483 results on '"Albrecht, Randy A."'
Search Results
2. Proteomic and genetic analyses of influenza A viruses identify pan-viral host targets
- Author
-
Haas, Kelsey M, McGregor, Michael J, Bouhaddou, Mehdi, Polacco, Benjamin J, Kim, Eun-Young, Nguyen, Thong T, Newton, Billy W, Urbanowski, Matthew, Kim, Heejin, Williams, Michael AP, Rezelj, Veronica V, Hardy, Alexandra, Fossati, Andrea, Stevenson, Erica J, Sukerman, Ellie, Kim, Tiffany, Penugonda, Sudhir, Moreno, Elena, Braberg, Hannes, Zhou, Yuan, Metreveli, Giorgi, Harjai, Bhavya, Tummino, Tia A, Melnyk, James E, Soucheray, Margaret, Batra, Jyoti, Pache, Lars, Martin-Sancho, Laura, Carlson-Stevermer, Jared, Jureka, Alexander S, Basler, Christopher F, Shokat, Kevan M, Shoichet, Brian K, Shriver, Leah P, Johnson, Jeffrey R, Shaw, Megan L, Chanda, Sumit K, Roden, Dan M, Carter, Tonia C, Kottyan, Leah C, Chisholm, Rex L, Pacheco, Jennifer A, Smith, Maureen E, Schrodi, Steven J, Albrecht, Randy A, Vignuzzi, Marco, Zuliani-Alvarez, Lorena, Swaney, Danielle L, Eckhardt, Manon, Wolinsky, Steven M, White, Kris M, Hultquist, Judd F, Kaake, Robyn M, García-Sastre, Adolfo, and Krogan, Nevan J
- Subjects
Microbiology ,Medical Microbiology ,Biomedical and Clinical Sciences ,Biological Sciences ,Infectious Diseases ,Prevention ,Emerging Infectious Diseases ,Vaccine Related ,Influenza ,Pneumonia & Influenza ,Biodefense ,Biotechnology ,2.1 Biological and endogenous factors ,2.2 Factors relating to the physical environment ,Aetiology ,Infection ,Humans ,Influenza A virus ,Influenza ,Human ,Influenza A Virus ,H5N1 Subtype ,Influenza A Virus ,H3N2 Subtype ,Proteomics ,Virus Replication ,COVID-19 ,SARS-CoV-2 ,Antiviral Agents ,Host-Pathogen Interactions - Abstract
Influenza A Virus (IAV) is a recurring respiratory virus with limited availability of antiviral therapies. Understanding host proteins essential for IAV infection can identify targets for alternative host-directed therapies (HDTs). Using affinity purification-mass spectrometry and global phosphoproteomic and protein abundance analyses using three IAV strains (pH1N1, H3N2, H5N1) in three human cell types (A549, NHBE, THP-1), we map 332 IAV-human protein-protein interactions and identify 13 IAV-modulated kinases. Whole exome sequencing of patients who experienced severe influenza reveals several genes, including scaffold protein AHNAK, with predicted loss-of-function variants that are also identified in our proteomic analyses. Of our identified host factors, 54 significantly alter IAV infection upon siRNA knockdown, and two factors, AHNAK and coatomer subunit COPB1, are also essential for productive infection by SARS-CoV-2. Finally, 16 compounds targeting our identified host factors suppress IAV replication, with two targeting CDK2 and FLT3 showing pan-antiviral activity across influenza and coronavirus families. This study provides a comprehensive network model of IAV infection in human cells, identifying functional host targets for pan-viral HDT.
- Published
- 2023
3. Author Correction: Discovery of SARS-CoV-2 antiviral drugs through large-scale compound repurposing
- Author
-
Riva, Laura, Yuan, Shuofeng, Yin, Xin, Martin-Sancho, Laura, Matsunaga, Naoko, Pache, Lars, Burgstaller-Muehlbacher, Sebastian, De Jesus, Paul D., Teriete, Peter, Hull, Mitchell V., Chang, Max W., Chan, Jasper Fuk-Woo, Cao, Jianli, Poon, Vincent Kwok-Man, Herbert, Kristina M., Cheng, Kuoyuan, Nguyen, Tu-Trinh H., Rubanov, Andrey, Pu, Yuan, Nguyen, Courtney, Choi, Angela, Rathnasinghe, Raveen, Schotsaert, Michael, Miorin, Lisa, Dejosez, Marion, Zwaka, Thomas P., Sit, Ko-Yung, Martinez-Sobrido, Luis, Liu, Wen-Chun, White, Kris M., Chapman, Mackenzie E., Lendy, Emma K., Glynne, Richard J., Albrecht, Randy, Ruppin, Eytan, Mesecar, Andrew D., Johnson, Jeffrey R., Benner, Christopher, Sun, Ren, Schultz, Peter G., Su, Andrew I., García-Sastre, Adolfo, Chatterjee, Arnab K., Yuen, Kwok-Yung, and Chanda, Sumit K.
