ,Virology ,Animals ,Humans ,Pandemics ,Vero Cells ,COVID-19 Serotherapy ,030304 developmental biology ,Host Microbial Interactions ,business.industry ,SARS-CoV-2 ,fungi ,Immunization, Passive ,COVID-19 ,Virus Internalization ,biology.organism_classification ,Antibodies, Neutralizing ,respiratory tract diseases ,body regions ,chemistry ,Polyclonal antibodies ,biology.protein ,Parasitology ,Glycoprotein ,business ,030217 neurology & neurosurgery - Abstract
Antibody-based interventions against SARS-CoV-2 could limit morbidity, mortality, and possibly transmission. An anticipated correlate of such countermeasures is the level of neutralizing antibodies against the SARS-CoV-2 spike protein, which engages with host ACE2 receptor for entry. Using an infectious molecular clone of vesicular stomatitis virus (VSV) expressing eGFP as a marker of infection, we replaced the glycoprotein gene (G) with the spike protein of SARS-CoV-2 (VSV-eGFP-SARS-CoV-2) and developed a high-throughput imaging-based neutralization assay at biosafety level 2. We also developed a focus-reduction neutralization test with a clinical isolate of SARS-CoV-2 at biosafety level 3. Comparing the neutralizing activities of various antibodies and ACE2-Fc soluble decoy protein in both assays revealed a high degree of concordance. These assays will help define correlates of protection for antibody-based countermeasures and vaccines against SARS-CoV-2. Additionally, replication-competent VSV-eGFP-SARS-CoV-2 provides a tool for testing inhibitors of SARS-CoV-2 mediated entry under reduced biosafety containment., Graphical Abstract, Highlights • Vesicular stomatitis virus encoding the SARS-CoV-2 spike replicates to high titers • Virus propagation is enhanced by a truncation in the cytoplasmic tail of the spike • Neutralization can be assessed by BSL2 and BSL3 high-throughput assays • SARS-CoV-2 and VSV-SARS-CoV-2-based neutralization assays correlate, Case, Rothlauf et al. generate a replication-competent vesicular stomatitis virus (VSV) expressing the SARS-CoV-2 spike and compare the neutralizing activity of antibodies with VSV-SARS-CoV-2 to fully infectious SARS-CoV-2. They show that VSV-SARS-CoV-2 is a useful BSL2 surrogate virus, as neutralization profiles strongly correlate with focus-reduction neutralization tests using SARS-CoV-2.
- Published
- 2020
23. Alterations in the oral microbiome in HIV-infected participants after antiretroviral therapy administration are influenced by immune status
- Author
-
Jennifer Webster-Cyriaque, Rachel M. Presti, Caroline H. Shiboski, Mark A. Jacobson, Edgar T. Overton, Todd T. Brown, Scott A. Handley, Mahmoud A. Ghannoum, Michael T. Yin, and Lindsay Droit
- Subjects
Male ,0301 basic medicine ,Saliva ,HIV Infections ,Medical and Health Sciences ,Cohort Studies ,Cluster Analysis ,2.1 Biological and endogenous factors ,Immunology and Allergy ,Aetiology ,Young adult ,Phylogeny ,Microbiota ,Bacterial ,Human microbiome ,human microbiome ,Middle Aged ,Biological Sciences ,Infectious Diseases ,Anti-Retroviral Agents ,HIV/AIDS ,Female ,Oral Microbiome ,Infection ,Sequence Analysis ,Cohort study ,Adult ,16S ,medicine.medical_specialty ,030106 microbiology ,Immunology ,Antiretroviral Therapy ,Young Adult ,03 medical and health sciences ,Acquired immunodeficiency syndrome (AIDS) ,Virology ,Internal medicine ,medicine ,Humans ,Highly Active ,Microbiome ,Dental/Oral and Craniofacial Disease ,Ribosomal ,business.industry ,Psychology and Cognitive Sciences ,HIV ,DNA ,medicine.disease ,CD4 Lymphocyte Count ,Clinical trial ,030104 developmental biology ,anti-HIV agents ,RNA ,business ,Follow-Up Studies - Abstract
ObjectiveTo characterize the oral bacterial microbiome in HIV-infected participants at baseline and after 24 weeks of EFV/FTC/TDF.DesignThirty-five participants co-enrolled in two AIDS Clinical Trials Group (ACTG) studies, A5272 and A5280, with paired saliva samples and complete data sets were assessed.MethodsPaired saliva samples were evaluated for bacterial microbiome using 16S rDNA PCR followed by Illumina sequencing. Diversity and differential abundance was compared between groups. A random forest classification scheme was used to determine the contribution of parameters in classifying participants' CD4+ T-cell count.ResultsBacterial communities demonstrated considerable variability both within participants and between timepoints, although they became more similar after 24 weeks of ART. At baseline, both the number of taxa detected and the average alpha diversity were variable between participants, but did not differ significantly based on CD4+ cell count, viral load or other factors. After 24 weeks of ART samples obtained from participants with persistently low CD4+ T-cell counts had significantly higher bacterial richness and diversity. Several differentially abundant taxa, including Porphyromonas species associated with periodontal disease, were identified, which discriminated between baseline and posttreatment samples. Analysis demonstrated that although inflammatory markers are important in untreated disease, the salivary microbiome may play an important role in CD4+ T-cell count recovery after ART.ConclusionShifts in the oral microbiome after ART initiation are complex, and may play an important role in immune function and inflammatory disease.
