208 results on '"Finberg RW"'
Search Results
2. Liposomal amphotericin B for empirical therapy in patients with persistent fever and neutropenia.
- Author
-
Walsh TJ, Finberg RW, Arndt C, Hiemenz J, Schwartz C, Bodensteiner D, Pappas P, Seibel N, Greenberg RN, Dummer S, Schuster M, Holcenberg JS, and National Institute of Allergy and Infectious Diseases Mycoses Study Group
- Published
- 1999
3. Sources and sequelae of bacterial contamination of hematopoietic stem cell components: implications for the safety of hematotherapy and graft engineering.
- Author
-
Webb IJ, Coral FS, Andersen JW, Elias AD, Finberg RW, Nadler LM, Ritz J, Anderson KC, Webb, I J, Coral, F S, Andersen, J W, Elias, A D, Finberg, R W, Nadler, L M, Ritz, J, and Anderson, K C
- Published
- 1996
4. ADAM9 promotes type I interferon-mediated innate immunity during encephalomyocarditis virus infection.
- Author
-
Bazzone LE, Zhu J, King M, Liu G, Guo Z, MacKay CR, Kyawe PP, Qaisar N, Rojas-Quintero J, Owen CA, Brass AL, McDougall W, Baer CE, Cashman T, Trivedi CM, Gack MU, Finberg RW, and Kurt-Jones EA
- Subjects
- Animals, Mice, Adaptor Proteins, Signal Transducing metabolism, Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing immunology, HEK293 Cells, Mice, Inbred C57BL, Mice, Knockout, Signal Transduction immunology, ADAM Proteins metabolism, ADAM Proteins genetics, ADAM Proteins immunology, Cardiovirus Infections immunology, Cardiovirus Infections virology, Encephalomyocarditis virus immunology, Immunity, Innate, Interferon Type I metabolism, Interferon Type I immunology, Interferon-Induced Helicase, IFIH1 metabolism, Interferon-Induced Helicase, IFIH1 genetics, Interferon-Induced Helicase, IFIH1 immunology, Membrane Proteins metabolism, Membrane Proteins genetics, Membrane Proteins immunology, Myocarditis immunology, Myocarditis virology
- Abstract
Viral myocarditis, an inflammatory disease of the heart, causes significant morbidity and mortality. Type I interferon (IFN)-mediated antiviral responses protect against myocarditis, but the mechanisms are poorly understood. We previously identified A Disintegrin And Metalloproteinase domain 9 (ADAM9) as an important factor in viral pathogenesis. ADAM9 is implicated in a range of human diseases, including inflammatory diseases; however, its role in viral infection is unknown. Here, we demonstrate that mice lacking ADAM9 are more susceptible to encephalomyocarditis virus (EMCV)-induced death and fail to mount a characteristic type I IFN response. This defect in type I IFN induction is specific to positive-sense, single-stranded RNA (+ ssRNA) viruses and involves melanoma differentiation-associated protein 5 (MDA5)-a key receptor for +ssRNA viruses. Mechanistically, ADAM9 binds to MDA5 and promotes its oligomerization and thereby downstream mitochondrial antiviral-signaling protein (MAVS) activation in response to EMCV RNA stimulation. Our findings identify a role for ADAM9 in the innate antiviral response, specifically MDA5-mediated IFN production, which protects against virus-induced cardiac damage, and provide a potential therapeutic target for treatment of viral myocarditis., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
5. Divalent siRNAs are bioavailable in the lung and efficiently block SARS-CoV-2 infection.
- Author
-
Hariharan VN, Shin M, Chang CW, O'Reilly D, Biscans A, Yamada K, Guo Z, Somasundaran M, Tang Q, Monopoli K, Krishnamurthy PM, Devi G, McHugh N, Cooper DA, Echeverria D, Cruz J, Chan IL, Liu P, Lim SY, McConnell J, Singh SP, Hildebrand S, Sousa J, Davis SM, Kennedy Z, Ferguson C, Godinho BMDC, Thillier Y, Caiazzi J, Ly S, Muhuri M, Kelly K, Humphries F, Cousineau A, Parsi KM, Li Q, Wang Y, Maehr R, Gao G, Korkin D, McDougall WM, Finberg RW, Fitzgerald KA, Wang JP, Watts JK, and Khvorova A
- Subjects
- Humans, Animals, Mice, RNA, Small Interfering genetics, SARS-CoV-2 genetics, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Oligonucleotides, Lung, COVID-19 therapy
- Abstract
The continuous evolution of SARS-CoV-2 variants complicates efforts to combat the ongoing pandemic, underscoring the need for a dynamic platform for the rapid development of pan-viral variant therapeutics. Oligonucleotide therapeutics are enhancing the treatment of numerous diseases with unprecedented potency, duration of effect, and safety. Through the systematic screening of hundreds of oligonucleotide sequences, we identified fully chemically stabilized siRNAs and ASOs that target regions of the SARS-CoV-2 genome conserved in all variants of concern, including delta and omicron. We successively evaluated candidates in cellular reporter assays, followed by viral inhibition in cell culture, with eventual testing of leads for in vivo antiviral activity in the lung. Previous attempts to deliver therapeutic oligonucleotides to the lung have met with only modest success. Here, we report the development of a platform for identifying and generating potent, chemically modified multimeric siRNAs bioavailable in the lung after local intranasal and intratracheal delivery. The optimized divalent siRNAs showed robust antiviral activity in human cells and mouse models of SARS-CoV-2 infection and represent a new paradigm for antiviral therapeutic development for current and future pandemics.
- Published
- 2023
- Full Text
- View/download PDF
6. Anti-SARS-CoV-2 Activity of Adamantanes In Vitro and in Animal Models of Infection.
- Author
-
Lim SY, Guo Z, Liu P, McKay LGA, Storm N, Griffiths A, Qu MD, Finberg RW, Somasundaran M, and Wang JP
- Abstract
Coronavirus disease 2019 (COVID-19) has had devastating effects worldwide, with particularly high morbidity and mortality in outbreaks on residential care facilities. Amantadine, originally licensed as an antiviral agent for therapy and prophylaxis against influenza A virus, has beneficial effects on patients with Parkinson's disease and is used for treatment of Parkinson's disease, multiple sclerosis, acquired brain injury, and various other neurological disorders. Recent observational data suggest an inverse relationship between the use of amantadine and COVID-19. Adamantanes, including amantadine and rimantadine, are reported to have in vitro activity against severe acute respiratory syndrome coronavirus (SARS-CoV) and, more recently, SARS-CoV-2. We hypothesized that adamantanes have antiviral activity against SARS-CoV-2, including variant strains. To assess the activity of adamantanes against SARS-CoV-2, we used in vitro and in vivo models of infection. We established that amantadine, rimantadine, and tromantadine inhibit the growth of SARS-CoV-2 in vitro in cultured human epithelial cells. While neither rimantadine nor amantadine reduces lung viral titers in mice infected with mouse-adapted SARS-CoV-2, rimantadine significantly reduces viral titers in the lungs in golden Syrian hamsters infected with SARS-CoV-2. In summary, rimantadine has antiviral activity against SARS-CoV-2 in human alveolar epithelial cells and in the hamster model of SARS-CoV-2 lung infection. The evaluation of amantadine or rimantadine in human randomized controlled trials can definitively address applications for the treatment or prevention of COVID-19., Competing Interests: Conflicts of Interest: The authors declare no conflict of interest.
- Published
- 2022
- Full Text
- View/download PDF
7. Baricitinib versus dexamethasone for adults hospitalised with COVID-19 (ACTT-4): a randomised, double-blind, double placebo-controlled trial.
- Author
-
Wolfe CR, Tomashek KM, Patterson TF, Gomez CA, Marconi VC, Jain MK, Yang OO, Paules CI, Palacios GMR, Grossberg R, Harkins MS, Mularski RA, Erdmann N, Sandkovsky U, Almasri E, Pineda JR, Dretler AW, de Castilla DL, Branche AR, Park PK, Mehta AK, Short WR, McLellan SLF, Kline S, Iovine NM, El Sahly HM, Doernberg SB, Oh MD, Huprikar N, Hohmann E, Kelley CF, Holodniy M, Kim ES, Sweeney DA, Finberg RW, Grimes KA, Maves RC, Ko ER, Engemann JJ, Taylor BS, Ponce PO, Larson L, Melendez DP, Seibert AM, Rouphael NG, Strebe J, Clark JL, Julian KG, de Leon AP, Cardoso A, de Bono S, Atmar RL, Ganesan A, Ferreira JL, Green M, Makowski M, Bonnett T, Beresnev T, Ghazaryan V, Dempsey W, Nayak SU, Dodd LE, Beigel JH, and Kalil AC
- Subjects
- Adolescent, Adult, Azetidines, Dexamethasone, Double-Blind Method, Female, Humans, Male, Middle Aged, Oxygen, Purines, Pyrazoles, SARS-CoV-2, Sulfonamides, Treatment Outcome, COVID-19 Drug Treatment
- Abstract
Background: Baricitinib and dexamethasone have randomised trials supporting their use for the treatment of patients with COVID-19. We assessed the combination of baricitinib plus remdesivir versus dexamethasone plus remdesivir in preventing progression to mechanical ventilation or death in hospitalised patients with COVID-19., Methods: In this randomised, double-blind, double placebo-controlled trial, patients were enrolled at 67 trial sites in the USA (60 sites), South Korea (two sites), Mexico (two sites), Singapore (two sites), and Japan (one site). Hospitalised adults (≥18 years) with COVID-19 who required supplemental oxygen administered by low-flow (≤15 L/min), high-flow (>15 L/min), or non-invasive mechanical ventilation modalities who met the study eligibility criteria (male or non-pregnant female adults ≥18 years old with laboratory-confirmed SARS-CoV-2 infection) were enrolled in the study. Patients were randomly assigned (1:1) to receive either baricitinib, remdesivir, and placebo, or dexamethasone, remdesivir, and placebo using a permuted block design. Randomisation was stratified by study site and baseline ordinal score at enrolment. All patients received remdesivir (≤10 days) and either baricitinib (or matching oral placebo) for a maximum of 14 days or dexamethasone (or matching intravenous placebo) for a maximum of 10 days. The primary outcome was the difference in mechanical ventilation-free survival by day 29 between the two treatment groups in the modified intention-to-treat population. Safety analyses were done in the as-treated population, comprising all participants who received one dose of the study drug. The trial is registered with ClinicalTrials.gov, NCT04640168., Findings: Between Dec 1, 2020, and April 13, 2021, 1047 patients were assessed for eligibility. 1010 patients were enrolled and randomly assigned, 516 (51%) to baricitinib plus remdesivir plus placebo and 494 (49%) to dexamethasone plus remdesivir plus placebo. The mean age of the patients was 58·3 years (SD 14·0) and 590 (58%) of 1010 patients were male. 588 (58%) of 1010 patients were White, 188 (19%) were Black, 70 (7%) were Asian, and 18 (2%) were American Indian or Alaska Native. 347 (34%) of 1010 patients were Hispanic or Latino. Mechanical ventilation-free survival by day 29 was similar between the study groups (Kaplan-Meier estimates of 87·0% [95% CI 83·7 to 89·6] in the baricitinib plus remdesivir plus placebo group and 87·6% [84·2 to 90·3] in the dexamethasone plus remdesivir plus placebo group; risk difference 0·6 [95% CI -3·6 to 4·8]; p=0·91). The odds ratio for improved status in the dexamethasone plus remdesivir plus placebo group compared with the baricitinib plus remdesivir plus placebo group was 1·01 (95% CI 0·80 to 1·27). At least one adverse event occurred in 149 (30%) of 503 patients in the baricitinib plus remdesivir plus placebo group and 179 (37%) of 482 patients in the dexamethasone plus remdesivir plus placebo group (risk difference 7·5% [1·6 to 13·3]; p=0·014). 21 (4%) of 503 patients in the baricitinib plus remdesivir plus placebo group had at least one treatment-related adverse event versus 49 (10%) of 482 patients in the dexamethasone plus remdesivir plus placebo group (risk difference 6·0% [2·8 to 9·3]; p=0·00041). Severe or life-threatening grade 3 or 4 adverse events occurred in 143 (28%) of 503 patients in the baricitinib plus remdesivir plus placebo group and 174 (36%) of 482 patients in the dexamethasone plus remdesivir plus placebo group (risk difference 7·7% [1·8 to 13·4]; p=0·012)., Interpretation: In hospitalised patients with COVID-19 requiring supplemental oxygen by low-flow, high-flow, or non-invasive ventilation, baricitinib plus remdesivir and dexamethasone plus remdesivir resulted in similar mechanical ventilation-free survival by day 29, but dexamethasone was associated with significantly more adverse events, treatment-related adverse events, and severe or life-threatening adverse events. A more individually tailored choice of immunomodulation now appears possible, where side-effect profile, ease of administration, cost, and patient comorbidities can all be considered., Funding: National Institute of Allergy and Infectious Diseases., Competing Interests: Declaration of interests We declare no competing interests., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
8. A Newly Engineered A549 Cell Line Expressing ACE2 and TMPRSS2 Is Highly Permissive to SARS-CoV-2, Including the Delta and Omicron Variants.