- Published
- 2024
- Full Text
- View/download PDF
4. The IRE1α-XBP1 arm of the unfolded protein response is a host factor activated in SARS-CoV-2 infection
- Author
-
Fernández, Jose Javier, Marín, Arturo, Rosales, Romel, Penrice-Randal, Rebekah, Mlcochova, Petra, Alvarez, Yolanda, Villalón-Letelier, Fernando, Yildiz, Soner, Pérez, Enrique, Rathnasinghe, Raveen, Cupic, Anastasija, Kehrer, Thomas, Uccellini, Melissa B., Alonso, Sara, Martínez, Fernando, McGovern, Briana Lynn, Clark, Jordan J., Sharma, Parul, Bayón, Yolanda, Alonso, Andrés, Albrecht, Randy A., White, Kris M., Schotsaert, Michael, Miorin, Lisa, Stewart, James P., Hiscox, Julian A., Gupta, Ravindra K., Irigoyen, Nerea, García-Sastre, Adolfo, Crespo, Mariano Sánchez, and Fernández, Nieves
- Published
- 2024
- Full Text
- View/download PDF
5. Mutations in SARS-CoV-2 variants of concern link to increased spike cleavage and virus transmission
- Author
-
Escalera, Alba, Gonzalez-Reiche, Ana S, Aslam, Sadaf, Mena, Ignacio, Laporte, Manon, Pearl, Rebecca L, Fossati, Andrea, Rathnasinghe, Raveen, Alshammary, Hala, van de Guchte, Adriana, Farrugia, Keith, Qin, Yiren, Bouhaddou, Mehdi, Kehrer, Thomas, Zuliani-Alvarez, Lorena, Meekins, David A, Balaraman, Velmurugan, McDowell, Chester, Richt, Jürgen A, Bajic, Goran, Sordillo, Emilia Mia, Dejosez, Marion, Zwaka, Thomas P, Krogan, Nevan J, Simon, Viviana, Albrecht, Randy A, van Bakel, Harm, García-Sastre, Adolfo, and Aydillo, Teresa
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Emerging Infectious Diseases ,Lung ,Prevention ,Biodefense ,Vaccine Related ,Aetiology ,2.2 Factors relating to the physical environment ,Infection ,Good Health and Well Being ,COVID-19 ,Humans ,Mutation ,SARS-CoV-2 ,Spike Glycoprotein ,Coronavirus ,H655Y mutation ,fusion ,gamma ,omicron ,spike cleavage ,syncytia formation ,variants of concern ,Microbiology ,Immunology ,Biochemistry and cell biology ,Medical microbiology - Abstract
SARS-CoV-2 lineages have diverged into highly prevalent variants termed "variants of concern" (VOCs). Here, we characterized emerging SARS-CoV-2 spike polymorphisms in vitro and in vivo to understand their impact on transmissibility and virus pathogenicity and fitness. We demonstrate that the substitution S:655Y, represented in the gamma and omicron VOCs, enhances viral replication and spike protein cleavage. The S:655Y substitution was transmitted more efficiently than its ancestor S:655H in the hamster infection model and was able to outcompete S:655H in the hamster model and in a human primary airway system. Finally, we analyzed a set of emerging SARS-CoV-2 variants to investigate how different sets of mutations may impact spike processing. All VOCs tested exhibited increased spike cleavage and fusogenic capacity. Taken together, our study demonstrates that the spike mutations present in VOCs that become epidemiologically prevalent in humans are linked to an increase in spike processing and virus transmission.