- Published
- 2018
24. Reduced antibody activity against SARS-CoV-2 B.1.617.2 delta virus in serum of mRNA-vaccinated individuals receiving tumor necrosis factor-α inhibitors
- Author
-
Lindsay Droit, Michael A. Paley, Rachel M. Presti, Florian Krammer, Jane A. O’Halloran, Rita E. Chen, Michael S. Diamond, Matthew J. Gorman, Dansu Yuan, Juan Manuel Carreño, Alfred H.J. Kim, Douglas A. Lauffenburger, Daniel Y. Zhu, Scott A. Handley, Gregory F. Wu, Wooseob Kim, Parakkal Deepak, Samantha Burdess, Salim Chahin, Galit Alter, Laura A. VanBlargan, Jackson S. Turner, and Ali H. Ellebedy
- Subjects
COVID-19 Vaccines ,Influenza vaccine ,medicine.drug_class ,Antibodies, Viral ,variants of concern ,Monoclonal antibody ,Virus ,TNF inhibitors ,antibody ,medicine ,Humans ,RNA, Messenger ,Neutralizing antibody ,BNT162 Vaccine ,Vaccines, Synthetic ,immunosuppression ,biology ,SARS-CoV-2 ,Tumor Necrosis Factor-alpha ,business.industry ,General Medicine ,neutralization ,Influenza research ,COVID-19 Drug Treatment ,Vaccination ,mRNA vaccine ,Immunization ,Spike Glycoprotein, Coronavirus ,Immunology ,biology.protein ,mRNA Vaccines ,Clinical and Translational Article ,Hepatitis Delta Virus ,Antibody ,business ,Fc effector functions - Abstract
Background Although vaccines effectively prevent coronavirus disease 2019 (COVID-19) in healthy individuals, they appear to be less immunogenic in individuals with chronic inflammatory disease (CID) or receiving chronic immunosuppression therapy. Methods Here we assessed a cohort of 77 individuals with CID treated as monotherapy with chronic immunosuppressive drugs for antibody responses in serum against historical and variant severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viruses after immunization with the BNT162b2 mRNA vaccine. Findings Longitudinal analysis showed the greatest reductions in neutralizing antibodies and Fc effector function capacity in individuals treated with tumor necrosis factor alpha (TNF-α) inhibitors (TNFi), and this pattern appeared to be worse against the B.1.617.2 delta virus. Within 5 months of vaccination, serum neutralizing titers of all TNFi-treated individuals tested fell below the presumed threshold correlate for antibody-mediated protection. However, TNFi-treated individuals receiving a third mRNA vaccine dose boosted their serum neutralizing antibody titers by more than 16-fold. Conclusions Vaccine boosting or administration of long-acting prophylaxis (e.g., monoclonal antibodies) will likely be required to prevent SARS-CoV-2 infection in this susceptible population. Funding This study was supported by grants and contracts from the NIH (R01 AI157155, R01AI151178, and HHSN75N93019C00074; NIAID Centers of Excellence for Influenza Research and Response (CEIRR) contracts HHSN272201400008C and 75N93021C00014; and Collaborative Influenza Vaccine Innovation Centers [CIVIC] contract 75N93019C00051)., Graphical abstract, Context and significance In most individuals, mRNA vaccines effectively prevent severe disease following SARS-CoV-2 infection. However, the protective immunity induced by mRNA vaccines is diminished in immunocompromised individuals, and the effect of variant strains is unexplored. Here we evaluated serum antibody responses in individuals with chronic inflammatory disease after immunization with the Pfizer BNT162b2 mRNA vaccine. The lowest neutralizing antibody titers were observed in individuals treated with TNF-α inhibitors, and this pattern appeared to be worse against the delta virus, with the antibody levels falling below the presumed threshold correlate of protection. Administration of a third vaccine dose substantially boosted serum neutralizing titers. Our data suggest that vaccine boosting is needed to prevent SARS-CoV-2 infection in some immunocompromised populations, especially those receiving TNF-α inhibitor therapies., Chen et al. assess serum antibodies from BNT162b2 mRNA-vaccinated individuals with chronic inflammatory disease receiving single immunosuppressive drug therapies. Individuals receiving TNF-α inhibitors (TNFi) had reduced antibody neutralizing and Fc effector function activity against the B.1.351 and B.1.617.2 variants. A third vaccine dose markedly boosted neutralizing titers in TNFi recipients.