- Author
-
Chang CW, Parsi KM, Somasundaran M, Vanderleeden E, Liu P, Cruz J, Cousineau A, Finberg RW, and Kurt-Jones EA
- Subjects
- A549 Cells, Antiviral Agents pharmacology, Humans, Peptidyl-Dipeptidase A metabolism, SARS-CoV-2 genetics, Serine Endopeptidases genetics, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus metabolism, Angiotensin-Converting Enzyme 2 genetics, COVID-19
- Abstract
New variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continue to emerge, causing surges, breakthrough infections, and devastating losses-underscoring the importance of identifying SARS-CoV-2 antivirals. A simple, accessible human cell culture model permissive to SARS-CoV-2 variants is critical for identifying and assessing antivirals in a high-throughput manner. Although human alveolar A549 cells are a valuable model for studying respiratory virus infections, they lack two essential host factors for SARS-CoV-2 infection: angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2). SARS-CoV-2 uses the ACE2 receptor for viral entry and TMPRSS2 to prime the SARS-CoV-2 spike protein, both of which are negligibly expressed in A549 cells. Here, we report the generation of a suitable human cell line for SARS-CoV-2 studies by transducing human ACE2 and TMPRSS2 into A549 cells. We show that subclones highly expressing ACE2 and TMPRSS2 ("ACE2plus" and the subclone "ACE2plusC3") are susceptible to infection with SARS-CoV-2, including the delta and omicron variants. These subclones express more ACE2 and TMPRSS2 transcripts than existing commercial A549 cells engineered to express ACE2 and TMPRSS2. Additionally, the antiviral drugs EIDD-1931, remdesivir, nirmatrelvir, and nelfinavir strongly inhibit SARS-CoV-2 variants in our infection model. Our data show that ACE2plusC3 cells are highly permissive to SARS-CoV-2 infection and can be used to identify anti-SARS-CoV-2 drugs.
- Published
- 2022
- Full Text
- View/download PDF
9. Broadly recognized, cross-reactive SARS-CoV-2 CD4 T cell epitopes are highly conserved across human coronaviruses and presented by common HLA alleles.
- Author
-
Becerra-Artiles A, Calvo-Calle JM, Co MD, Nanaware PP, Cruz J, Weaver GC, Lu L, Forconi C, Finberg RW, Moormann AM, and Stern LJ
- Subjects
- Alleles, CD4-Positive T-Lymphocytes, COVID-19 Vaccines, Epitopes, T-Lymphocyte, HLA Antigens, Humans, Receptors, Antigen, T-Cell, mRNA Vaccines, COVID-19, SARS-CoV-2
- Abstract
Sequence homology between SARS-CoV-2 and common-cold human coronaviruses (HCoVs) raises the possibility that memory responses to prior HCoV infection can affect T cell response in COVID-19. We studied T cell responses to SARS-CoV-2 and HCoVs in convalescent COVID-19 donors and identified a highly conserved SARS-CoV-2 sequence, S
811-831 , with overlapping epitopes presented by common MHC class II proteins HLA-DQ5 and HLA-DP4. These epitopes are recognized by low-abundance CD4 T cells from convalescent COVID-19 donors, mRNA vaccine recipients, and uninfected donors. TCR sequencing revealed a diverse repertoire with public TCRs. T cell cross-reactivity is driven by the high conservation across human and animal coronaviruses of T cell contact residues in both HLA-DQ5 and HLA-DP4 binding frames, with distinct patterns of HCoV cross-reactivity explained by MHC class II binding preferences and substitutions at secondary TCR contact sites. These data highlight S811-831 as a highly conserved CD4 T cell epitope broadly recognized across human populations., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
10. Quantitative structural analysis of influenza virus by cryo-electron tomography and convolutional neural networks.
- Author
-
Huang QJ, Song K, Xu C, Bolon DNA, Wang JP, Finberg RW, Schiffer CA, and Somasundaran M
- Subjects
- Cryoelectron Microscopy methods, Electron Microscope Tomography methods, Hemagglutinin Glycoproteins, Influenza Virus, Humans, Neural Networks, Computer, Influenza A Virus, H1N1 Subtype, Influenza, Human
- Abstract
Influenza viruses pose severe public health threats globally. Influenza viruses are extensively pleomorphic, in shape, size, and organization of viral proteins. Analysis of influenza morphology and ultrastructure can help elucidate viral structure-function relationships and aid in therapeutics and vaccine development. While cryo-electron tomography (cryoET) can depict the 3D organization of pleomorphic influenza, the low signal-to-noise ratio inherent to cryoET and viral heterogeneity have precluded detailed characterization of influenza viruses. In this report, we leveraged convolutional neural networks and cryoET to characterize the morphological architecture of the A/Puerto Rico/8/34 (H1N1) influenza strain. Our pipeline improved the throughput of cryoET analysis and accurately identified viral components within tomograms. Using this approach, we successfully characterized influenza morphology, glycoprotein density, and conducted subtomogram averaging of influenza glycoproteins. Application of this processing pipeline can aid in the structural characterization of not only influenza viruses, but other pleomorphic viruses and infected cells., Competing Interests: Declaration of interests C.A.S. is on the Scientific Advisory Board of Gandeeva Therapeutics., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
11. Identification of a Permissive Secondary Mutation That Restores the Enzymatic Activity of Oseltamivir Resistance Mutation H275Y.
- Author
-
Jiang L, Samant N, Liu P, Somasundaran M, Jensen JD, Marasco WA, Kowalik TF, Schiffer CA, Finberg RW, Wang JP, and Bolon DNA
- Subjects
- Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Humans, Influenza A virus drug effects, Influenza A virus enzymology, Influenza A virus genetics, Influenza, Human drug therapy, Mutation, Neuraminidase genetics, Neuraminidase metabolism, Drug Resistance, Viral genetics, Influenza A Virus, H1N1 Subtype drug effects, Influenza A Virus, H1N1 Subtype enzymology, Influenza A Virus, H1N1 Subtype genetics, Oseltamivir pharmacology, Viral Proteins genetics, Viral Proteins metabolism
- Abstract
Many oseltamivir resistance mutations exhibit fitness defects in the absence of drug pressure that hinders their propagation in hosts. Secondary permissive mutations can rescue fitness defects and facilitate the segregation of resistance mutations in viral populations. Previous studies have identified a panel of permissive or compensatory mutations in neuraminidase (NA) that restore the growth defect of the predominant oseltamivir resistance mutation (H275Y) in H1N1 influenza A virus. In prior work, we identified a hyperactive mutation (Y276F) that increased NA activity by approximately 70%. While Y276F had not been previously identified as a permissive mutation, we hypothesized that Y276F may counteract the defects caused by H275Y by buffering its reduced NA expression and enzyme activity. In this study, we measured the relative fitness, NA activity, and surface expression, as well as sensitivity to oseltamivir, for several oseltamivir resistance mutations, including H275Y in the wild-type and Y276F genetic background. Our results demonstrate that Y276F selectively rescues the fitness defect of H275Y by restoring its NA surface expression and enzymatic activity, elucidating the local compensatory structural impacts of Y276F on the adjacent H275Y. IMPORTANCE The potential for influenza A virus (IAV) to cause pandemics makes understanding evolutionary mechanisms that impact drug resistance critical for developing surveillance and treatment strategies. Oseltamivir is the most widely used therapeutic strategy to treat IAV infections, but mutations in IAV can lead to drug resistance. The main oseltamivir resistance mutation, H275Y, occurs in the neuraminidase (NA) protein of IAV and reduces drug binding as well as NA function. Here, we identified a new helper mutation, Y276F, that can rescue the functional defects of H275Y and contribute to the evolution of drug resistance in IAV.
- Published
- 2022
- Full Text
- View/download PDF
12. US201 Study: A Phase 2, Randomized Proof-of-Concept Trial of Favipiravir for the Treatment of COVID-19.
- Author
-
Finberg RW, Ashraf M, Julg B, Ayoade F, Marathe JG, Issa NC, Wang JP, Jaijakul S, Baden LR, and Epstein C
- Abstract
Background: Favipiravir is used to treat influenza, and studies demonstrate that it has antiviral activity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)., Methods: We performed a randomized, open-label, multicenter, phase 2 proof-of-concept trial of favipiravir in hospitalized adult patients with polymerase chain reaction (PCR)-positive coronavirus disease 2019 (COVID-19). Patients were randomized to standard of care (SOC) or favipiravir treatment (1800mg per os twice a day [b.i.d.] on day 1, followed by 1000mg b.i.d. for 13 days). The primary end point was time to viral clearance on day 29., Results: Fifty patients were enrolled and stratified by disease severity (critical disease, severe disease, or mild to moderate disease). Nineteen patients were censored from the event of viral clearance based on being SARS-CoV-2 PCR-negative at the study outset, being PCR-positive at day 29, or because of loss to follow-up. Data from the 31 remaining patients who achieved viral clearance show enhanced viral clearance in the favipiravir group compared with the SOC group by day 29, with 72% of the favipiravir group and 52% of the SOC group being evaluable for viral clearance through day 29. The median time to viral clearance was 16.0 days (90% CI, 12.0 to 29.0) in the favipiravir group and 30.0 days (90% CI, 12.0 to 31.0) in the SOC group. A post hoc analysis revealed an effect in the subgroup of patients who were neutralizing antibody-negative at randomization. Treatment-emergent adverse events were equally distributed between the groups., Conclusions: We demonstrate that favipiravir can be safely administered to hospitalized adults with COVID-19 and believe that further studies are warranted., Clinicaltrialsgov Registration: NCT04358549., (© The Author(s) 2021. Published by Oxford University Press on behalf of Infectious Diseases Society of America.)
- Published
- 2021
- Full Text
- View/download PDF
13. Human nasal wash RNA-Seq reveals distinct cell-specific innate immune responses in influenza versus SARS-CoV-2.
- Author
-
Gao KM, Derr AG, Guo Z, Nündel K, Marshak-Rothstein A, Finberg RW, and Wang JP
- Subjects
- Adult, Female, Humans, Male, Nasal Lavage, COVID-19 genetics, COVID-19 immunology, Immunity, Innate, Influenza A virus genetics, Influenza A virus immunology, Influenza, Human genetics, Influenza, Human immunology, Macrophages immunology, Macrophages virology, RNA-Seq, SARS-CoV-2 genetics, SARS-CoV-2 immunology
- Abstract
BACKGROUNDInfluenza A virus (IAV) and SARS-CoV-2 are pandemic viruses causing millions of deaths, yet their clinical manifestations are distinctly different.METHODSWith the hypothesis that upper airway immune and epithelial cell responses are also distinct, we performed single-cell RNA sequencing (scRNA-Seq) on nasal wash cells freshly collected from adults with either acute COVID-19 or influenza or from healthy controls. We focused on major cell types and subtypes in a subset of donor samples.ResultsNasal wash cells were enriched for macrophages and neutrophils for both individuals with influenza and those with COVID-19 compared with healthy controls. Hillock-like epithelial cells, M2-like macrophages, and age-dependent B cells were enriched in COVID-19 samples. A global decrease in IFN-associated transcripts in neutrophils, macrophages, and epithelial cells was apparent in COVID-19 samples compared with influenza samples. The innate immune response to SARS-CoV-2 appears to be maintained in macrophages, despite evidence for limited epithelial cell immune sensing. Cell-to-cell interaction analyses revealed a decrease in epithelial cell interactions in COVID-19 and highlighted differences in macrophage-macrophage interactions for COVID-19 and influenza.ConclusionsOur study demonstrates that scRNA-Seq can define host and viral transcriptional activity at the site of infection and reveal distinct local epithelial and immune cell responses for COVID-19 and influenza that may contribute to their divergent disease courses.FundingMassachusetts Consortium on Pathogen Readiness, the Mathers Foundation, and the Department of Defense (W81XWH2110029) "COVID-19 Expansion for AIRe Program."
- Published
- 2021
- Full Text
- View/download PDF
14. SARS-CoV-2 Initiates Programmed Cell Death in Platelets.
- Author
-
Koupenova M, Corkrey HA, Vitseva O, Tanriverdi K, Somasundaran M, Liu P, Soofi S, Bhandari R, Godwin M, Parsi KM, Cousineau A, Maehr R, Wang JP, Cameron SJ, Rade J, Finberg RW, and Freedman JE
- Subjects
- A549 Cells, Adult, Blood Platelets ultrastructure, Blood Platelets virology, Blotting, Western, COVID-19 blood, COVID-19 virology, Female, Host-Pathogen Interactions, Humans, Male, Microscopy, Electron, Transmission, Necroptosis, SARS-CoV-2 physiology, Angiotensin-Converting Enzyme 2 metabolism, Apoptosis, Blood Platelets metabolism, COVID-19 metabolism, Serine Endopeptidases metabolism
- Abstract
[Figure: see text].
- Published
- 2021
- Full Text
- View/download PDF
15. High-throughput human primary cell-based airway model for evaluating influenza, coronavirus, or other respiratory viruses in vitro.