- Published
- 2022
6. Restriction factor compendium for influenza A virus reveals a mechanism for evasion of autophagy
- Author
-
Martin-Sancho, Laura, Tripathi, Shashank, Rodriguez-Frandsen, Ariel, Pache, Lars, Sanchez-Aparicio, Maite, McGregor, Michael J, Haas, Kelsey M, Swaney, Danielle L, Nguyen, Thong T, Mamede, João I, Churas, Christopher, Pratt, Dexter, Rosenthal, Sara B, Riva, Laura, Nguyen, Courtney, Beltran-Raygoza, Nish, Soonthornvacharin, Stephen, Wang, Guojun, Jimenez-Morales, David, De Jesus, Paul D, Moulton, Hong M, Stein, David A, Chang, Max W, Benner, Chris, Ideker, Trey, Albrecht, Randy A, Hultquist, Judd F, Krogan, Nevan J, García-Sastre, Adolfo, and Chanda, Sumit K
- Subjects
Microbiology ,Biochemistry and Cell Biology ,Biological Sciences ,Biodefense ,Vaccine Related ,Prevention ,Influenza ,Emerging Infectious Diseases ,Infectious Diseases ,Pneumonia & Influenza ,2.2 Factors relating to the physical environment ,2.1 Biological and endogenous factors ,Aetiology ,Infection ,Antiviral Agents ,Autophagy ,GTPase-Activating Proteins ,Host-Pathogen Interactions ,Humans ,Immune Evasion ,Influenza A virus ,Lysosomes ,Protein Binding ,Viral Matrix Proteins ,Virus Replication ,rab GTP-Binding Proteins ,rab7 GTP-Binding Proteins ,Medical Microbiology - Abstract
The fate of influenza A virus (IAV) infection in the host cell depends on the balance between cellular defence mechanisms and viral evasion strategies. To illuminate the landscape of IAV cellular restriction, we generated and integrated global genetic loss-of-function screens with transcriptomics and proteomics data. Our multi-omics analysis revealed a subset of both IFN-dependent and independent cellular defence mechanisms that inhibit IAV replication. Amongst these, the autophagy regulator TBC1 domain family member 5 (TBC1D5), which binds Rab7 to enable fusion of autophagosomes and lysosomes, was found to control IAV replication in vitro and in vivo and to promote lysosomal targeting of IAV M2 protein. Notably, IAV M2 was observed to abrogate TBC1D5-Rab7 binding through a physical interaction with TBC1D5 via its cytoplasmic tail. Our results provide evidence for the molecular mechanism utilised by IAV M2 protein to escape lysosomal degradation and traffic to the cell membrane, where it supports IAV budding and growth.
- Published
- 2021
7. TOP1 inhibition therapy protects against SARS-CoV-2-induced lethal inflammation
- Author
-
Ho, Jessica Sook Yuin, Mok, Bobo Wing-Yee, Campisi, Laura, Jordan, Tristan, Yildiz, Soner, Parameswaran, Sreeja, Wayman, Joseph A, Gaudreault, Natasha N, Meekins, David A, Indran, Sabarish V, Morozov, Igor, Trujillo, Jessie D, Fstkchyan, Yesai S, Rathnasinghe, Raveen, Zhu, Zeyu, Zheng, Simin, Zhao, Nan, White, Kris, Ray-Jones, Helen, Malysheva, Valeriya, Thiecke, Michiel J, Lau, Siu-Ying, Liu, Honglian, Zhang, Anna Junxia, Lee, Andrew Chak-Yiu, Liu, Wen-Chun, Jangra, Sonia, Escalera, Alba, Aydillo, Teresa, Melo, Betsaida Salom, Guccione, Ernesto, Sebra, Robert, Shum, Elaine, Bakker, Jan, Kaufman, David A, Moreira, Andre L, Carossino, Mariano, Balasuriya, Udeni BR, Byun, Minji, Albrecht, Randy A, Schotsaert, Michael, Garcia-Sastre, Adolfo, Chanda, Sumit K, Miraldi, Emily R, Jeyasekharan, Anand D, TenOever, Benjamin R, Spivakov, Mikhail, Weirauch, Matthew T, Heinz, Sven, Chen, Honglin, Benner, Christopher, Richt, Juergen A, and Marazzi, Ivan
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Biodefense ,Infectious Diseases ,Vaccine Related ,Pneumonia ,Emerging Infectious Diseases ,Lung ,Pneumonia & Influenza ,Prevention ,Development of treatments and therapeutic interventions ,5.1 Pharmaceuticals ,Inflammatory and immune system ,Good Health and Well Being ,Animals ,COVID-19 ,Chlorocebus aethiops ,DNA Topoisomerases ,Type I ,Humans ,Inflammation ,Mesocricetus ,Mice ,Mice ,Transgenic ,SARS-CoV-2 ,THP-1 Cells ,Topoisomerase I Inhibitors ,Topotecan ,Vero Cells ,COVID-19 Drug Treatment ,chromatin ,cytokine storm ,epigenetics ,inducible genes ,inflammation ,topoisomerase ,topotecan ,transcription ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
The ongoing pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is currently affecting millions of lives worldwide. Large retrospective studies indicate that an elevated level of inflammatory cytokines and pro-inflammatory factors are associated with both increased disease severity and mortality. Here, using multidimensional epigenetic, transcriptional, in vitro, and in vivo analyses, we report that topoisomerase 1 (TOP1) inhibition suppresses lethal inflammation induced by SARS-CoV-2. Therapeutic treatment with two doses of topotecan (TPT), an FDA-approved TOP1 inhibitor, suppresses infection-induced inflammation in hamsters. TPT treatment as late as 4 days post-infection reduces morbidity and rescues mortality in a transgenic mouse model. These results support the potential of TOP1 inhibition as an effective host-directed therapy against severe SARS-CoV-2 infection. TPT and its derivatives are inexpensive clinical-grade inhibitors available in most countries. Clinical trials are needed to evaluate the efficacy of repurposing TOP1 inhibitors for severe coronavirus disease 2019 (COVID-19) in humans.