- Published
- 2021
25. Multi-Omics Analyses Show Disease, Diet, and Transcriptome Interactions With the Virome
- Author
-
Kathie A. Mihindukulasuriya, Abigail J. Johnson, Ruben A. T. Mars, Mauro D'Amato, Tonya Ward, Krishna R. Kalari, Tenghao Zheng, Lindsay Droit, Heather Lekatz, Scott A. Handley, Dan Knights, Ran Blekhman, Purna C. Kashyap, Sambhawa Priya, and Gianrico Farrugia
- Subjects
Adult ,Male ,Microviridae ,Context (language use) ,Myoviridae ,Article ,Irritable Bowel Syndrome ,Transcriptome ,Virology ,Humans ,Bacteriophages ,Human virome ,Longitudinal Studies ,Microbiome ,KEGG ,Genetics ,Hepatology ,biology ,Virome ,Gene Expression Profiling ,Gastroenterology ,Middle Aged ,biology.organism_classification ,Diet ,Gastrointestinal Microbiome ,Intestines ,Gene Expression Regulation ,Metagenomics ,Case-Control Studies ,Host-Pathogen Interactions ,Viruses ,Metagenome ,Female - Abstract
Background & Aims The gut virome includes eukaryotic viruses and bacteriophages that can shape the gut bacterial community and elicit host responses. The virome can be implicated in diseases, such as irritable bowel syndrome (IBS), where gut bacteria play an important role in pathogenesis. We provide a comprehensive and longitudinal characterization of the virome, including DNA and RNA viruses and paired multi-omics data in a cohort of healthy subjects and patients with IBS. Methods We selected 2 consecutive stool samples per subject from a longitudinal study cohort and performed metagenomic sequencing on DNA and RNA viruses after enriching for viral-like particles. Viral sequence abundance was evaluated over time, as well as in the context of diet, bacterial composition and function, metabolite levels, colonic gene expression, host genetics, and IBS subsets. Results We found that the gut virome was temporally stable and correlated with the colonic transcriptome. We identified IBS-subset–specific changes in phage populations; Microviridae, Myoviridae, and Podoviridae species were elevated in diarrhea-predominant IBS, and other Microviridae and Myoviridae species were elevated in constipation-predominant IBS compared to healthy controls. We identified correlations between subsets of the virome and bacterial composition (unclassifiable "dark matter" and phages) and diet (eukaryotic viruses). Conclusions We found that the gut virome is stable over time but varies among subsets of patients with IBS. It can be affected by diet and potentially influences host function via interactions with gut bacteria and/or altering host gene expression.