- Author
-
Gard AL, Luu RJ, Miller CR, Maloney R, Cain BP, Marr EE, Burns DM, Gaibler R, Mulhern TJ, Wong CA, Alladina J, Coppeta JR, Liu P, Wang JP, Azizgolshani H, Fezzie RF, Balestrini JL, Isenberg BC, Medoff BD, Finberg RW, and Borenstein JT
- Subjects
- Bronchi cytology, Bronchi virology, COVID-19 virology, Cell Culture Techniques instrumentation, Cell Line, Coronavirus drug effects, Coronavirus Infections drug therapy, Equipment Design, High-Throughput Screening Assays instrumentation, Humans, Influenza A virus drug effects, Influenza, Human drug therapy, Respiratory Mucosa virology, Respiratory Tract Infections drug therapy, Respiratory Tract Infections virology, SARS-CoV-2 drug effects, COVID-19 Drug Treatment, Antiviral Agents pharmacology, Coronavirus Infections virology, Influenza, Human virology, Microbial Sensitivity Tests instrumentation, Microfluidic Analytical Techniques instrumentation, Respiratory Mucosa cytology
- Abstract
Influenza and other respiratory viruses present a significant threat to public health, national security, and the world economy, and can lead to the emergence of global pandemics such as from COVID-19. A barrier to the development of effective therapeutics is the absence of a robust and predictive preclinical model, with most studies relying on a combination of in vitro screening with immortalized cell lines and low-throughput animal models. Here, we integrate human primary airway epithelial cells into a custom-engineered 96-device platform (PREDICT96-ALI) in which tissues are cultured in an array of microchannel-based culture chambers at an air-liquid interface, in a configuration compatible with high resolution in-situ imaging and real-time sensing. We apply this platform to influenza A virus and coronavirus infections, evaluating viral infection kinetics and antiviral agent dosing across multiple strains and donor populations of human primary cells. Human coronaviruses HCoV-NL63 and SARS-CoV-2 enter host cells via ACE2 and utilize the protease TMPRSS2 for spike protein priming, and we confirm their expression, demonstrate infection across a range of multiplicities of infection, and evaluate the efficacy of camostat mesylate, a known inhibitor of HCoV-NL63 infection. This new capability can be used to address a major gap in the rapid assessment of therapeutic efficacy of small molecules and antiviral agents against influenza and other respiratory viruses including coronaviruses., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
16. Expression of SARS coronavirus 1 spike protein from a herpesviral vector induces innate immune signaling and neutralizing antibody responses.
- Author
-
Kurt-Jones EA, Dudek TE, Watanabe D, Mandell L, Che J, Zhou S, Cao L, Greenough T, Babcock GJ, Diaz F, Oh HS, Zhou C, Finberg RW, and Knipe DM
- Subjects
- Animals, Antibodies, Neutralizing blood, Antibodies, Viral blood, Antibodies, Viral immunology, Cell Fusion, Cell Line, Cytokines immunology, Genetic Vectors, Humans, Mice, Severe acute respiratory syndrome-related coronavirus genetics, Signal Transduction, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus metabolism, Vaccines, Virus-Like Particle immunology, Viral Vaccines immunology, Antibodies, Neutralizing immunology, Herpesvirus 1, Human genetics, Immunity, Innate, Severe acute respiratory syndrome-related coronavirus immunology, Spike Glycoprotein, Coronavirus immunology
- Abstract
SARS coronavirus 1 (SARS-CoV-1) causes a respiratory infection that can lead to acute respiratory distress characterized by inflammation and high levels of cytokines in the lung tissue. In this study we constructed a herpes simplex virus 1 replication-defective mutant vector expressing SARS-CoV-1 spike protein as a potential vaccine vector and to probe the effects of spike protein on host cells. The spike protein expressed from this vector is functional in that it localizes to the surface of infected cells and induces fusion of ACE2-expressing cells. In immunized mice, the recombinant vector induced antibodies that bind to spike protein in an ELISA assay and that show neutralizing activity. The spike protein expressed from this vector can induce the expression of cytokines in an ACE2-independent, MyD88-dependent process. These results argue that the SARS-CoV-1 spike protein intrinsically activates signaling pathways that induce cytokines and contribute directly to the inflammatory process of SARS., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
17. Baricitinib plus Remdesivir for Hospitalized Adults with Covid-19.
- Author
-
Kalil AC, Patterson TF, Mehta AK, Tomashek KM, Wolfe CR, Ghazaryan V, Marconi VC, Ruiz-Palacios GM, Hsieh L, Kline S, Tapson V, Iovine NM, Jain MK, Sweeney DA, El Sahly HM, Branche AR, Regalado Pineda J, Lye DC, Sandkovsky U, Luetkemeyer AF, Cohen SH, Finberg RW, Jackson PEH, Taiwo B, Paules CI, Arguinchona H, Erdmann N, Ahuja N, Frank M, Oh MD, Kim ES, Tan SY, Mularski RA, Nielsen H, Ponce PO, Taylor BS, Larson L, Rouphael NG, Saklawi Y, Cantos VD, Ko ER, Engemann JJ, Amin AN, Watanabe M, Billings J, Elie MC, Davey RT, Burgess TH, Ferreira J, Green M, Makowski M, Cardoso A, de Bono S, Bonnett T, Proschan M, Deye GA, Dempsey W, Nayak SU, Dodd LE, and Beigel JH
- Subjects
- Adenosine Monophosphate adverse effects, Adenosine Monophosphate therapeutic use, Adult, Aged, Alanine adverse effects, Alanine therapeutic use, Antiviral Agents adverse effects, Azetidines adverse effects, COVID-19 mortality, COVID-19 therapy, Double-Blind Method, Drug Therapy, Combination, Female, Hospital Mortality, Hospitalization, Humans, Janus Kinase Inhibitors adverse effects, Janus Kinase Inhibitors therapeutic use, Male, Middle Aged, Oxygen Inhalation Therapy, Purines adverse effects, Pyrazoles adverse effects, Respiration, Artificial, Sulfonamides adverse effects, Treatment Outcome, Adenosine Monophosphate analogs & derivatives, Alanine analogs & derivatives, Antiviral Agents therapeutic use, Azetidines therapeutic use, Purines therapeutic use, Pyrazoles therapeutic use, Sulfonamides therapeutic use, COVID-19 Drug Treatment
- Abstract
Background: Severe coronavirus disease 2019 (Covid-19) is associated with dysregulated inflammation. The effects of combination treatment with baricitinib, a Janus kinase inhibitor, plus remdesivir are not known., Methods: We conducted a double-blind, randomized, placebo-controlled trial evaluating baricitinib plus remdesivir in hospitalized adults with Covid-19. All the patients received remdesivir (≤10 days) and either baricitinib (≤14 days) or placebo (control). The primary outcome was the time to recovery. The key secondary outcome was clinical status at day 15., Results: A total of 1033 patients underwent randomization (with 515 assigned to combination treatment and 518 to control). Patients receiving baricitinib had a median time to recovery of 7 days (95% confidence interval [CI], 6 to 8), as compared with 8 days (95% CI, 7 to 9) with control (rate ratio for recovery, 1.16; 95% CI, 1.01 to 1.32; P = 0.03), and a 30% higher odds of improvement in clinical status at day 15 (odds ratio, 1.3; 95% CI, 1.0 to 1.6). Patients receiving high-flow oxygen or noninvasive ventilation at enrollment had a time to recovery of 10 days with combination treatment and 18 days with control (rate ratio for recovery, 1.51; 95% CI, 1.10 to 2.08). The 28-day mortality was 5.1% in the combination group and 7.8% in the control group (hazard ratio for death, 0.65; 95% CI, 0.39 to 1.09). Serious adverse events were less frequent in the combination group than in the control group (16.0% vs. 21.0%; difference, -5.0 percentage points; 95% CI, -9.8 to -0.3; P = 0.03), as were new infections (5.9% vs. 11.2%; difference, -5.3 percentage points; 95% CI, -8.7 to -1.9; P = 0.003)., Conclusions: Baricitinib plus remdesivir was superior to remdesivir alone in reducing recovery time and accelerating improvement in clinical status among patients with Covid-19, notably among those receiving high-flow oxygen or noninvasive ventilation. The combination was associated with fewer serious adverse events. (Funded by the National Institute of Allergy and Infectious Diseases; ClinicalTrials.gov number, NCT04401579.)., (Copyright © 2020 Massachusetts Medical Society.)
- Published
- 2021
- Full Text
- View/download PDF
18. Unique structural solution from a V H 3-30 antibody targeting the hemagglutinin stem of influenza A viruses.
- Author
-
Harshbarger WD, Deming D, Lockbaum GJ, Attatippaholkun N, Kamkaew M, Hou S, Somasundaran M, Wang JP, Finberg RW, Zhu QK, Schiffer CA, and Marasco WA
- Subjects
- Antibodies, Neutralizing metabolism, Antibodies, Viral metabolism, Epitopes chemistry, Epitopes metabolism, Hemagglutinin Glycoproteins, Influenza Virus chemistry, Hemagglutinin Glycoproteins, Influenza Virus metabolism, Humans, Influenza A virus metabolism, Influenza Vaccines immunology, Influenza, Human immunology, Influenza, Human virology, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Epitopes immunology, Hemagglutinin Glycoproteins, Influenza Virus immunology, Influenza A virus immunology
- Abstract
Broadly neutralizing antibodies (bnAbs) targeting conserved influenza A virus (IAV) hemagglutinin (HA) epitopes can provide valuable information for accelerating universal vaccine designs. Here, we report structural details for heterosubtypic recognition of HA from circulating and emerging IAVs by the human antibody 3I14. Somatic hypermutations play a critical role in shaping the HCDR3, which alone and uniquely among V
H 3-30 derived antibodies, forms contacts with five sub-pockets within the HA-stem hydrophobic groove. 3I14 light-chain interactions are also key for binding HA and contribute a large buried surface area spanning two HA protomers. Comparison of 3I14 to bnAbs from several defined classes provide insights to the bias selection of VH 3-30 antibodies and reveals that 3I14 represents a novel structural solution within the VH 3-30 repertoire. The structures reported here improve our understanding of cross-group heterosubtypic binding activity, providing the basis for advancing immunogen designs aimed at eliciting a broadly protective response to IAV.- Published
- 2021
- Full Text
- View/download PDF
19. Remdesivir for the Treatment of Covid-19 - Final Report.
- Author
-
Beigel JH, Tomashek KM, Dodd LE, Mehta AK, Zingman BS, Kalil AC, Hohmann E, Chu HY, Luetkemeyer A, Kline S, Lopez de Castilla D, Finberg RW, Dierberg K, Tapson V, Hsieh L, Patterson TF, Paredes R, Sweeney DA, Short WR, Touloumi G, Lye DC, Ohmagari N, Oh MD, Ruiz-Palacios GM, Benfield T, Fätkenheuer G, Kortepeter MG, Atmar RL, Creech CB, Lundgren J, Babiker AG, Pett S, Neaton JD, Burgess TH, Bonnett T, Green M, Makowski M, Osinusi A, Nayak S, and Lane HC
- Subjects
- Adenosine Monophosphate administration & dosage, Adenosine Monophosphate adverse effects, Adenosine Monophosphate therapeutic use, Administration, Intravenous, Adult, Aged, Alanine administration & dosage, Alanine adverse effects, Alanine therapeutic use, Antiviral Agents administration & dosage, Antiviral Agents adverse effects, Betacoronavirus, COVID-19, Coronavirus Infections mortality, Coronavirus Infections therapy, Double-Blind Method, Extracorporeal Membrane Oxygenation, Female, Humans, Kaplan-Meier Estimate, Male, Middle Aged, Oxygen Inhalation Therapy, Pandemics, Pneumonia, Viral mortality, Pneumonia, Viral therapy, Respiration, Artificial, SARS-CoV-2, Time Factors, Young Adult, COVID-19 Drug Treatment, Adenosine Monophosphate analogs & derivatives, Alanine analogs & derivatives, Antiviral Agents therapeutic use, Coronavirus Infections drug therapy, Pneumonia, Viral drug therapy
- Abstract
Background: Although several therapeutic agents have been evaluated for the treatment of coronavirus disease 2019 (Covid-19), no antiviral agents have yet been shown to be efficacious., Methods: We conducted a double-blind, randomized, placebo-controlled trial of intravenous remdesivir in adults who were hospitalized with Covid-19 and had evidence of lower respiratory tract infection. Patients were randomly assigned to receive either remdesivir (200 mg loading dose on day 1, followed by 100 mg daily for up to 9 additional days) or placebo for up to 10 days. The primary outcome was the time to recovery, defined by either discharge from the hospital or hospitalization for infection-control purposes only., Results: A total of 1062 patients underwent randomization (with 541 assigned to remdesivir and 521 to placebo). Those who received remdesivir had a median recovery time of 10 days (95% confidence interval [CI], 9 to 11), as compared with 15 days (95% CI, 13 to 18) among those who received placebo (rate ratio for recovery, 1.29; 95% CI, 1.12 to 1.49; P<0.001, by a log-rank test). In an analysis that used a proportional-odds model with an eight-category ordinal scale, the patients who received remdesivir were found to be more likely than those who received placebo to have clinical improvement at day 15 (odds ratio, 1.5; 95% CI, 1.2 to 1.9, after adjustment for actual disease severity). The Kaplan-Meier estimates of mortality were 6.7% with remdesivir and 11.9% with placebo by day 15 and 11.4% with remdesivir and 15.2% with placebo by day 29 (hazard ratio, 0.73; 95% CI, 0.52 to 1.03). Serious adverse events were reported in 131 of the 532 patients who received remdesivir (24.6%) and in 163 of the 516 patients who received placebo (31.6%)., Conclusions: Our data show that remdesivir was superior to placebo in shortening the time to recovery in adults who were hospitalized with Covid-19 and had evidence of lower respiratory tract infection. (Funded by the National Institute of Allergy and Infectious Diseases and others; ACTT-1 ClinicalTrials.gov number, NCT04280705.)., (Copyright © 2020 Massachusetts Medical Society.)
- Published
- 2020
- Full Text
- View/download PDF
20. SARS-CoV-2 Receptor ACE2 Is an Interferon-Stimulated Gene in Human Airway Epithelial Cells and Is Detected in Specific Cell Subsets across Tissues.