- Published
- 2021
8. Discovery of SARS-CoV-2 antiviral drugs through large-scale compound repurposing
- Author
-
Riva, Laura, Yuan, Shuofeng, Yin, Xin, Martin-Sancho, Laura, Matsunaga, Naoko, Pache, Lars, Burgstaller-Muehlbacher, Sebastian, De Jesus, Paul D, Teriete, Peter, Hull, Mitchell V, Chang, Max W, Chan, Jasper Fuk-Woo, Cao, Jianli, Poon, Vincent Kwok-Man, Herbert, Kristina M, Cheng, Kuoyuan, Nguyen, Tu-Trinh H, Rubanov, Andrey, Pu, Yuan, Nguyen, Courtney, Choi, Angela, Rathnasinghe, Raveen, Schotsaert, Michael, Miorin, Lisa, Dejosez, Marion, Zwaka, Thomas P, Sit, Ko-Yung, Martinez-Sobrido, Luis, Liu, Wen-Chun, White, Kris M, Chapman, Mackenzie E, Lendy, Emma K, Glynne, Richard J, Albrecht, Randy, Ruppin, Eytan, Mesecar, Andrew D, Johnson, Jeffrey R, Benner, Christopher, Sun, Ren, Schultz, Peter G, Su, Andrew I, García-Sastre, Adolfo, Chatterjee, Arnab K, Yuen, Kwok-Yung, and Chanda, Sumit K
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Emerging Infectious Diseases ,Rare Diseases ,Lung ,Orphan Drug ,Biodefense ,Infectious Diseases ,Vaccine Related ,Pneumonia & Influenza ,Pneumonia ,Prevention ,Development of treatments and therapeutic interventions ,5.1 Pharmaceuticals ,6.1 Pharmaceuticals ,Evaluation of treatments and therapeutic interventions ,Infection ,Good Health and Well Being ,Adenosine Monophosphate ,Alanine ,Alveolar Epithelial Cells ,Antiviral Agents ,Betacoronavirus ,COVID-19 ,Cell Line ,Coronavirus Infections ,Cysteine Proteinase Inhibitors ,Dose-Response Relationship ,Drug ,Drug Evaluation ,Preclinical ,Drug Repositioning ,Drug Synergism ,Gene Expression Regulation ,Humans ,Hydrazones ,Induced Pluripotent Stem Cells ,Models ,Biological ,Morpholines ,Pandemics ,Pneumonia ,Viral ,Pyrimidines ,Reproducibility of Results ,SARS-CoV-2 ,Small Molecule Libraries ,Triazines ,Virus Internalization ,Virus Replication ,COVID-19 Drug Treatment ,General Science & Technology - Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in 2019 has triggered an ongoing global pandemic of the severe pneumonia-like disease coronavirus disease 2019 (COVID-19)1. The development of a vaccine is likely to take at least 12-18 months, and the typical timeline for approval of a new antiviral therapeutic agent can exceed 10 years. Thus, repurposing of known drugs could substantially accelerate the deployment of new therapies for COVID-19. Here we profiled a library of drugs encompassing approximately 12,000 clinical-stage or Food and Drug Administration (FDA)-approved small molecules to identify candidate therapeutic drugs for COVID-19. We report the identification of 100 molecules that inhibit viral replication of SARS-CoV-2, including 21 drugs that exhibit dose-response relationships. Of these, thirteen were found to harbour effective concentrations commensurate with probable achievable therapeutic doses in patients, including the PIKfyve kinase inhibitor apilimod2-4 and the cysteine protease inhibitors MDL-28170, Z LVG CHN2, VBY-825 and ONO 5334. Notably, MDL-28170, ONO 5334 and apilimod were found to antagonize viral replication in human pneumocyte-like cells derived from induced pluripotent stem cells, and apilimod also demonstrated antiviral efficacy in a primary human lung explant model. Since most of the molecules identified in this study have already advanced into the clinic, their known pharmacological and human safety profiles will enable accelerated preclinical and clinical evaluation of these drugs for the treatment of COVID-19.