- Published
- 2021
26. A potently neutralizing SARS-CoV-2 antibody inhibits variants of concern by utilizing unique binding residues in a highly conserved epitope
- Author
-
Saravanan Raju, Pavlo Gilchuk, Lindsay Droit, James Brett Case, Haiyan Zhao, James E. Crowe, Adrianus C. M. Boon, Laura A. VanBlargan, Daved H. Fremont, David Wang, Pei Yong Shi, Scott A. Handley, Rita E. Chen, Traci L. Bricker, Sean P. J. Whelan, Amarendra Pegu, Bradley Whitener, Brittany K. Smith, Michael S. Diamond, Astha Joshi, Adrian Creanga, Lucas J. Adams, Zhuoming Liu, Emma S. Winkler, and Ishmael D. Aziati
- Subjects
Genetically modified mouse ,medicine.drug_class ,viruses ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Amino Acid Motifs ,Immunology ,Monoclonal antibody ,Article ,Epitope ,Virus ,Epitopes ,Mice ,Protein Domains ,Neutralization Tests ,medicine ,Animals ,Humans ,Immunology and Allergy ,Potency ,Receptor ,biology ,SARS-CoV-2 ,Antibodies, Monoclonal ,COVID-19 ,Antibodies, Neutralizing ,Virology ,Infectious Diseases ,Spike Glycoprotein, Coronavirus ,biology.protein ,Immunoglobulin Light Chains ,Antibody - Abstract
With the emergence of SARS-CoV-2 variants with increased transmissibility and potential resistance, antibodies and vaccines with broadly inhibitory activity are needed. Here we developed a panel of neutralizing anti-SARS-CoV-2 monoclonal antibodies (mAbs) that bound the receptor binding domain of the spike protein at distinct epitopes and blocked virus attachment to its host receptor, human angiotensin converting enzyme-2 (hACE2). Although several potently neutralizing mAbs protected K18-hACE2 transgenic mice against infection caused by ancestral SARS-CoV-2 strains, others induced escape variants in vivo or lost neutralizing activity against emerging strains. One mAb, SARS2-38, potently neutralized all SARS-CoV-2 variants of concern tested and protected mice against challenge by multiple SARS-CoV-2 strains. Structural analysis showed that SARS2-38 engaged a conserved epitope proximal to the receptor binding motif. Thus, treatment with or induction of neutralizing antibodies that bind conserved spike epitopes may limit the loss of potency of therapies or vaccines against emerging SARS-CoV-2 variants., Graphical Abstract, VanBlargan et al. describe a potently neutralizing mAb, SARS2-38, that recognizes a panel of SARS-CoV-2 variants and confers therapeutic protection in vivo. Structure analysis of SARS2-38 bound to the viral spike protein reveals the basis of its broadly neutralizing activity, highlighting an epitope target for antibody therapeutics and vaccine design.
- Published
- 2021
27. The Intestinal Microbiome Restricts Alphavirus Infection and Dissemination through a Bile Acid-Type I IFN Signaling Axis
- Author
-
Barry L. Hykes, Larissa B. Thackray, Michael S. Diamond, Matthias Mack, Marco Colonna, Amanda Swain, Dana L. Vanlandingham, Scott A. Handley, Emma S. Winkler, Swathi Shrihari, Lindsay Droit, Maxim N. Artyomov, Yan-Jang S. Huang, Prabhakar S. Andhey, Marina Cella, Thaddeus S. Stappenbeck, and Kranthi K. Chebrolu
- Subjects
0303 health sciences ,Deoxycholic acid ,virus diseases ,Viremia ,Plasmacytoid dendritic cell ,Alphavirus ,Biology ,biology.organism_classification ,medicine.disease ,Virology ,General Biochemistry, Genetics and Molecular Biology ,Virus ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,chemistry ,Interferon ,medicine ,Microbiome ,Alphavirus infection ,030217 neurology & neurosurgery ,030304 developmental biology ,medicine.drug - Abstract
Chikungunya virus (CHIKV), an emerging alphavirus, has infected millions of people. However, the factors modulating disease outcome remain poorly understood. Here, we show in germ-free mice or in oral antibiotic-treated conventionally housed mice with depleted intestinal microbiomes that greater CHIKV infection and spread occurs within 1 day of virus inoculation. Alteration of the microbiome alters TLR7-MyD88 signaling in plasmacytoid dendritic cells (pDCs) and blunts systemic production of type I interferon (IFN). Consequently, circulating monocytes express fewer IFN-stimulated genes and become permissive for CHIKV infection. Reconstitution with a single bacterial species, Clostridium scindens, or its derived metabolite, the secondary bile acid deoxycholic acid, can restore pDC- and MyD88-dependent type I IFN responses to restrict systemic CHIKV infection and transmission back to vector mosquitoes. Thus, symbiotic intestinal bacteria modulate antiviral immunity and levels of circulating alphaviruses within hours of infection through a bile acid-pDC-IFN signaling axis, which affects viremia, dissemination, and potentially transmission.