- Author
-
Ziegler CGK, Allon SJ, Nyquist SK, Mbano IM, Miao VN, Tzouanas CN, Cao Y, Yousif AS, Bals J, Hauser BM, Feldman J, Muus C, Wadsworth MH 2nd, Kazer SW, Hughes TK, Doran B, Gatter GJ, Vukovic M, Taliaferro F, Mead BE, Guo Z, Wang JP, Gras D, Plaisant M, Ansari M, Angelidis I, Adler H, Sucre JMS, Taylor CJ, Lin B, Waghray A, Mitsialis V, Dwyer DF, Buchheit KM, Boyce JA, Barrett NA, Laidlaw TM, Carroll SL, Colonna L, Tkachev V, Peterson CW, Yu A, Zheng HB, Gideon HP, Winchell CG, Lin PL, Bingle CD, Snapper SB, Kropski JA, Theis FJ, Schiller HB, Zaragosi LE, Barbry P, Leslie A, Kiem HP, Flynn JL, Fortune SM, Berger B, Finberg RW, Kean LS, Garber M, Schmidt AG, Lingwood D, Shalek AK, and Ordovas-Montanes J
- Subjects
- Adolescent, Alveolar Epithelial Cells immunology, Angiotensin-Converting Enzyme 2, Animals, Betacoronavirus physiology, COVID-19, Cell Line, Cells, Cultured, Child, Coronavirus Infections virology, Enterocytes immunology, Goblet Cells immunology, HIV Infections immunology, Humans, Influenza, Human immunology, Interferon Type I immunology, Lung cytology, Lung pathology, Macaca mulatta, Mice, Mycobacterium tuberculosis, Nasal Mucosa immunology, Pandemics, Peptidyl-Dipeptidase A metabolism, Pneumonia, Viral virology, Receptors, Virus genetics, SARS-CoV-2, Serine Endopeptidases metabolism, Single-Cell Analysis, Tuberculosis immunology, Up-Regulation, Alveolar Epithelial Cells metabolism, Enterocytes metabolism, Goblet Cells metabolism, Interferon Type I metabolism, Nasal Mucosa cytology, Peptidyl-Dipeptidase A genetics
- Abstract
There is pressing urgency to understand the pathogenesis of the severe acute respiratory syndrome coronavirus clade 2 (SARS-CoV-2), which causes the disease COVID-19. SARS-CoV-2 spike (S) protein binds angiotensin-converting enzyme 2 (ACE2), and in concert with host proteases, principally transmembrane serine protease 2 (TMPRSS2), promotes cellular entry. The cell subsets targeted by SARS-CoV-2 in host tissues and the factors that regulate ACE2 expression remain unknown. Here, we leverage human, non-human primate, and mouse single-cell RNA-sequencing (scRNA-seq) datasets across health and disease to uncover putative targets of SARS-CoV-2 among tissue-resident cell subsets. We identify ACE2 and TMPRSS2 co-expressing cells within lung type II pneumocytes, ileal absorptive enterocytes, and nasal goblet secretory cells. Strikingly, we discovered that ACE2 is a human interferon-stimulated gene (ISG) in vitro using airway epithelial cells and extend our findings to in vivo viral infections. Our data suggest that SARS-CoV-2 could exploit species-specific interferon-driven upregulation of ACE2, a tissue-protective mediator during lung injury, to enhance infection., Competing Interests: Declaration of Interests A.R. is an SAB member of ThermoFisher Scientific, Neogene Therapeutics, Asimov, and Syros Pharmaceuticals; a co-founder of and equity holder in Celsius Therapeutics; and an equity holder in Immunitas Therapeutics. A.K.S. reports compensation for consulting and/or SAB membership from Merck, Honeycomb Biotechnologies, Cellarity, Cogen Therapeutics, Orche Bio, and Dahlia Biosciences. L.S.K. is on the SAB for HiFiBio; she reports research funding from Kymab Limited, Bristol Meyers Squibb, Magenta Therapeutics, BlueBird Bio, and Regeneron Pharmaceuticals and consulting fees from Equillium, FortySeven, Inc, Novartis, Inc, EMD Serono, Gilead Sciences, and Takeda Pharmaceuticals. A.S. is an employee of Johnson and Johnson. N.K. is an inventor on a patent using thyroid hormone mimetics in acute lung injury that is now being considered for intervention in COVID-19 patients. J.L. is a scientific consultant for 10X Genomics, Inc. O.R.R, is a co-inventor on patent applications filed by the Broad Institute to inventions relating to single-cell genomics applications, such as in PCT/US2018/060860 and US Provisional Application No. 62/745,259. S.T. in the last three years was a consultant at Genentech, Biogen, and Roche and is a member of the SAB of Foresite Labs. M.H.W. is now an employee of Pfizer. F.J.T. reports receiving consulting fees from Roche Diagnostics GmbH and ownership interest in Cellarity, Inc. P.H. is a co-inventor on a patent using artificial intelligence and high-resolution microscopy for COVID-19 infection testing based on serology., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
21. The role of platelets in mediating a response to human influenza infection.
- Author
-
Koupenova M, Corkrey HA, Vitseva O, Manni G, Pang CJ, Clancy L, Yao C, Rade J, Levy D, Wang JP, Finberg RW, Kurt-Jones EA, and Freedman JE
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Complement C3 immunology, Complement C3 metabolism, Disease Models, Animal, Extracellular Traps immunology, Extracellular Traps metabolism, Female, Humans, Influenza A virus isolation & purification, Influenza, Human blood, Influenza, Human virology, Male, Membrane Glycoproteins genetics, Membrane Glycoproteins immunology, Membrane Glycoproteins metabolism, Mice, Inbred C57BL, Mice, Knockout, Middle Aged, Neutrophils immunology, Toll-Like Receptor 7 genetics, Toll-Like Receptor 7 immunology, Toll-Like Receptor 7 metabolism, Blood Platelets immunology, Influenza A virus immunology, Influenza, Human immunology, Platelet Activation immunology
- Abstract
Influenza infection increases the incidence of myocardial infarction but the reason is unknown. Platelets mediate vascular occlusion through thrombotic functions but are also recognized to have immunomodulatory activity. To determine if platelet processes are activated during influenza infection, we collected blood from 18 patients with acute influenza infection. Microscopy reveals activated platelets, many containing viral particles and extracellular-DNA associated with platelets. To understand the mechanism, we isolate human platelets and treat them with influenza A virus. Viral-engulfment leads to C3 release from platelets as a function of TLR7 and C3 leads to neutrophil-DNA release and aggregation. TLR7 specificity is confirmed in murine models lacking the receptor, and platelet depletion models support platelet-mediated C3 and neutrophil-DNA release post-influenza infection. These findings demonstrate that the initial intrinsic defense against influenza is mediated by platelet-neutrophil cross-communication that tightly regulates host immune and complement responses but can also lead to thrombotic vascular occlusion.
- Published
- 2019
- Full Text
- View/download PDF
22. Phase 2b Study of Pimodivir (JNJ-63623872) as Monotherapy or in Combination With Oseltamivir for Treatment of Acute Uncomplicated Seasonal Influenza A: TOPAZ Trial.
- Author
-
Finberg RW, Lanno R, Anderson D, Fleischhackl R, van Duijnhoven W, Kauffman RS, Kosoglou T, Vingerhoets J, and Leopold L
- Subjects
- Acute Disease, Adult, Antiviral Agents adverse effects, Antiviral Agents blood, Antiviral Agents pharmacokinetics, Diarrhea chemically induced, Double-Blind Method, Drug Resistance, Viral genetics, Drug Therapy, Combination, Early Termination of Clinical Trials, Female, Humans, Influenza, Human virology, Male, Middle Aged, Oseltamivir blood, Pyridines adverse effects, Pyridines blood, Pyridines pharmacokinetics, Pyrimidines adverse effects, Pyrimidines blood, Pyrimidines pharmacokinetics, Pyrroles adverse effects, Pyrroles blood, Pyrroles pharmacokinetics, Time Factors, Viral Load, Viral Proteins genetics, Virus Shedding, Antiviral Agents therapeutic use, Influenza A virus genetics, Influenza A virus physiology, Influenza, Human drug therapy, Oseltamivir therapeutic use, Pyridines therapeutic use, Pyrimidines therapeutic use, Pyrroles therapeutic use
- Abstract
Background: Pimodivir, a first-in-class inhibitor of influenza virus polymerase basic protein 2, is being developed for hospitalized and high-risk patients with influenza A., Methods: In this double-blinded phase 2b study, adults with acute uncomplicated influenza A were randomized 1:1:1:1 to receive one of the following treatments twice daily for 5 days: placebo, pimodivir 300 mg or 600 mg, or pimodivir 600 mg plus oseltamivir 75 mg. Antiviral activity, safety, and pharmacokinetics of pimodivir alone or in combination were evaluated., Results: Of 292 patients randomized, 223 were treated and had confirmed influenza A virus infection. The trial was stopped early because the primary end point was met; the area under the curve of the viral load, determined by quantitative reverse transcription-polymerase chain reaction analysis, in nasal secretions from baseline to day 8 significantly decreased in the active treatment groups, compared with the placebo group (300 mg group, -3.6 day*log10 copies/mL [95% confidence interval {CI}, -7.1 to -0.1]; 600 mg group, -4.5 [95%CI -8.0 to -1.0]; and combination group, -8.6 [95% CI, -12.0 to -5.1]). Pimodivir plus oseltamivir yielded a significantly lower viral load titer over time than placebo and a trend for a shorter time to symptom resolution than placebo. Pimodivir plasma concentrations increased in a dose-proportional manner. The most commonly reported adverse event was mild or moderate diarrhea., Conclusions: Pimodivir (with or without oseltamivir) resulted in significant virologic improvements over placebo, demonstrated trends in clinical improvement, and was well tolerated. Pimodivir 600 mg twice daily is in further development., Clinical Trials Registration: NCT02342249, 2014-004068-39, and CR107745., (© The Author(s) 2018. Published by Oxford University Press for the Infectious Diseases Society of America. All rights reserved. For permissions, e-mail: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
23. A Disintegrin and Metalloproteinase 9 Domain (ADAM9) Is a Major Susceptibility Factor in the Early Stages of Encephalomyocarditis Virus Infection.
- Author
-
Bazzone LE, King M, MacKay CR, Kyawe PP, Meraner P, Lindstrom D, Rojas-Quintero J, Owen CA, Wang JP, Brass AL, Kurt-Jones EA, and Finberg RW
- Subjects
- Animals, Cell Line, Gene Knockout Techniques, Genetic Testing, Humans, Mice, Mice, Knockout, Models, Biological, ADAM Proteins metabolism, Cardiovirus Infections genetics, Disease Resistance, Encephalomyocarditis virus growth & development, Membrane Proteins metabolism
- Abstract
Encephalomyocarditis virus (EMCV) is a picornavirus that produces lytic infections in murine and human cells. Employing a genome-wide CRISPR-Cas9 knockout screen to find host factors required for EMCV infection, we identified a role for ADAM9 in EMCV infection. CRISPR-mediated deletion of ADAM9 in multiple human cell lines rendered the cells highly resistant to EMCV infection and cell death. Primary fibroblasts from ADAM9 KO mice were also strongly resistant to EMCV infection and cell death. In contrast, ADAM9 KO and WT cells were equally susceptible to infection with other viruses, including the picornavirus Coxsackie virus B. ADAM9 KO cells failed to produce viral progeny when incubated with EMCV. However, bypassing EMCV entry into cells through delivery of viral RNA directly to the cytosol yielded infectious EMCV virions from ADAM9 KO cells, suggesting that ADAM9 is not required for EMCV replication post-entry. These findings establish that ADAM9 is required for the early stage of EMCV infection, likely for virus entry or viral genome delivery to the cytosol. IMPORTANCE Viral myocarditis is a leading cause of death in the United States, contributing to numerous unexplained deaths in people ≤35 years old. Enteroviruses contribute to many cases of human myocarditis. Encephalomyocarditis virus (EMCV) infection causes viral myocarditis in rodent models, but its receptor requirements have not been fully identified. CRISPR-Cas9 screens can identify host dependency factors essential for EMCV infection and enhance our understanding of key events that follow viral infection, potentially leading to new strategies for preventing viral myocarditis. Using a CRISPR-Cas9 screen, we identified ad isintegrin a nd m etalloproteinase 9 domain (ADAM9) as a major factor required for the early stages of EMCV infection in both human and murine infection., (Copyright © 2019 Bazzone et al.)
- Published
- 2019
- Full Text
- View/download PDF
24. Mutations in Influenza A Virus Neuraminidase and Hemagglutinin Confer Resistance against a Broadly Neutralizing Hemagglutinin Stem Antibody.