- Published
- 2020
9. Viral Determinants in H5N1 Influenza A Virus Enable Productive Infection of HeLa Cells.
- Author
-
Rodriguez-Frandsen, Ariel, Martin-Sancho, Laura, Gounder, Anshu, Chang, Max, Liu, Wen-Chun, De Jesus, Paul, von Recum-Knepper, Jessica, Dutra, Miriam, Huffmaster, Nicholas, Chavarria, Monica, Mena, Ignacio, Riva, Laura, Nguyen, Courtney, Dobariya, Saunil, Herbert, Kristina, Benner, Christopher, Albrecht, Randy, García-Sastre, Adolfo, and Chanda, Sumit
- Subjects
H5N1 ,HeLa ,heterokaryon ,highly pathogenic ,influenza A virus ,A549 Cells ,Animals ,Birds ,Cell Line ,Dogs ,HEK293 Cells ,HeLa Cells ,Humans ,Influenza A Virus ,H1N1 Subtype ,Influenza A Virus ,H3N2 Subtype ,Influenza A Virus ,H5N1 Subtype ,Influenza A virus ,Influenza in Birds ,Influenza ,Human ,Madin Darby Canine Kidney Cells ,Viral Tropism ,Virus Replication - Abstract
Influenza A virus (IAV) is a human respiratory pathogen that causes yearly global epidemics, as well as sporadic pandemics due to human adaptation of pathogenic strains. Efficient replication of IAV in different species is, in part, dictated by its ability to exploit the genetic environment of the host cell. To investigate IAV tropism in human cells, we evaluated the replication of IAV strains in a diverse subset of epithelial cell lines. HeLa cells were refractory to the growth of human H1N1 and H3N2 viruses and low-pathogenic avian influenza (LPAI) viruses. Interestingly, a human isolate of the highly pathogenic avian influenza (HPAI) H5N1 virus successfully propagated in HeLa cells to levels comparable to those in a human lung cell line. Heterokaryon cells generated by fusion of HeLa and permissive cells supported H1N1 virus growth, suggesting the absence of a host factor(s) required for the replication of H1N1, but not H5N1, viruses in HeLa cells. The absence of this factor(s) was mapped to reduced nuclear import, replication, and translation, as well as deficient viral budding. Using reassortant H1N1:H5N1 viruses, we found that the combined introduction of nucleoprotein (NP) and hemagglutinin (HA) from an H5N1 virus was necessary and sufficient to enable H1N1 virus growth. Overall, this study suggests that the absence of one or more cellular factors in HeLa cells results in abortive replication of H1N1, H3N2, and LPAI viruses, which can be circumvented upon the introduction of H5N1 virus NP and HA. Further understanding of the molecular basis of this restriction will provide important insights into the virus-host interactions that underlie IAV pathogenesis and tropism.IMPORTANCE Many zoonotic avian influenza A viruses have successfully crossed the species barrier and caused mild to life-threatening disease in humans. While human-to-human transmission is limited, there is a risk that these zoonotic viruses may acquire adaptive mutations enabling them to propagate efficiently and cause devastating human pandemics. Therefore, it is important to identify viral determinants that provide these viruses with a replicative advantage in human cells. Here, we tested the growth of influenza A virus in a subset of human cell lines and found that abortive replication of H1N1 viruses in HeLa cells can be circumvented upon the introduction of H5N1 virus HA and NP. Overall, this work leverages the genetic diversity of multiple human cell lines to highlight viral determinants that could contribute to H5N1 virus pathogenesis and tropism.