- Published
- 2020
28. A commensal Clostridium species restricts systemic alphavirus dissemination through a type I interferon signaling axis
- Author
-
Emma S Winkler, Larissa B Thackray, Barry L Hykes, Scott A Handley, Lindsay Droit, Prabhakar Andhey, Matthias Mack, Swathi Shrihari, Amanda Swain, Marina Cella, Marco Colonna, Maxim Artyomov, Thaddeus S Stappenbeck, and Michael S Diamond
- Subjects
Immunology ,Immunology and Allergy - Abstract
The role of the gut microbiota in shaping antiviral immune responses at non-intestinal sites has been described in multiple models of viral pathogenesis. Although chikungunya virus (CHIKV), an emerging alphavirus, is a substantial global health threat, the impact of the microbiota in modulating immune responses to alphavirus infection is entirely unexplored. Here, we show that CHIKV infection of oral antibiotic-treated or germ-free mice resulted in increased viremia within one day of infection, and this enhanced viral dissemination. Microbiota depletion resulted in greater CHIKV infection of circulating monocytes, which was linked to dampened systemic type I interferon (IFN) responses. Differences in CHIKV tropism and replication following microbiota depletion depended on MyD88 signaling in plasmacytoid dendritic cells, which contributed to systemic type I IFN production within hours of infection. Colonization of antibiotic-treated mice with a single bacterial microbiota-derived Clostridium species resulted in MyD88-dependent type I IFN responses that restricted CHIKV infection in blood. We propose that select bacterial symbionts impact antiviral immunity to alphaviruses within hours of infection through a type I IFN signaling axis and by virtue of effects on viremia may determine disease severity and affect vector transmission and epidemic spread.
- Published
- 2020
29. Virome biogeography in the lower gastrointestinal tract of rhesus macaques with chronic diarrhea
- Author
-
Anne M. Paredes, Herbert W. Virgin, David Wang, Faith Schiro, Lindsay Droit, Xuemei Si, Margaret H. Gilbert, Rudolf P. Bohm, Peter J. Didier, Scott A. Handley, and Guoyan Zhao
- Subjects
Diarrhea ,Colon ,Rectum ,Lower Gastrointestinal Tract ,Biology ,Virus ,Article ,Microbiology ,03 medical and health sciences ,Contig Mapping ,Feces ,Chronic diarrhea ,Ileum ,Virology ,medicine ,Terminal ileum ,Animals ,Human virome ,Bacteriophages ,030304 developmental biology ,0303 health sciences ,Gastrointestinal tract ,030302 biochemistry & molecular biology ,Primate Diseases ,Biodiversity ,Macaca mulatta ,medicine.anatomical_structure ,Chronic Disease ,Metagenome - Abstract
The composition of gastrointestinal tract viromes has been associated with multiple diseases. Our understanding of virus communities in the GI tract is still very limited due to challenges in sampling from different GI sites. Here we defined the GI viromes of 15 rhesus macaques with chronic diarrhea. Luminal content samples from terminal ileum, proximal and distal colon were collected at necropsy while samples from the rectum were collected antemortem using a fecal loop. The composition of and ecological parameters associated with the terminal ileum virome were distinct from the colon and rectum samples; these differences were driven by bacteriophages rather than eukaryotic viruses. The six contigs that were most discriminative of the viromes were distantly related to bacteriophages from three different families. Our analysis provides support for using fecal loop sampling of the rectum as a proxy of the colonic virome in humans.
- Published
- 2018
30. Early life dynamics of the human gut virome and bacterial microbiome in infants
- Author
-
Irma K. Bauer, Lori R. Holtz, Lindsay Droit, Guoyan Zhao, Barbara B. Warner, I. Malick Ndao, Yanjiao Zhou, David Wang, Phillip I. Tarr, and Efrem S. Lim
- Subjects
medicine.medical_specialty ,biology ,Microbiota ,Infant ,General Medicine ,crAssphage ,biology.organism_classification ,General Biochemistry, Genetics and Molecular Biology ,Early life ,Microbiology ,Cohort Studies ,Intestines ,Immune system ,Medical microbiology ,Human gut ,Microbial ecology ,medicine ,Humans ,Human virome ,Microbiome - Abstract
The early years of life are important for immune development and influence health in adulthood. Although it has been established that the gut bacterial microbiome is rapidly acquired after birth, less is known about the viral microbiome (or 'virome'), consisting of bacteriophages and eukaryotic RNA and DNA viruses, during the first years of life. Here, we characterized the gut virome and bacterial microbiome in a longitudinal cohort of healthy infant twins. The virome and bacterial microbiome were more similar between co-twins than between unrelated infants. From birth to 2 years of age, the eukaryotic virome and the bacterial microbiome expanded, but this was accompanied by a contraction of and shift in the bacteriophage virome composition. The bacteriophage-bacteria relationship begins from birth with a high predator-low prey dynamic, consistent with the Lotka-Volterra prey model. Thus, in contrast to the stable microbiome observed in adults, the infant microbiome is highly dynamic and associated with early life changes in the composition of bacteria, viruses and bacteriophages with age.