- Author
-
Prachanronarong KL, Canale AS, Liu P, Somasundaran M, Hou S, Poh YP, Han T, Zhu Q, Renzette N, Zeldovich KB, Kowalik TF, Kurt-Yilmaz N, Jensen JD, Bolon DNA, Marasco WA, Finberg RW, Schiffer CA, and Wang JP
- Subjects
- Animals, Antibodies, Neutralizing pharmacology, Antibodies, Viral pharmacology, Dogs, Hemagglutinin Glycoproteins, Influenza Virus chemistry, Influenza A Virus, H1N1 Subtype drug effects, Influenza Vaccines, Madin Darby Canine Kidney Cells, Models, Molecular, Neuraminidase chemistry, Neutralization Tests, Reverse Genetics, Sequence Analysis, RNA, Viral Proteins chemistry, Viral Proteins genetics, Drug Resistance, Viral, Hemagglutinin Glycoproteins, Influenza Virus genetics, Influenza A Virus, H1N1 Subtype genetics, Mutation, Neuraminidase genetics
- Abstract
Influenza A virus (IAV), a major cause of human morbidity and mortality, continuously evolves in response to selective pressures. Stem-directed, broadly neutralizing antibodies (sBnAbs) targeting the influenza virus hemagglutinin (HA) are a promising therapeutic strategy, but neutralization escape mutants can develop. We used an integrated approach combining viral passaging, deep sequencing, and protein structural analyses to define escape mutations and mechanisms of neutralization escape in vitro for the F10 sBnAb. IAV was propagated with escalating concentrations of F10 over serial passages in cultured cells to select for escape mutations. Viral sequence analysis revealed three mutations in HA and one in neuraminidase (NA). Introduction of these specific mutations into IAV through reverse genetics confirmed their roles in resistance to F10. Structural analyses revealed that the selected HA mutations (S123G, N460S, and N203V) are away from the F10 epitope but may indirectly impact influenza virus receptor binding, endosomal fusion, or budding. The NA mutation E329K, which was previously identified to be associated with antibody escape, affects the active site of NA, highlighting the importance of the balance between HA and NA function for viral survival. Thus, whole-genome population sequencing enables the identification of viral resistance mutations responding to antibody-induced selective pressure. IMPORTANCE Influenza A virus is a public health threat for which currently available vaccines are not always effective. Broadly neutralizing antibodies that bind to the highly conserved stem region of the influenza virus hemagglutinin (HA) can neutralize many influenza virus strains. To understand how influenza virus can become resistant or escape such antibodies, we propagated influenza A virus in vitro with escalating concentrations of antibody and analyzed viral populations by whole-genome sequencing. We identified HA mutations near and distal to the antibody binding epitope that conferred resistance to antibody neutralization. Additionally, we identified a neuraminidase (NA) mutation that allowed the virus to grow in the presence of high concentrations of the antibody. Virus carrying dual mutations in HA and NA also grew under high antibody concentrations. We show that NA mutations mediate the escape of neutralization by antibodies against HA, highlighting the importance of a balance between HA and NA for optimal virus function., (Copyright © 2019 American Society for Microbiology.)
- Published
- 2019
- Full Text
- View/download PDF
25. Synonymous Mutations at the Beginning of the Influenza A Virus Hemagglutinin Gene Impact Experimental Fitness.
- Author
-
Canale AS, Venev SV, Whitfield TW, Caffrey DR, Marasco WA, Schiffer CA, Kowalik TF, Jensen JD, Finberg RW, Zeldovich KB, Wang JP, and Bolon DNA
- Subjects
- Amino Acid Substitution, Animals, Dogs, Evolution, Molecular, HEK293 Cells, Humans, Influenza A Virus, H1N1 Subtype genetics, Madin Darby Canine Kidney Cells, Models, Molecular, Phylogeny, RNA Folding, Swine, Virus Replication, Genetic Fitness, Hemagglutinin Glycoproteins, Influenza Virus chemistry, Hemagglutinin Glycoproteins, Influenza Virus genetics, Influenza A Virus, H1N1 Subtype growth & development, Silent Mutation
- Abstract
The fitness effects of synonymous mutations can provide insights into biological and evolutionary mechanisms. We analyzed the experimental fitness effects of all single-nucleotide mutations, including synonymous substitutions, at the beginning of the influenza A virus hemagglutinin (HA) gene. Many synonymous substitutions were deleterious both in bulk competition and for individually isolated clones. Investigating protein and RNA levels of a subset of individually expressed HA variants revealed that multiple biochemical properties contribute to the observed experimental fitness effects. Our results indicate that a structural element in the HA segment viral RNA may influence fitness. Examination of naturally evolved sequences in human hosts indicates a preference for the unfolded state of this structural element compared to that found in swine hosts. Our overall results reveal that synonymous mutations may have greater fitness consequences than indicated by simple models of sequence conservation, and we discuss the implications of this finding for commonly used evolutionary tests and analyses., (Copyright © 2018. Published by Elsevier Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
26. Structural Determination of the Broadly Reactive Anti-IGHV1-69 Anti-idiotypic Antibody G6 and Its Idiotope.
- Author
-
Avnir Y, Prachanronarong KL, Zhang Z, Hou S, Peterson EC, Sui J, Zayed H, Kurella VB, McGuire AT, Stamatatos L, Hilbert BJ, Bohn MF, Kowalik TF, Jensen JD, Finberg RW, Wang JP, Goodall M, Jefferis R, Zhu Q, Kurt Yilmaz N, Schiffer CA, and Marasco WA
- Subjects
- Amino Acid Sequence, Antibodies, Anti-Idiotypic genetics, Antibodies, Anti-Idiotypic immunology, Antibodies, Monoclonal, Humanized genetics, Antibodies, Monoclonal, Humanized immunology, Antibodies, Neutralizing genetics, Antibodies, Neutralizing immunology, Antibodies, Viral genetics, Antibodies, Viral immunology, Antibody Specificity, Binding Sites, Cloning, Molecular, Crystallography, X-Ray, Gene Expression, Hemagglutinin Glycoproteins, Influenza Virus genetics, Hemagglutinin Glycoproteins, Influenza Virus immunology, Humans, Models, Molecular, Orthomyxoviridae chemistry, Protein Binding, Protein Interaction Domains and Motifs, Protein Structure, Secondary, Receptors, Antigen, B-Cell genetics, Receptors, Antigen, B-Cell immunology, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins immunology, Sequence Alignment, Sequence Homology, Amino Acid, Antibodies, Anti-Idiotypic chemistry, Antibodies, Monoclonal, Humanized chemistry, Antibodies, Neutralizing chemistry, Antibodies, Viral chemistry, Hemagglutinin Glycoproteins, Influenza Virus chemistry, Receptors, Antigen, B-Cell chemistry
- Abstract
The heavy chain IGHV1-69 germline gene exhibits a high level of polymorphism and shows biased use in protective antibody (Ab) responses to infections and vaccines. It is also highly expressed in several B cell malignancies and autoimmune diseases. G6 is an anti-idiotypic monoclonal Ab that selectively binds to IGHV1-69 heavy chain germline gene 51p1 alleles that have been implicated in these Ab responses and disease processes. Here, we determine the co-crystal structure of humanized G6 (hG6.3) in complex with anti-influenza hemagglutinin stem-directed broadly neutralizing Ab D80. The core of the hG6.3 idiotope is a continuous string of CDR-H2 residues starting with M53 and ending with N58. G6 binding studies demonstrate the remarkable breadth of binding to 51p1 IGHV1-69 Abs with diverse CDR-H3, light chain, and antigen binding specificities. These studies detail the broad expression of the G6 cross-reactive idiotype (CRI) that further define its potential role in precision medicine., (Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
27. Eliminating Residents Increases the Cost of Care.
- Author
-
DeMarco DM, Forster R, Gakis T, and Finberg RW
- Subjects
- Health Care Sector, Hospital Costs, Humans, Medicare, United States, Education, Medical, Graduate economics, Financing, Government economics, Hospitals, Teaching economics, Internship and Residency economics, Training Support economics
- Abstract
Background: Academic health centers are facing a potential reduction in Medicare financing for graduate medical education (GME). Both the Medicare Payment Advisory Commission and the National Commission on Fiscal Responsibility and Reform (Deficit Commission) have suggested cutting approximately half the funding that teaching hospitals receive for indirect medical education. Because of the effort that goes into teaching trainees, who are only transient employees, hospital executives often see teaching programs as a drain on resources., Objective: In light of the possibility of a Medicare cut to GME programs, we undertook an analysis to assess the financial risk of training programs to our institution and the possibility of saving money by reducing resident positions., Methods: The chief administrative officer, in collaboration with the hospital chief financial officer, performed a financial analysis to examine the possibility of decreasing costs by reducing residency programs at the University of Massachusetts Memorial Medical Center., Results: Despite the real costs of our training programs, the analysis demonstrated that GME programs have a positive impact on hospital finances., Conclusions: Reducing or eliminating GME programs would have a negative impact on our hospital's bottom line., Competing Interests: Conflict of interest: The authors declare they have no competing interests.
- Published
- 2017
- Full Text
- View/download PDF
28. Association of hospital contact precaution policies with emergency department admission time.
- Author
-
Kotkowski K, Ellison RT 3rd, Barysauskas C, Barton B, Allison J, Mack D, Finberg RW, and Reznek M
- Subjects
- Adult, Carrier State diagnosis, Emergency Service, Hospital, Hospitals, Humans, Organizational Policy, Retrospective Studies, Time Factors, Cross Infection prevention & control, Emergency Medicine methods, Gram-Positive Bacterial Infections diagnosis, Infection Control methods, Methicillin-Resistant Staphylococcus aureus isolation & purification, Patient Admission, Vancomycin-Resistant Enterococci isolation & purification
- Abstract
Background: Contact precautions are a widely accepted strategy to reduce in-hospital transmission of meticillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant enterococci (VRE). However, these practices may have unintended deleterious effects on patients., Aim: To evaluate the effect of a modification in hospital-wide contact precaution practices on emergency department (ED) admission times., Methods: During the study period, the hospital changed its contact precaution policy from requiring contact precautions for all patients with a history of MRSA or VRE to only those who presented with clinical conditions likely to contaminate the environment with pathogens. An interrupted time series analysis of ED admission times for adults for one year preceding and one year following this change was performed at a two-campus hospital. The main outcome was admission time, defined as time from decision to admit to arrival in an inpatient bed, for patients with MRSA or VRE compared with all other patients. The in-hospital MRSA and VRE acquisition rates were evaluated over the same period and have been published previously., Findings: At one campus, admission time decreased immediately by 161min for MRSA patients (P=0.008) and 135min for VRE patients (P=0.003), and both continued to decrease over the duration of the study. There was no significant change in admission time at the second campus., Conclusions: Modifying contact precaution requirements for MRSA and VRE may be associated with improved ED admission time without significantly altering in-hospital MRSA and VRE acquisition., (Copyright © 2017 The Healthcare Infection Society. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
29. The Combined Effect of Oseltamivir and Favipiravir on Influenza A Virus Evolution.
- Author
-
Ormond L, Liu P, Matuszewski S, Renzette N, Bank C, Zeldovich K, Bolon DN, Kowalik TF, Finberg RW, Jensen JD, and Wang JP
- Subjects
- Animals, Cell Line, Dogs, Genetics, Population, Influenza A virus classification, Mutation Rate, Orthomyxoviridae Infections virology, Amides pharmacology, Antiviral Agents pharmacology, Biological Evolution, Influenza A virus drug effects, Orthomyxoviridae Infections drug therapy, Oseltamivir pharmacology, Pyrazines pharmacology
- Abstract
Influenza virus inflicts a heavy death toll annually and resistance to existing antiviral drugs has generated interest in the development of agents with novel mechanisms of action. Favipiravir is an antiviral drug that acts by increasing the genome-wide mutation rate of influenza A virus (IAV). Potential synergistic benefits of combining oseltamivir and favipiravir have been demonstrated in animal models of influenza, but the population-level effects of combining the drugs are unknown. In order to elucidate the underlying evolutionary processes at play, we performed genome-wide sequencing of IAV experimental populations subjected to serial passaging in vitro under a combined protocol of oseltamivir and favipiravir. We describe the interplay between mutation, selection, and genetic drift that ultimately culminates in population extinction. In particular, selective sweeps around oseltamivir resistance mutations reduce genome-wide variation while deleterious mutations hitchhike to fixation given the increased mutational load generated by favipiravir. This latter effect reduces viral fitness and accelerates extinction compared with IAV populations treated with favipiravir alone, but risks spreading both established and newly emerging mutations, including possible drug resistance mutations, if transmission occurs before the viral populations are eradicated., (© The Author 2017. Published by Oxford University Press on behalf of the Society for Molecular Biology and Evolution.)
- Published
- 2017
- Full Text
- View/download PDF
30. Molecular Basis for Differential Patterns of Drug Resistance in Influenza N1 and N2 Neuraminidase.
- Author
-
Prachanronarong KL, Özen A, Thayer KM, Yilmaz LS, Zeldovich KB, Bolon DN, Kowalik TF, Jensen JD, Finberg RW, Wang JP, Kurt-Yilmaz N, and Schiffer CA
- Subjects
- Antiviral Agents chemistry, Antiviral Agents metabolism, Antiviral Agents pharmacology, Binding Sites, Drug Resistance, Viral drug effects, Enzyme Inhibitors metabolism, Enzyme Inhibitors pharmacology, Humans, Hydrogen Bonding, Influenza, Human pathology, Influenza, Human virology, Molecular Dynamics Simulation, Mutagenesis, Site-Directed, Neuraminidase genetics, Neuraminidase metabolism, Oseltamivir chemistry, Oseltamivir metabolism, Oseltamivir pharmacology, Protein Binding, Protein Structure, Quaternary, Static Electricity, Substrate Specificity, Thermodynamics, Zanamivir chemistry, Zanamivir metabolism, Zanamivir pharmacology, Enzyme Inhibitors chemistry, Influenza A virus enzymology, Neuraminidase antagonists & inhibitors
- Abstract
Neuraminidase (NA) inhibitors are used for the prevention and treatment of influenza A virus infections. Two subtypes of NA, N1 and N2, predominate in viruses that infect humans, but differential patterns of drug resistance have emerged in each subtype despite highly homologous active sites. To understand the molecular basis for the selection of these drug resistance mutations, structural and dynamic analyses on complexes of N1 and N2 NA with substrates and inhibitors were performed. Comparison of dynamic substrate and inhibitor envelopes and interactions at the active site revealed how differential patterns of drug resistance have emerged for specific drug resistance mutations, at residues I222, S246, and H274 in N1 and E119 in N2. Our results show that the differences in intermolecular interactions, especially van der Waals contacts, of the inhibitors versus substrates at the NA active site effectively explain the selection of resistance mutations in the two subtypes. Avoiding such contacts that render inhibitors vulnerable to resistance by better mimicking the dynamics and intermolecular interactions of substrates can lead to the development of novel inhibitors that avoid drug resistance in both subtypes.