- Published
- 2020
10. Immunogenicity of chimeric haemagglutinin-based, universal influenza virus vaccine candidates: interim results of a randomised, placebo-controlled, phase 1 clinical trial
- Author
-
Bernstein, David I, Guptill, Jeffrey, Naficy, Abdollah, Nachbagauer, Raffael, Berlanda-Scorza, Francesco, Feser, Jodi, Wilson, Patrick C, Solórzano, Alicia, Van der Wielen, Marie, Walter, Emmanuel B, Albrecht, Randy A, Buschle, Kristen N, Chen, Yao-qing, Claeys, Carine, Dickey, Michelle, Dugan, Haley L, Ermler, Megan E, Freeman, Debra, Gao, Min, Gast, Christopher, Guthmiller, Jenna J, Hai, Rong, Henry, Carole, Lan, Linda Yu-Ling, McNeal, Monica, Palm, Anna-Karin E, Shaw, Dustin G, Stamper, Christopher T, Sun, Weina, Sutton, Victoria, Tepora, Micah E, Wahid, Rahnuma, Wenzel, Heather, Wohlbold, Teddy John, Innis, Bruce L, García-Sastre, Adolfo, Palese, Peter, and Krammer, Florian
- Subjects
Clinical Trials and Supportive Activities ,Influenza ,Immunization ,Prevention ,Biotechnology ,Vaccine Related ,Biodefense ,Pneumonia & Influenza ,Infectious Diseases ,Emerging Infectious Diseases ,Clinical Research ,Prevention of disease and conditions ,and promotion of well-being ,6.1 Pharmaceuticals ,Evaluation of treatments and therapeutic interventions ,3.4 Vaccines ,Infection ,Good Health and Well Being ,Adjuvants ,Immunologic ,Adult ,Female ,Healthy Volunteers ,Hemagglutinins ,Humans ,Influenza Vaccines ,Influenza ,Human ,Male ,Vaccination ,Vaccines ,Attenuated ,Vaccines ,Inactivated ,Clinical Sciences ,Medical Microbiology ,Public Health and Health Services ,Microbiology - Abstract
BackgroundInfluenza viruses cause substantial annual morbidity and mortality globally. Current vaccines protect against influenza only when well matched to the circulating strains. However, antigenic drift can cause considerable mismatches between vaccine and circulating strains, substantially reducing vaccine effectiveness. Moreover, current seasonal vaccines are ineffective against pandemic influenza, and production of a vaccine matched to a newly emerging virus strain takes months. Therefore, there is an unmet medical need for a broadly protective influenza virus vaccine. We aimed to test the ability of chimeric H1 haemagglutinin-based universal influenza virus vaccine candidates to induce broadly cross-reactive antibodies targeting the stalk domain of group 1 haemagglutinin-expressing influenza viruses.MethodsWe did a randomised, observer-blinded, phase 1 study in healthy adults in two centres in the USA. Participants were randomly assigned to one of three prime-boost, chimeric haemagglutinin-based vaccine regimens or one of two placebo groups. The vaccine regimens included a chimeric H8/1, intranasal, live-attenuated vaccine on day 1 followed by a non-adjuvanted, chimeric H5/1, intramuscular, inactivated vaccine on day 85; the same regimen but with the inactivated vaccine being adjuvanted with AS03; and an AS03-adjuvanted, chimeric H8/1, intramuscular, inactivated vaccine followed by an AS03-adjuvanted, chimeric H5/1, intramuscular, inactivated vaccine. In this planned interim analysis, the primary endpoints of reactogenicity and safety were assessed by blinded study group. We also assessed anti-H1 haemagglutinin stalk, anti-H2, anti-H9, and anti-H18 IgG antibody titres and plasmablast and memory B-cell responses in peripheral blood. This trial is registered with ClinicalTrials.gov, number NCT03300050.FindingsBetween Oct 10, 2017, and Nov 27, 2017, 65 participants were enrolled and randomly assigned. The adjuvanted inactivated vaccine, but not the live-attenuated vaccine, induced a substantial serum IgG antibody response after the prime immunisation, with a seven times increase in anti-H1 stalk antibody titres on day 29. After boost immunisation, all vaccine regimens induced detectable anti-H1 stalk antibody (2·2-5·6 times induction over baseline), cross-reactive serum IgG antibody, and peripheral blood plasmablast responses. An unsolicited adverse event was reported for 29 (48%) of 61 participants. Solicited local adverse events were reported in 12 (48%) of 25 participants following prime vaccination with intramuscular study product or placebo, in 12 (33%) of 36 after prime immunisation with intranasal study product or placebo, and in 18 (32%) of 56 following booster doses of study product or placebo. Solicited systemic adverse events were reported in 14 (56%) of 25 after prime immunisation with intramuscular study product or placebo, in 22 (61%) of 36 after immunisation with intranasal study product or placebo, and in 21 (38%) of 56 after booster doses of study product or placebo. Disaggregated safety data were not available at the time of this interim analysis.InterpretationThe tested chimeric haemagglutinin-based, universal influenza virus vaccine regimens elicited cross-reactive serum IgG antibodies that targeted the conserved haemagglutinin stalk domain. This is the first proof-of-principle study to show that high anti-stalk titres can be induced by a rationally designed vaccine in humans and opens up avenues for further development of universal influenza virus vaccines. On the basis of the blinded study group, the vaccine regimens were tolerable and no safety concerns were observed.FundingBill & Melinda Gates Foundation.