- Published
- 2015
31. The Genome of Yoka Poxvirus
- Author
-
Lindsay Droit, Guoyan Zhao, Herbert W. Virgin, Vsevolod L. Popov, Chris Upton, Robert B. Tesh, Nicole S. Little, and David Wang
- Subjects
food.ingredient ,viruses ,Molecular Sequence Data ,Immunology ,Genome, Viral ,Orthopoxvirus ,Suipoxvirus ,complex mixtures ,Microbiology ,Capripoxvirus ,chemistry.chemical_compound ,food ,Virology ,Animals ,Cluster Analysis ,Poxviridae ,Phylogeny ,Yatapoxvirus ,Genetics ,biology ,Structure and Assembly ,virus diseases ,Sequence Analysis, DNA ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Central African Republic ,Culicidae ,chemistry ,Insect Science ,DNA, Viral ,Parapoxvirus ,Vaccinia ,Cervidpoxvirus - Abstract
Yoka poxvirus was isolated almost four decades ago from a mosquito pool in the Central African Republic. Its classification as a poxvirus is based solely upon the morphology of virions visualized by electron microscopy. Here we describe sequencing of the Yoka poxvirus genome using a combination of Roche/454 and Illumina next-generation sequencing technologies. A single consensus contig of 175 kb in length that encodes 186 predicted genes was generated. Multiple methods were used to show that Yoka poxvirus is most closely related to viruses in the Orthopoxvirus genus, but it is clearly distinct from previously described poxviruses. Collectively, the phylogenetic and genomic sequence analyses suggest that Yoka poxvirus is the prototype member of a new genus in the family Poxviridae. Poxviruses are currently classified into two subfamilies, the Chordopoxvirinae (ChPV) and the Entomopoxivirinae. The ChPV infect vertebrates, and the Entomopoxivirinae infect insects. Within the ChPV subfamily, there are nine genera currently recognized, with classification based initially on morphological and biological characteristics: Avipoxvirus, Capripoxvirus, Cervidpoxvirus, Leporipoxvirus, Molluscipoxvirus, Orthopoxvirus, Parapoxvirus, Suipoxvirus, and Yatapoxvirus. More recently, comparative genome analysis has confirmed this poxvirus genus classification (10, 15). The ChPV genomes are composed of linear double-stranded DNA, vary in size from 134 (6) to 365 kb (22), and contain between 130 and 328 open reading frames (ORFs). It has been shown that the ends of the vaccinia virus genome contain covalently closed hairpin loops, indicating that the linear doublestranded DNA molecule consists of a single, continuous polynucleotide chain (4). Typically, terminal inverted repeats are located at the termini of poxvirus genomes. The relatively conserved central region of the ChPV genomes contains essential genes with roles in transcription, replication, and virion assembly and ranges in size from about 80 to 100 kb. The variable genes in poxviruses are located predominantly at either end of the genomes. These genes include some with host range restrictions and immune subversion functions (17), although the functions of many genes in these regions of the poxvirus genomes are not known. Complete genomic sequences of representative viruses from all nine of the ChPV genera have been obtained. Yoka poxvirus is an unclassified poxvirus that was originally isolated from a pool of mosquitoes (Aedes simpsoni) collected in the Central African Republic in 1972 (25). Yoka poxvirus causes the cytopathic effect (CPE) in Vero, CER, PS, and BHK-21 cells but does not form plaques; suckling mice die within 6 days after intracranial inoculation. Virions of poxvirus shape can be observed in the cytoplasm, and the virus does not react serologically with antibodies to variola or vaccinia viruses (25). Based on electron microscopy, Yoka poxvirus was placed in the Poxviridae family, and it was suggested to be either an avipox, orthopox, leporipox, goatpox, or sheeppox virus but not a tanapox or a parapox virus (25). Here we report the genome sequence of Yoka poxvirus and provide data suggesting that Yoka poxvirus is the prototype member of a new genus within the family Poxviridae.