- Published
- 2016
- Full Text
- View/download PDF
31. Properdin Levels in Individuals with Chemotherapy-Induced Neutropenia.
- Author
-
Tsyrkunou A, Agarwal S, Koirala B, Finberg RW, Nath R, Barton B, Levitz SM, Wang JP, and Ram S
- Abstract
Background: Neutrophils produce and carry key components of the alternative pathway (AP) of complement, including properdin (P). The effect of chemotherapy-induced absolute neutropenia on circulating P levels and AP function has not been previously established., Methods: We prospectively measured free P levels in serum from 27 individuals expected to develop neutropenia after administration of chemotherapy for hematological malignancies in preparation for hematopoietic stem cell transplantation and here describe the relationship between serum P levels and the neutrophil count over time., Results: When the absolute neutrophil count (ANC) was >500 cells/mm
3 pre-chemotherapy, P levels were significantly higher than P levels associated with an ANC ≤500 cells/mm3 (median values 8392 ng/mL and 6355 ng/mL, respectively; P = .001). Pairwise comparison between pre-chemotherapy P levels and P levels at initial or last documented neutropenia before recovery showed a significant decline ( P < .0001). No correlation was observed between P levels during neutropenia and after recovery of neutropenia in 20 subjects for which postneutropenia samples were obtained. A small but significant ( P = .02) decrease in AP hemolytic activity was noted between baseline (preneutropenia) and samples obtained at the onset of neutropenia, but only with low (6.25%) and not higher (12.5 or 25%) serum concentrations., Conclusions: A decline in P levels and AP activity could contribute to the increased risk of infection in neutropenic patients and warrants further study., (© The Author 2016. Published by Oxford University Press on behalf of Infectious Diseases Society of America.)- Published
- 2016
- Full Text
- View/download PDF
32. An experimental evaluation of drug-induced mutational meltdown as an antiviral treatment strategy.
- Author
-
Bank C, Renzette N, Liu P, Matuszewski S, Shim H, Foll M, Bolon DN, Zeldovich KB, Kowalik TF, Finberg RW, Wang JP, and Jensen JD
- Subjects
- Animals, Dogs, Evolution, Molecular, Influenza A Virus, H1N1 Subtype drug effects, Madin Darby Canine Kidney Cells, Polymorphism, Genetic, Amides pharmacology, Anti-Retroviral Agents pharmacology, Drug Resistance, Viral genetics, Influenza A Virus, H1N1 Subtype genetics, Mutation Rate, Pyrazines pharmacology
- Abstract
The rapid evolution of drug resistance remains a critical public health concern. The treatment of influenza A virus (IAV) has proven particularly challenging, due to the ability of the virus to develop resistance against current antivirals and vaccines. Here, we evaluate a novel antiviral drug therapy, favipiravir, for which the mechanism of action in IAV involves an interaction with the viral RNA-dependent RNA polymerase resulting in an effective increase in the viral mutation rate. We used an experimental evolution framework, combined with novel population genetic method development for inference from time-sampled data, to evaluate the effectiveness of favipiravir against IAV. Evaluating whole genome polymorphism data across 15 time points under multiple drug concentrations and in controls, we present the first evidence for the ability of IAV populations to effectively adapt to low concentrations of favipiravir. In contrast, under high concentrations, we observe population extinction, indicative of mutational meltdown. We discuss the observed dynamics with respect to the evolutionary forces at play and emphasize the utility of evolutionary theory to inform drug development., (© 2016 The Author(s). Evolution © 2016 The Society for the Study of Evolution.)
- Published
- 2016
- Full Text
- View/download PDF
33. A Balance between Inhibitor Binding and Substrate Processing Confers Influenza Drug Resistance.
- Author
-
Jiang L, Liu P, Bank C, Renzette N, Prachanronarong K, Yilmaz LS, Caffrey DR, Zeldovich KB, Schiffer CA, Kowalik TF, Jensen JD, Finberg RW, Wang JP, and Bolon DNA
- Subjects
- Animals, Cell Line, Dogs, Enzyme Inhibitors pharmacology, Humans, Influenza A virus enzymology, Influenza A virus genetics, Influenza, Human virology, Models, Molecular, Neuraminidase metabolism, Orthomyxoviridae Infections drug therapy, Orthomyxoviridae Infections virology, Point Mutation, Viral Proteins metabolism, Antiviral Agents pharmacology, Drug Resistance, Viral, Influenza A virus drug effects, Influenza, Human drug therapy, Neuraminidase genetics, Oseltamivir pharmacology, Viral Proteins genetics
- Abstract
The therapeutic benefits of the neuraminidase (NA) inhibitor oseltamivir are dampened by the emergence of drug resistance mutations in influenza A virus (IAV). To investigate the mechanistic features that underlie resistance, we developed an approach to quantify the effects of all possible single-nucleotide substitutions introduced into important regions of NA. We determined the experimental fitness effects of 450 nucleotide mutations encoding positions both surrounding the active site and at more distant sites in an N1 strain of IAV in the presence and absence of oseltamivir. NA mutations previously known to confer oseltamivir resistance in N1 strains, including H275Y and N295S, were adaptive in the presence of drug, indicating that our experimental system captured salient features of real-world selection pressures acting on NA. We identified mutations, including several at position 223, that reduce the apparent affinity for oseltamivir in vitro. Position 223 of NA is located adjacent to a hydrophobic portion of oseltamivir that is chemically distinct from the substrate, making it a hotspot for substitutions that preferentially impact drug binding relative to substrate processing. Furthermore, two NA mutations, K221N and Y276F, each reduce susceptibility to oseltamivir by increasing NA activity without altering drug binding. These results indicate that competitive expansion of IAV in the face of drug pressure is mediated by a balance between inhibitor binding and substrate processing., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
34. Uveitis and Systemic Inflammatory Markers in Convalescent Phase of Ebola Virus Disease.
- Author
-
Chancellor JR, Padmanabhan SP, Greenough TC, Sacra R, Ellison RT 3rd, Madoff LC, Droms RJ, Hinkle DM, Asdourian GK, Finberg RW, Stroher U, Uyeki TM, and Cerón OM
- Subjects
- Hemorrhagic Fever, Ebola diagnosis, Humans, Liberia, Male, Tomography, Optical Coherence, Uveitis drug therapy, Ebolavirus genetics, Hemorrhagic Fever, Ebola complications, Hemorrhagic Fever, Ebola virology, Uveitis diagnosis, Uveitis etiology
- Abstract
We report a case of probable Zaire Ebola virus-related ophthalmologic complications in a physician from the United States who contracted Ebola virus disease in Liberia. Uveitis, immune activation, and nonspecific increase in antibody titers developed during convalescence. This case highlights immune phenomena that could complicate management of Ebola virus disease-related uveitis during convalescence.
- Published
- 2016
- Full Text
- View/download PDF
35. Influenza A virus preferentially snatches noncoding RNA caps.
- Author
-
Gu W, Gallagher GR, Dai W, Liu P, Li R, Trombly MI, Gammon DB, Mello CC, Wang JP, and Finberg RW
- Subjects
- Base Sequence, Cell Line, Tumor, Gene Expression Regulation, Viral, Genes, Viral, Humans, Influenza A Virus, H1N1 Subtype metabolism, RNA Processing, Post-Transcriptional, RNA, Messenger genetics, RNA, Messenger metabolism, RNA, Viral genetics, RNA, Viral metabolism, Influenza A Virus, H1N1 Subtype genetics, RNA Caps metabolism
- Abstract
Influenza A virus (IAV) lacks the enzyme for adding 5' caps to its RNAs and snatches the 5' ends of host capped RNAs to prime transcription. Neither the preference of the host RNA sequences snatched nor the effect of cap-snatching on host processes is completely defined. Previous studies of influenza cap-snatching used poly(A)-selected RNAs from infected cells or relied on annotated host genes to define the snatched host RNAs, and thus lack details on many noncoding host RNAs including snRNAs, snoRNAs, and promoter-associated capped small (cs)RNAs, which are made by "paused" Pol II during transcription initiation. In this study, we used a nonbiased technique, CapSeq, to identify host and viral-capped RNAs including nonpolyadenylated RNAs in the same samples, and investigated the substrate-product correlation between the host RNAs and the viral RNAs. We demonstrated that noncoding host RNAs, particularly U1 and U2, are the preferred cap-snatching source over mRNAs or pre-mRNAs. We also found that csRNAs are highly snatched by IAV. Because the functions of csRNAs remain mostly unknown, especially in somatic cells, our finding reveals that csRNAs at least play roles in the process of IAV infection. Our findings support a model where nascent RNAs including csRNAs are the preferred targets for cap-snatching by IAV and raise questions about how IAV might use snatching preferences to modulate host-mRNA splicing and transcription., (© 2015 Gu et al.; Published by Cold Spring Harbor Laboratory Press for the RNA Society.)
- Published
- 2015
- Full Text
- View/download PDF
36. Positive Selection Drives Preferred Segment Combinations during Influenza Virus Reassortment.
- Author
-
Zeldovich KB, Liu P, Renzette N, Foll M, Pham ST, Venev SV, Gallagher GR, Bolon DN, Kurt-Jones EA, Jensen JD, Caffrey DR, Schiffer CA, Kowalik TF, Wang JP, and Finberg RW
- Subjects
- Animals, Computational Biology, Dogs, Evolution, Molecular, Gene Frequency, Genotype, Hemagglutinin Glycoproteins, Influenza Virus genetics, High-Throughput Nucleotide Sequencing, Influenza A Virus, H1N1 Subtype genetics, Influenza A Virus, H1N2 Subtype genetics, Influenza A Virus, H3N2 Subtype genetics, Limit of Detection, Madin Darby Canine Kidney Cells, Nucleoproteins genetics, Sequence Analysis, RNA, Genome, Viral, Alphainfluenzavirus genetics, Reassortant Viruses genetics, Selection, Genetic
- Abstract
Influenza A virus (IAV) has a segmented genome that allows for the exchange of genome segments between different strains. This reassortment accelerates evolution by breaking linkage, helping IAV cross species barriers to potentially create highly virulent strains. Challenges associated with monitoring the process of reassortment in molecular detail have limited our understanding of its evolutionary implications. We applied a novel deep sequencing approach with quantitative analysis to assess the in vitro temporal evolution of genomic reassortment in IAV. The combination of H1N1 and H3N2 strains reproducibly generated a new H1N2 strain with the hemagglutinin and nucleoprotein segments originating from H1N1 and the remaining six segments from H3N2. By deep sequencing the entire viral genome, we monitored the evolution of reassortment, quantifying the relative abundance of all IAV genome segments from the two parent strains over time and measuring the selection coefficients of the reassorting segments. Additionally, we observed several mutations coemerging with reassortment that were not found during passaging of pure parental IAV strains. Our results demonstrate how reassortment of the segmented genome can accelerate viral evolution in IAV, potentially enabled by the emergence of a small number of individual mutations., (© The Author 2015. Published by Oxford University Press on behalf of the Society for Molecular Biology and Evolution. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2015
- Full Text
- View/download PDF
37. Heparin octasaccharide decoy liposomes inhibit replication of multiple viruses.
- Author
-
Hendricks GL, Velazquez L, Pham S, Qaisar N, Delaney JC, Viswanathan K, Albers L, Comolli JC, Shriver Z, Knipe DM, Kurt-Jones EA, Fygenson DK, Trevejo JM, Wang JP, and Finberg RW
- Subjects
- Animals, Antiviral Agents administration & dosage, Antiviral Agents chemistry, Heparitin Sulfate administration & dosage, Parainfluenza Virus 3, Human growth & development, Respiratory Syncytial Viruses growth & development, Simplexvirus growth & development, Vero Cells, Antiviral Agents pharmacology, Heparitin Sulfate pharmacology, Liposomes, Parainfluenza Virus 3, Human drug effects, Respiratory Syncytial Viruses drug effects, Simplexvirus drug effects, Virus Replication drug effects
- Abstract
Heparan sulfate (HS) is a ubiquitous glycosaminoglycan that serves as a cellular attachment site for a number of significant human pathogens, including respiratory syncytial virus (RSV), human parainfluenza virus 3 (hPIV3), and herpes simplex virus (HSV). Decoy receptors can target pathogens by binding to the receptor pocket on viral attachment proteins, acting as 'molecular sinks' and preventing the pathogen from binding to susceptible host cells. Decoy receptors functionalized with HS could bind to pathogens and prevent infection, so we generated decoy liposomes displaying HS-octasaccharide (HS-octa). These decoy liposomes significantly inhibited RSV, hPIV3, and HSV infectivity in vitro to a greater degree than the original HS-octa building block. The degree of inhibition correlated with the density of HS-octa displayed on the liposome surface. Decoy liposomes with HS-octa inhibited infection of viruses to a greater extent than either full-length heparin or HS-octa alone. Decoy liposomes were effective when added prior to infection or following the initial infection of cells in vitro. By targeting the well-conserved receptor-binding sites of HS-binding viruses, decoy liposomes functionalized with HS-octa are a promising therapeutic antiviral agent and illustrate the utility of the liposome delivery platform., (Copyright © 2015 Elsevier B.V. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
38. Viral infection of engrafted human islets leads to diabetes.
- Author
-
Gallagher GR, Brehm MA, Finberg RW, Barton BA, Shultz LD, Greiner DL, Bortell R, and Wang JP
- Subjects
- Animals, Diabetes Mellitus, Type 1 immunology, Enterovirus B, Human, Humans, Hyperglycemia immunology, Islets of Langerhans immunology, Mice, Coxsackievirus Infections immunology, Diabetes Mellitus, Type 1 virology, Hyperglycemia virology, Islets of Langerhans virology, Islets of Langerhans Transplantation
- Abstract
Type 1 diabetes (T1D) is characterized by the destruction of the insulin-producing β-cells of pancreatic islets. Genetic and environmental factors both contribute to T1D development. Viral infection with enteroviruses is a suspected trigger for T1D, but a causal role remains unproven and controversial. Studies in animals are problematic because of species-specific differences in host cell susceptibility and immune responses to candidate viral pathogens such as coxsackievirus B (CVB). In order to resolve the controversial role of viruses in human T1D, we developed a viral infection model in immunodeficient mice bearing human islet grafts. Hyperglycemia was induced in mice by specific ablation of native β-cells. Human islets, which are naturally susceptible to CVB infection, were transplanted to restore normoglycemia. Transplanted mice were infected with CVB4 and monitored for hyperglycemia. Forty-seven percent of CVB4-infected mice developed hyperglycemia. Human islet grafts from infected mice contained viral RNA, expressed viral protein, and had reduced insulin levels compared with grafts from uninfected mice. Human-specific gene expression profiles in grafts from infected mice revealed the induction of multiple interferon-stimulated genes. Thus, human islets can become severely dysfunctional with diminished insulin production after CVB infection of β-cells, resulting in diabetes., (© 2015 by the American Diabetes Association. Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered.)