- Published
- 2020
11. NS2 is a key determinant of compatibility in reassortant avian influenza virus with heterologous H7N9-derived NS segment
- Author
-
Liu, Yee-Chen, Liao, Guan-Ru, Tsai, April Y., Tseng, Ching-Yu, Kuan, Chih-Ying, Tsai, Ruei-Sheng, Albrecht, Randy A., Kuo, Rei-Lin, Cheng, Ivan-Chen, Liang, Wan-Ting, Ou, Shan-Chia, and Hsu, Wei-Li
- Published
- 2023
- Full Text
- View/download PDF
12. Host-Specific NS5 Ubiquitination Determines Yellow Fever Virus Tropism.
- Author
-
Miorin, Lisa, Laurent-Rolle, Maudry, Pisanelli, Giuseppe, Co, Pierre Hendrick, Albrecht, Randy A, García-Sastre, Adolfo, and Morrison, Juliet
- Subjects
Infectious Diseases ,Vaccine Related ,Prevention ,Biodefense ,Biotechnology ,Rare Diseases ,Emerging Infectious Diseases ,Vector-Borne Diseases ,Immunization ,Aetiology ,2.1 Biological and endogenous factors ,Infection ,Good Health and Well Being ,Animals ,HEK293 Cells ,Humans ,Interferon-alpha ,Interferon-beta ,Mice ,Mice ,Knockout ,STAT2 Transcription Factor ,Ubiquitination ,Viral Nonstructural Proteins ,Viral Tropism ,Yellow fever virus ,Zika Virus ,NS5 ,STAT2 ,flavivirus ,host tropism ,interferon ,interferon antagonism ,yellow fever virus ,Biological Sciences ,Agricultural and Veterinary Sciences ,Medical and Health Sciences ,Virology - Abstract
The recent yellow fever virus (YFV) epidemic in Brazil in 2017 and Zika virus (ZIKV) epidemic in 2015 serve to remind us of the importance of flaviviruses as emerging human pathogens. With the current global flavivirus threat, there is an urgent need for antivirals and vaccines to curb the spread of these viruses. However, the lack of suitable animal models limits the research questions that can be answered. A common trait of all flaviviruses studied thus far is their ability to antagonize interferon (IFN) signaling so as to enhance viral replication and dissemination. Previously, we reported that YFV NS5 requires the presence of type I IFN (IFN-α/β) for its engagement with human signal transducer and activator of transcription 2 (hSTAT2). In this manuscript, we report that like the NS5 proteins of ZIKV and dengue virus (DENV), YFV NS5 protein is able to bind hSTAT2 but not murine STAT2 (mSTAT2). Contrary to what has been demonstrated with ZIKV NS5 and DENV NS5, replacing mSTAT2 with hSTAT2 cannot rescue the YFV NS5-STAT2 interaction, as YFV NS5 is also unable to interact with hSTAT2 in murine cells. We show that the IFN-α/β-dependent ubiquitination of YFV NS5 that is required for STAT2 binding in human cells is absent in murine cells. In addition, we demonstrate that mSTAT2 restricts YFV replication in vivo These data serve as further impetus for the development of an immunocompetent mouse model that can serve as a disease model for multiple flaviviruses.IMPORTANCE Flaviviruses such as yellow fever virus (YFV), Zika virus (ZIKV), and dengue virus (DENV) are important human pathogens. A common flavivirus trait is the antagonism of interferon (IFN) signaling to enhance viral replication and spread. We report that like ZIKV NS5 and DENV NS5, YFV NS5 binds human STAT2 (hSTAT2) but not mouse STAT2 (mSTAT2), a type I IFN (IFN-α/β) pathway component. Additionally, we show that contrary to what has been demonstrated with ZIKV NS5 and DENV NS5, YFV NS5 is unable to interact with hSTAT2 in murine cells. We demonstrate that mSTAT2 restricts YFV replication in mice and that this correlates with a lack of IFN-α/β-induced YFV NS5 ubiquitination in murine cells. The lack of suitable animal models limits flavivirus pathogenesis, vaccine, and drug research. These data serve as further impetus for the development of an immunocompetent mouse model that can serve as a disease model for multiple flaviviruses.