- Published
- 2011
32. Analysis of the Airways Virome after Lung Transplantation and Its Association with Clinical Outcomes
- Author
-
Herbert W. Virgin, Derek E. Byers, Brian C. Keller, Lindsay Droit, Ramsey R. Hachem, Guoyan Zhao, Scott A. Handley, and Chandni Desai
- Subjects
Pulmonary and Respiratory Medicine ,Transplantation ,medicine.medical_specialty ,Adverse outcomes ,business.industry ,medicine.medical_treatment ,Gastroenterology ,Macrolide resistance ,Internal medicine ,Cohort ,medicine ,Lung transplantation ,Surgery ,Human virome ,In patient ,Favorable outcome ,Cardiology and Cardiovascular Medicine ,business ,Clearance - Abstract
s S135 patients had a favorable outcome after prolonged therapy. 2/6 had positive cultures at the time of the transplant (one despite therapy, one diagnosed at transplant) and both developed early recurrences and died within the first year of transplant. 20 patients developed de novo infections at a median of 15.5 months (0.5-132 months) post LTx. 10/20 did not receive treatment, despite the fact that 7 out of these 10 had some form of radiological abnormalities. 7/10 untreated patients cleared the infection spontaneously. None of the untreated cohort had deaths attributable to MAb infection. Out of the 10 patients who received MAb treatment, 7 died at a median of 41 months (12-83 months) after LTx, with 3 deaths being attributable to MAb infection. There was no relationship found between deaths/attributable deaths and macrolide resistance or rejection therapy. Conclusion: In patients who are infected with MAb pre-LTx, those with positive smear or culture at the time of transplant had poorer outcomes compared to those who had documented negative cultures. Half of the de novo Mab infections post-LTx were not treated without any MAb-attributable adverse outcomes.
- Published
- 2016
33. Gammaherpesvirus gene expression and DNA synthesis are facilitated by viral protein kinase and histone variant H2AX
- Author
-
Bryan C. Mounce, Sarah Kohler, Lindsay Droit, Fei Chin Tsan, Justin M. Reitsma, Lisa Ann Cirillo, and Vera L. Tarakanova
- Subjects
DNA Replication ,Gene Expression Regulation, Viral ,Chromatin Immunoprecipitation ,Rhadinovirus ,DNA Repair ,Transcription, Genetic ,DNA repair ,DNA damage ,viruses ,Eukaryotic DNA replication ,Biology ,DNA damage response ,Immediate early protein ,Gammaherpesvirus ,Article ,Immediate-Early Proteins ,Histones ,03 medical and health sciences ,Mice ,Viral Proteins ,Virology ,Animals ,H2AX ,030304 developmental biology ,Regulation of gene expression ,Mice, Knockout ,0303 health sciences ,DNA synthesis ,Macrophages ,030302 biochemistry & molecular biology ,DNA replication ,biochemical phenomena, metabolism, and nutrition ,Molecular biology ,3. Good health ,Mice, Inbred C57BL ,Lytic cycle ,DNA, Viral ,Protein Kinases ,Gammaherpesvirus protein kinase - Abstract
Gammaherpesvirus protein kinases are an attractive therapeutic target as they support lytic replication and latency. Via an unknown mechanism these kinases enhance expression of select viral genes and DNA synthesis. Importantly, the kinase phenotypes have not been examined in primary cell types. Mouse gammaherpesvirus-68 (MHV68) protein kinase orf36 activates the DNA damage response (DDR) and facilitates lytic replication in primary macrophages. Significantly, H2AX, a DDR component and putative orf36 substrate, enhances MHV68 replication. Here we report that orf36 facilitated expression of RTA, an immediate early MHV68 gene, and DNA synthesis during de novo infection of primary macrophages. H2AX expression supported efficient RTA transcription and phosphorylated H2AX associated with RTA promoter. Furthermore, viral DNA synthesis was attenuated in H2AX-deficient macrophages, suggesting that the DDR system was exploited throughout the replication cycle. The interactions between a cancer-associated gammaherpesvirus and host tumor suppressor system have important implications for the pathogenesis of gammaherpesvirus infection.