- Published
- 2015
- Full Text
- View/download PDF
39. A chimeric EBV gp350/220-based VLP replicates the virion B-cell attachment mechanism and elicits long-lasting neutralizing antibodies in mice.
- Author
-
Ogembo JG, Muraswki MR, McGinnes LW, Parcharidou A, Sutiwisesak R, Tison T, Avendano J, Agnani D, Finberg RW, Morrison TG, and Fingeroth JD
- Subjects
- Animals, Antigens, CD metabolism, Cell Adhesion, Cell Line, Humans, Immunization, Immunoglobulin G metabolism, Mice, Inbred BALB C, Neutralization Tests, Protein Binding, Antibodies, Neutralizing biosynthesis, B-Lymphocytes cytology, Recombinant Proteins metabolism, Viral Matrix Proteins immunology, Virion metabolism
- Abstract
Epstein-Barr virus (EBV), an oncogenic gammaherpesvirus, causes acute infectious mononucleosis (AIM) and is linked to the development of several human malignancies. There is an urgent need for a vaccine that is safe, prevents infection and/or limits disease. Unique among human herpesviruses, glycoprotein (gp)350/220, which initiates EBV attachment to susceptible host cells, is the major ligand on the EBV envelope and is highly conserved. Interaction between gp350/220 and complement receptor type 2 (CR2)/CD21 and/or (CR1)/CD35 on B-cells is required for infection. Potent antibody responses to gp350/220 occur in animal models and humans. Thus, gp350/220 provides an attractive candidate for prophylactic subunit vaccine development. However, in a recent Phase II clinical trial immunization with soluble recombinant gp350 reduced the incidence of AIM, but did not prevent infection. Despite various attempts to produce an EBV vaccine, no vaccine is licensed. Herein we describe a sub-unit vaccine against EBV based on a novel Newcastle disease virus (NDV)-virus-like particle (VLP) platform consisting of EBVgp350/220 ectodomain fused to NDV-fusion (F) protein. The chimeric protein EBVgp350/220-F is incorporated into the membrane of a VLP composed of the NDV matrix and nucleoprotein. The particles resemble native EBV in diameter and shape and bind CD21 and CD35. Immunization of BALB/c mice with EBVgp350/220-F VLPs elicited strong, long-lasting neutralizing antibody responses when assessed in vitro. This chimeric VLP is predicted to provide a superior safety profile as it is efficiently produced in Chinese hamster ovary (CHO) cells using a platform devoid of human nucleic acid and EBV-transforming genes.
- Published
- 2015
- Full Text
- View/download PDF
40. Increased survival in B-cell-deficient mice during experimental cerebral malaria suggests a role for circulating immune complexes.
- Author
-
de Oliveira RB, Wang JP, Ram S, Gazzinelli RT, Finberg RW, and Golenbock DT
- Subjects
- Animals, Female, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Receptors, Complement 3b genetics, Receptors, Complement 3b immunology, Survival Analysis, Antigen-Antibody Complex immunology, Antigen-Antibody Complex toxicity, B-Lymphocytes immunology, Malaria, Cerebral immunology, Malaria, Cerebral pathology, Plasmodium berghei immunology
- Abstract
The pathogenesis of malaria, an insect-borne disease that takes millions of lives every year, is still not fully understood. Complement receptor 1 (CR1) has been described as a receptor for Plasmodium falciparum, which causes cerebral malaria in humans. We investigated the role of CR1 in an experimental model of cerebral malaria. Transgenic mice expressing human CR1 (hCR1(+)) on erythrocytes were infected with Plasmodium berghei ANKA and developed cerebral malaria. No difference in survival was observed in hCR1(+) mice compared to wild-type mice following infection with P. berghei ANKA; however, hCR1 detection was significantly diminished on erythrocytes between days 7 and 10 postinfection. hCR1 levels returned to baseline by day 17 postinfection in surviving animals. Immunoblot assays revealed that total erythrocyte hCR1 levels were diminished, confirming that immune complexes in association with erythrocyte hCR1 were likely removed from erythrocytes in vivo by clearance following immune adherence. Decreases in hCR1 were completely dependent on C3 expression, as mice treated with cobra venom factor (which consumes and depletes C3) retained hCR1 on erythrocytes during C3 depletion through day 7; erythrocyte hCR1 decreases were observed only when C3 levels recovered on day 9. B-cell-deficient mice exhibit a marked increase in survival following infection with P. berghei ANKA, which suggests that immune complexes play a central role in the pathogenesis of experimental cerebral malaria. Together, our findings highlight the importance of complement and immune complexes in experimental cerebral malaria. IMPORTANCE Cerebral malaria is a deadly complication of infection with Plasmodium falciparum. Despite its high prevalence, relatively little is understood about its pathogenesis. We have determined that immune complexes are generated and deposited on erythrocytes specifically expressing human complement receptor 1 in a mouse model of cerebral malaria. We also provide evidence demonstrating the importance of immunoglobulins in the pathogenesis of cerebral malaria in mice. These findings may have important implications in human cerebral malaria.
- Published
- 2014
- Full Text
- View/download PDF
41. Influenza virus drug resistance: a time-sampled population genetics perspective.
- Author
-
Foll M, Poh YP, Renzette N, Ferrer-Admetlla A, Bank C, Shim H, Malaspinas AS, Ewing G, Liu P, Wegmann D, Caffrey DR, Zeldovich KB, Bolon DN, Wang JP, Kowalik TF, Schiffer CA, Finberg RW, and Jensen JD
- Subjects
- Bayes Theorem, Genetic Drift, Humans, Influenza A Virus, H1N1 Subtype drug effects, Influenza, Human genetics, Influenza, Human virology, Mutation, Oseltamivir pharmacology, Drug Resistance, Viral genetics, Genetics, Population, Influenza A Virus, H1N1 Subtype genetics, Selection, Genetic
- Abstract
The challenge of distinguishing genetic drift from selection remains a central focus of population genetics. Time-sampled data may provide a powerful tool for distinguishing these processes, and we here propose approximate Bayesian, maximum likelihood, and analytical methods for the inference of demography and selection from time course data. Utilizing these novel statistical and computational tools, we evaluate whole-genome datasets of an influenza A H1N1 strain in the presence and absence of oseltamivir (an inhibitor of neuraminidase) collected at thirteen time points. Results reveal a striking consistency amongst the three estimation procedures developed, showing strongly increased selection pressure in the presence of drug treatment. Importantly, these approaches re-identify the known oseltamivir resistance site, successfully validating the approaches used. Enticingly, a number of previously unknown variants have also been identified as being positively selected. Results are interpreted in the light of Fisher's Geometric Model, allowing for a quantification of the increased distance to optimum exerted by the presence of drug, and theoretical predictions regarding the distribution of beneficial fitness effects of contending mutations are empirically tested. Further, given the fit to expectations of the Geometric Model, results suggest the ability to predict certain aspects of viral evolution in response to changing host environments and novel selective pressures.
- Published
- 2014
- Full Text
- View/download PDF
42. Evolution of the influenza A virus genome during development of oseltamivir resistance in vitro.
- Author
-
Renzette N, Caffrey DR, Zeldovich KB, Liu P, Gallagher GR, Aiello D, Porter AJ, Kurt-Jones EA, Bolon DN, Poh YP, Jensen JD, Schiffer CA, Kowalik TF, Finberg RW, and Wang JP
- Subjects
- Animals, Cell Line, Dogs, High-Throughput Screening Assays, In Vitro Techniques, Influenza A Virus, H1N1 Subtype growth & development, Inhibitory Concentration 50, Mutation, Viral Plaque Assay, Antiviral Agents pharmacology, Drug Resistance, Viral genetics, Evolution, Molecular, Genome, Viral, Influenza A Virus, H1N1 Subtype genetics, Oseltamivir pharmacology
- Abstract
Influenza A virus (IAV) is a major cause of morbidity and mortality throughout the world. Current antiviral therapies include oseltamivir, a neuraminidase inhibitor that prevents the release of nascent viral particles from infected cells. However, the IAV genome can evolve rapidly, and oseltamivir resistance mutations have been detected in numerous clinical samples. Using an in vitro evolution platform and whole-genome population sequencing, we investigated the population genomics of IAV during the development of oseltamivir resistance. Strain A/Brisbane/59/2007 (H1N1) was grown in Madin-Darby canine kidney cells with or without escalating concentrations of oseltamivir over serial passages. Following drug treatment, the H274Y resistance mutation fixed reproducibly within the population. The presence of the H274Y mutation in the viral population, at either a low or a high frequency, led to measurable changes in the neuraminidase inhibition assay. Surprisingly, fixation of the resistance mutation was not accompanied by alterations of viral population diversity or differentiation, and oseltamivir did not alter the selective environment. While the neighboring K248E mutation was also a target of positive selection prior to H274Y fixation, H274Y was the primary beneficial mutation in the population. In addition, once evolved, the H274Y mutation persisted after the withdrawal of the drug, even when not fixed in viral populations. We conclude that only selection of H274Y is required for oseltamivir resistance and that H274Y is not deleterious in the absence of the drug. These collective results could offer an explanation for the recent reproducible rise in oseltamivir resistance in seasonal H1N1 IAV strains in humans.
- Published
- 2014
- Full Text
- View/download PDF
43. Tuning innate immune activation by surface texturing of polymer microparticles: the role of shape in inflammasome activation.
- Author
-
Vaine CA, Patel MK, Zhu J, Lee E, Finberg RW, Hayward RC, and Kurt-Jones EA
- Subjects
- Animals, Carrier Proteins metabolism, Humans, Interleukin-1 metabolism, Interleukin-1beta metabolism, Macrophages immunology, Membrane Microdomains metabolism, Mice, Mice, Knockout, NLR Family, Pyrin Domain-Containing 3 Protein, Neutrophils immunology, Neutrophils metabolism, Particle Size, Phagocytosis immunology, Signal Transduction, Surface Properties, Immunity, Innate, Inflammasomes immunology, Polymers chemistry
- Abstract
Polymeric microparticles have been widely investigated as platforms for delivery of drugs, vaccines, and imaging contrast agents and are increasingly used in a variety of clinical applications. Microparticles activate the inflammasome complex and induce the processing and secretion of IL-1β, a key innate immune cytokine. Recent work suggests that although receptors are clearly important for particle phagocytosis, other physical characteristics, especially shape, play an important role in the way microparticles activate cells. We examined the role of particle surface texturing not only on uptake efficiency but also on the subsequent immune cell activation of the inflammasome. Using a method based on emulsion processing of amphiphilic block copolymers, we prepared microparticles with similar overall sizes and surface chemistries but having either smooth or highly microtextured surfaces. In vivo, textured (budding) particles induced more rapid neutrophil recruitment to the injection site. In vitro, budding particles were more readily phagocytosed than smooth particles and induced more lipid raft recruitment to the phagosome. Remarkably, budding particles also induced stronger IL-1β secretion than smooth particles through activation of the NLRP3 inflammasome. These findings demonstrate a pronounced role of particle surface topography in immune cell activation, suggesting that shape is a major determinant of inflammasome activation.
- Published
- 2013
- Full Text
- View/download PDF
44. Human monoclonal antibody MBL-HCV1 delays HCV viral rebound following liver transplantation: a randomized controlled study.