- Published
- 2011
34. Detection of novel sequences related to african Swine Fever virus in human serum and sewage
- Author
-
James M. Pipas, Stacy R. Finkbeiner, Rachel M. Presti, Herbert W. Virgin, Guoyan Zhao, Lori R. Holtz, Lindsay Droit, Joy Loh, Zoilmar Villasana, Rosina Girones, David Q.-H. Wang, Byron Calgua, and Collin Todd
- Subjects
Serum ,Genetic diversity ,biology ,Sewage ,business.industry ,Immunology ,Molecular Sequence Data ,Outbreak ,biology.organism_classification ,Microbiology ,African swine fever virus ,Virology ,African Swine Fever Virus ,Virus ,Blood serum ,Genetic Diversity and Evolution ,Phylogenetics ,Insect Science ,Humans ,Amino Acid Sequence ,business ,Virus classification ,Phylogeny - Abstract
The family Asfarviridae contains only a single virus species, African swine fever virus (ASFV). ASFV is a viral agent with significant economic impact due to its devastating effects on populations of domesticated pigs during outbreaks but has not been reported to infect humans. We report here the discovery of novel viral sequences in human serum and sewage which are clearly related to the asfarvirus family but highly divergent from ASFV. Detection of these sequences suggests that greater genetic diversity may exist among asfarviruses than previously thought and raises the possibility that human infection by asfarviruses may occur.
- Published
- 2009
35. S22 Analysis of the lung microbiome in human asthma using whole genome shot-gun metagenomics
- Author
-
Lindsay Droit, Caroline Smith, Ratko Djukanovic, Karl J. Staples, Scott A. Handley, Herbert W. Virgin, Tsc Hinks, Stephan D. Gadola, Brian C. Keller, and P H Howarth
- Subjects
Pulmonary and Respiratory Medicine ,COPD ,Lung microbiome ,Bronchiectasis ,medicine.diagnostic_test ,business.industry ,medicine.disease ,respiratory tract diseases ,Microbiology ,Bronchoalveolar lavage ,Metagenomics ,Immunology ,medicine ,Sputum ,Microbiome ,medicine.symptom ,business ,Asthma - Abstract
Introduction and Objectives Evidence is accumulating for the presence of bacteria in the airways and the existence of a commensal airway microbiome. Molecular techniques reveal complex microbial assemblages in bronchiectasis and COPD, but little is known about the airway microbiome in asthma or in health. Immunopathology in asthma may be driven by bacteria or fungi or even by chronic viral persistence. We undertook an analysis of the entire respiratory microbial metagenome in airway samples from carefully phenotyped subjects across a spectrum of asthma and health. Methods 55 subjects (9 mild asthmatics, 16 moderate asthmatics, 15 with severe neutrophilic asthma without bronchiectasis and 15 healthy controls) underwent detailed clinical and immunological phenotyping, sputum induction, and bronchoscopy during periods of clinical stability. Protected bronchoalveolar lavage (BAL) and induced sputum were analysed by whole genome shot-gun sequencing for RNA and DNA from bacterial, viral and fungal genomes. Data were analysed using the VirusHunter analysis pipeline. Results We found no evidence of novel viral species or of persistent viral infection. Protected BAL samples typically contained 500–5000 bacterial reads for organisms typical of the oral cavity or upper respiratory tract, consistent with microaspiration rather than a distinct airway microbiome. Bacterial abundance was not increased in asthma. Hierarchical cluster analysis revealed no general association between disease and the presence of bacteria, with the exception of two individuals with severe neutrophilic asthma in whom single pathogenic species were detected with high abundance. In the first subject, chronic Haemophilus influenzae infection was present, correlating with standard culture and a striking Th17 cell response in BAL. Specific antibiotic treatment resulted in a dramatic clinical improvement. In the second individual Tropheryma whipplei was present, and correlated with the presence of foamy macrophages and a deficiency of BAL Th17 cells. Conclusions : The data from this study argue against the existence of a distinct airway microbiome in health or in asthma, but rather that lung microbes are a result of microaspiration. Conversely, in specific cases chronic low grade infection may drive immunopathology in asthma. It remains to be seen whether very severe forms of asthma have a more prominent microbiome.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.