- Author
-
Chung RT, Gordon FD, Curry MP, Schiano TD, Emre S, Corey K, Markmann JF, Hertl M, Pomposelli JJ, Pomfret EA, Florman S, Schilsky M, Broering TJ, Finberg RW, Szabo G, Zamore PD, Khettry U, Babcock GJ, Ambrosino DM, Leav B, Leney M, Smith HL, and Molrine DC
- Subjects
- Aged, Biopsy, Double-Blind Method, Female, Genotype, Hepatitis C virology, Humans, Liver pathology, Male, Middle Aged, Pilot Projects, RNA, Viral analysis, Time Factors, Viral Envelope Proteins immunology, Antibodies, Monoclonal pharmacology, Hepacivirus physiology, Hepatitis C drug therapy, Liver Transplantation
- Abstract
Rapid allograft infection complicates liver transplantation (LT) in patients with hepatitis C virus (HCV). Pegylated interferon-α and ribavirin therapy after LT has significant toxicity and limited efficacy. The effect of a human monoclonal antibody targeting the HCV E2 glycoprotein (MBL-HCV1) on viral clearance was examined in a randomized, double-blind, placebo-controlled pilot study in patients infected with HCV genotype 1a undergoing LT. Subjects received 11 infusions of 50 mg/kg MBL-HCV1 (n=6) or placebo (n=5) intravenously with three infusions on day of transplant, a single infusion on days 1 through 7 and one infusion on day 14 after LT. MBL-HCV1 was well-tolerated and reduced viral load for a period ranging from 7 to 28 days. Median change in viral load (log10 IU/mL) from baseline was significantly greater (p=0.02) for the antibody-treated group (range -3.07 to -3.34) compared to placebo group (range -0.331 to -1.01) on days 3 through 6 posttransplant. MBL-HCV1 treatment significantly delayed median time to viral rebound compared to placebo treatment (18.7 days vs. 2.4 days, p<0.001). As with other HCV monotherapies, antibody-treated subjects had resistance-associated variants at the time of viral rebound. A combination study of MBL-HCV1 with a direct-acting antiviral is underway., (© Copyright 2013 The American Society of Transplantation and the American Society of Transplant Surgeons.)
- Published
- 2013
- Full Text
- View/download PDF
45. Sialylneolacto-N-tetraose c (LSTc)-bearing liposomal decoys capture influenza A virus.
- Author
-
Hendricks GL, Weirich KL, Viswanathan K, Li J, Shriver ZH, Ashour J, Ploegh HL, Kurt-Jones EA, Fygenson DK, Finberg RW, Comolli JC, and Wang JP
- Subjects
- Animals, Antiviral Agents administration & dosage, Cell Line, Chlorocebus aethiops, Dogs, Drug Evaluation, Preclinical, Epithelial Cells drug effects, Epithelial Cells virology, Female, Hemagglutination drug effects, Humans, Influenza A virus physiology, Liposomes, Mice, Mice, Inbred C57BL, Polysaccharides administration & dosage, Rous sarcoma virus drug effects, Sendai virus drug effects, Sialic Acids administration & dosage, Vero Cells, Virus Replication drug effects, Antiviral Agents pharmacology, Influenza A virus drug effects, Influenza, Human drug therapy, Polysaccharides pharmacology, Sialic Acids pharmacology
- Abstract
Influenza is a severe disease in humans and animals with few effective therapies available. All strains of influenza virus are prone to developing drug resistance due to the high mutation rate in the viral genome. A therapeutic agent that targets a highly conserved region of the virus could bypass resistance and also be effective against multiple strains of influenza. Influenza uses many individually weak ligand binding interactions for a high avidity multivalent attachment to sialic acid-bearing cells. Polymerized sialic acid analogs can form multivalent interactions with influenza but are not ideal therapeutics due to solubility and toxicity issues. We used liposomes as a novel means for delivery of the glycan sialylneolacto-N-tetraose c (LSTc). LSTc-bearing decoy liposomes form multivalent, polymer-like interactions with influenza virus. Decoy liposomes competitively bind influenza virus in hemagglutination inhibition assays and inhibit infection of target cells in a dose-dependent manner. Inhibition is specific for influenza virus, as inhibition of Sendai virus and respiratory syncytial virus is not observed. In contrast, monovalent LSTc does not bind influenza virus or inhibit infectivity. LSTc decoy liposomes prevent the spread of influenza virus during multiple rounds of replication in vitro and extend survival of mice challenged with a lethal dose of virus. LSTc decoy liposomes co-localize with fluorescently tagged influenza virus, whereas control liposomes do not. Considering the conservation of the hemagglutinin binding pocket and the ability of decoy liposomes to form high avidity interactions with influenza hemagglutinin, our decoy liposomes have potential as a new therapeutic agent against emerging influenza strains.
- Published
- 2013
- Full Text
- View/download PDF
46. Dephosphorylation of the RNA sensors RIG-I and MDA5 by the phosphatase PP1 is essential for innate immune signaling.
- Author
-
Wies E, Wang MK, Maharaj NP, Chen K, Zhou S, Finberg RW, and Gack MU
- Subjects
- Animals, Cell Line, Cells, Cultured, Chlorocebus aethiops, DEAD Box Protein 58, DEAD-box RNA Helicases genetics, DEAD-box RNA Helicases metabolism, HEK293 Cells, HeLa Cells, Humans, Immunity, Innate genetics, Immunoblotting, Interferon-Induced Helicase, IFIH1, Interferon-beta immunology, Interferon-beta metabolism, Mice, Mice, Knockout, Microscopy, Confocal, Molecular Sequence Data, Mutation, Phosphorylation, Protein Phosphatase 1 genetics, Protein Phosphatase 1 metabolism, RNA Interference, RNA, Viral genetics, RNA, Viral metabolism, Receptors, Immunologic, Signal Transduction genetics, Signal Transduction immunology, Vero Cells, DEAD-box RNA Helicases immunology, Immunity, Innate immunology, Protein Phosphatase 1 immunology, RNA, Viral immunology
- Abstract
RIG-I and MDA5 have emerged as key cytosolic sensors for the detection of RNA viruses and lead to antiviral interferon (IFN) production. Recent studies have highlighted the importance of posttranslational modifications for controlling RIG-I antiviral activity. However, the regulation of MDA5 signal-transducing ability remains unclear. Here, we show that MDA5 signaling activity is regulated by a dynamic balance between phosphorylation and dephosphorylation of its caspase recruitment domains (CARDs). Employing a phosphatome RNAi screen, we identified PP1α and PP1γ as the primary phosphatases that are responsible for MDA5 and RIG-I dephosphorylation and that lead to their activation. Silencing of PP1α and PP1γ enhanced RIG-I and MDA5 CARD phosphorylation and reduced antiviral IFN-β production. PP1α- and PP1γ-depleted cells were impaired in their ability to induce IFN-stimulated gene expression, which resulted in enhanced RNA virus replication. This work identifies PP1α and PP1γ as regulators of antiviral innate immune responses to various RNA viruses, including influenza virus, paramyxovirus, dengue virus, and picornavirus., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
47. Human complement receptor type 1/CD35 is an Epstein-Barr Virus receptor.
- Author
-
Ogembo JG, Kannan L, Ghiran I, Nicholson-Weller A, Finberg RW, Tsokos GC, and Fingeroth JD
- Subjects
- Antigens, CD19 metabolism, Cell Line, Humans, K562 Cells, Mutation, Precursor Cells, B-Lymphoid immunology, Precursor Cells, B-Lymphoid metabolism, Protein Binding, Receptors, Complement 3b genetics, Receptors, Complement 3d genetics, Temperature, Transfection, Viral Matrix Proteins metabolism, Virus Attachment, Virus Internalization, Herpesvirus 4, Human metabolism, Receptors, Complement 3b metabolism, Receptors, Complement 3d metabolism
- Abstract
Epstein-Barr virus (EBV) attachment to primary B cells initiates virus entry. Although CD21 is the only known receptor for EBVgp350/220, a recent report documents EBV-infected B cells from a patient genetically deficient in CD21. On normal resting B cells, CD21 forms two membrane complexes: one with CD19 and another with CD35. Whereas the CD21/CD19 complex is widely retained on immortalized and B cell tumor lines, the related complement-regulatory protein CD35 is lost. To determine the role(s) of CD35 in initial infection, we transduced a CD21-negative pre-B cell and myeloid leukemia line with CD35, CD21, or both. Cells expressing CD35 alone bound gp350/220 and became latently infected when the fusion receptor HLA II was coexpressed. Temporal, biophysical, and structural characteristics of CD35-mediated infection were distinct from CD21. Identification of CD35 as an EBV receptor uncovers a salient role in primary infection, addresses unsettled questions of virus tropism, and underscores the importance of EBVgp350/220 for vaccine development., (Copyright © 2013 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
48. Phomopsis bougainvilleicola prepatellar bursitis in a renal transplant recipient.
- Author
-
Cariello PF, Wickes BL, Sutton DA, Castlebury LA, Levitz SM, Finberg RW, Thompson EH, and Daly JS
- Subjects
- Bursitis diagnosis, DNA, Bacterial, Humans, Magnetic Resonance Imaging, Male, Middle Aged, Molecular Sequence Data, Mycoses diagnosis, Phylogeny, Ultrasonography, Ascomycota classification, Ascomycota genetics, Ascomycota growth & development, Bursitis microbiology, Kidney Transplantation adverse effects, Knee Injuries microbiology, Mycoses microbiology
- Abstract
Prepatellar bursitis is typically a monomicrobial bacterial infection. A fungal cause is rarely identified. We describe a 61-year-old man who had received a renal transplant 21 months prior to presentation whose synovial fluid and surgical specimens grew Phomopsis bougainvilleicola, a pycnidial coelomycete.
- Published
- 2013
- Full Text
- View/download PDF
49. Role of interferon regulatory factor 7 in T cell responses during acute lymphocytic choriomeningitis virus infection.
- Author
-
Zhou S, Cerny AM, Fitzgerald KA, Kurt-Jones EA, and Finberg RW
- Subjects
- Adaptive Immunity, Animals, CD4-Positive T-Lymphocytes virology, CD8-Positive T-Lymphocytes virology, Immunity, Innate, Interferon Regulatory Factor-7 genetics, Lymphocyte Activation, Lymphocytic Choriomeningitis virology, Mice, Mice, Inbred C57BL, Mice, Knockout, Signal Transduction, Virus Replication, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Interferon Regulatory Factor-7 immunology, Interferon Type I immunology, Lymphocytic Choriomeningitis immunology, Lymphocytic choriomeningitis virus immunology
- Abstract
Type I interferons (IFNs), predominantly IFN-α and -β, play critical roles in both innate and adaptive immune responses against viral infections. Interferon regulatory factor 7 (IRF7), a key innate immune molecule in the type I IFN signaling pathway, is essential for the type I IFN response to many viruses, including lymphocytic choriomeningitis virus (LCMV). Here, we show that although IRF7 knockout (KO) mice failed to control the replication of LCMV in the early stages of infection, they were capable of clearing LCMV infection. Despite the lack of type I IFN production, IRF7 KO mice generated normal CD4(+) T cell responses, and the expansion of naïve CD8(+) T cells into primary CD8(+) T cells specific for LCMV GP(33-41) was relatively normal. In contrast, the expansion of the LCMV NP(396)-specific CD8(+) T cells was severely impaired in IRF7 KO mice. We demonstrated that this defective CD8(+) T cell response is due neither to an impaired antigen-presenting system nor to any intrinsic role of IRF7 in CD8(+) T cells. The lack of a type I IFN response in IRF7 KO mice did not affect the formation of memory CD8(+) T cells. Thus, the present study provides new insight into the impact of the innate immune system on viral pathogenesis and demonstrates the critical contribution of innate immunity in controlling virus replication in the early stages of infection, which may shape the quality of CD8(+) T cell responses.
- Published
- 2012
- Full Text
- View/download PDF
50. Role of specific innate immune responses in herpes simplex virus infection of the central nervous system.
- Author
-
Wang JP, Bowen GN, Zhou S, Cerny A, Zacharia A, Knipe DM, Finberg RW, and Kurt-Jones EA
- Subjects
- Animals, Central Nervous System virology, Herpes Simplex virology, Herpesvirus 1, Human immunology, Humans, Interferon Type I immunology, Mice, Mice, Inbred C57BL, Receptor, Interferon alpha-beta immunology, Toll-Like Receptors immunology, Central Nervous System immunology, Herpes Simplex immunology, Herpesvirus 1, Human physiology, Immunity, Innate
- Abstract
Herpes simplex virus 1 (HSV-1) causes a spectrum of disease, including herpes labialis, herpes keratitis, and herpes encephalitis, which can be lethal. Viral recognition by pattern recognition receptors plays a central role in cytokine production and in the generation of antiviral immunity. The relative contributions of different Toll-like receptors (TLRs) in the innate immune response during central nervous system infection with HSV-1 have not been fully characterized. In this study, we investigate the roles of TLR2, TLR9, UNC93B1, and the type I interferon (IFN) receptor in a murine model of HSV-1 encephalitis. TLR2 is responsible for detrimental inflammatory cytokine production following intracranial infection with HSV-1, and the absence of TLR2 expression leads to increased survival in mice. We prove that inflammatory cytokine production by microglial cells, astrocytes, neutrophils, and monocytes is mediated predominantly by TLR2. We also demonstrate that type I IFNs are absolutely required for survival following intracranial HSV-1 infection, as mice lacking the type I IFN receptor succumb rapidly following infection and have high levels of HSV in the brain. However, the absence of TLR9 does not impact survival, type I IFN levels, or viral replication in the brain following infection. The absence of UNC93B1 leads to a survival disadvantage but does not impact viral replication or type I IFN levels in the brain in HSV-1-infected mice. These results illustrate the complex but important roles that innate immune receptors play in host responses to HSV-1 during infection of the central nervous system.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.