- Published
- 2019
13. Influenza virus infection causes global RNAPII termination defects
- Author
-
Zhao, Nan, Sebastiano, Vittorio, Moshkina, Natasha, Mena, Nacho, Hultquist, Judd, Jimenez-Morales, David, Ma, Yixuan, Rialdi, Alex, Albrecht, Randy, Fenouil, Romain, Sánchez-Aparicio, Maria Teresa, Ayllon, Juan, Ravisankar, Sweta, Haddad, Bahareh, Ho, Jessica Sook Yuin, Low, Diana, Jin, Jian, Yurchenko, Vyacheslav, Prinjha, Rab K, Tarakhovsky, Alexander, Squatrito, Massimo, Pinto, Dalila, Allette, Kimaada, Byun, Minji, Smith, Melissa Laird, Sebra, Robert, Guccione, Ernesto, Tumpey, Terrence, Krogan, Nevan, Greenbaum, Benjamin, van Bakel, Harm, García-Sastre, Adolfo, and Marazzi, Ivan
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Microbiology ,Clinical Sciences ,Medical Microbiology ,Infectious Diseases ,Emerging Infectious Diseases ,Biodefense ,Influenza ,Pneumonia & Influenza ,Prevention ,Genetics ,Vaccine Related ,2.1 Biological and endogenous factors ,2.2 Factors relating to the physical environment ,Aetiology ,Infection ,Humans ,Influenza A virus ,Influenza ,Human ,RNA Polymerase II ,Terminator Regions ,Genetic ,Virulence ,Chemical Sciences ,Medical and Health Sciences ,Biophysics ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences ,Chemical sciences - Abstract
Viral infection perturbs host cells and can be used to uncover regulatory mechanisms controlling cellular responses and susceptibility to infections. Using cell biological, biochemical, and genetic tools, we reveal that influenza A virus (IAV) infection induces global transcriptional defects at the 3' ends of active host genes and RNA polymerase II (RNAPII) run-through into extragenic regions. Deregulated RNAPII leads to expression of aberrant RNAs (3' extensions and host-gene fusions) that ultimately cause global transcriptional downregulation of physiological transcripts, an effect influencing antiviral response and virulence. This phenomenon occurs with multiple strains of IAV, is dependent on influenza NS1 protein, and can be modulated by SUMOylation of an intrinsically disordered region (IDR) of NS1 expressed by the 1918 pandemic IAV strain. Our data identify a strategy used by IAV to suppress host gene expression and indicate that polymorphisms in IDRs of viral proteins can affect the outcome of an infection.
- Published
- 2018
14. Transcription Elongation Can Affect Genome 3D Structure.
- Author
-
Heinz, Sven, Texari, Lorane, Hayes, Michael GB, Urbanowski, Matthew, Chang, Max W, Givarkes, Ninvita, Rialdi, Alexander, White, Kris M, Albrecht, Randy A, Pache, Lars, Marazzi, Ivan, García-Sastre, Adolfo, Shaw, Megan L, and Benner, Christopher
- Subjects
Cell Line ,Chromatin ,Macrophages ,Humans ,Piperidines ,Flavonoids ,RNA Polymerase II ,Carrier Proteins ,Cell Cycle Proteins ,Interferon-beta ,Proto-Oncogene Proteins ,Nuclear Proteins ,Chromosomal Proteins ,Non-Histone ,Viral Nonstructural Proteins ,RNA ,Small Interfering ,Transcription ,Genetic ,RNA Interference ,Binding Sites ,Protein Binding ,Genome ,Human ,Influenza A Virus ,H5N1 Subtype ,CCCTC-Binding Factor ,CTCF ,NS1 ,chromatin compaction ,cohesin ,genome 3D structure ,influenza A virus ,readthrough transcription ,transcription ,transcription elongation ,transcription termination ,Emerging Infectious Diseases ,Infectious Diseases ,Influenza ,Genetics ,Pneumonia & Influenza ,Human Genome ,1.1 Normal biological development and functioning ,Underpinning research ,Infection ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
How transcription affects genome 3D organization is not well understood. We found that during influenza A (IAV) infection, rampant transcription rapidly reorganizes host cell chromatin interactions. These changes occur at the ends of highly transcribed genes, where global inhibition of transcription termination by IAV NS1 protein causes readthrough transcription for hundreds of kilobases. In these readthrough regions, elongating RNA polymerase II disrupts chromatin interactions by inducing cohesin displacement from CTCF sites, leading to locus decompaction. Readthrough transcription into heterochromatin regions switches them from the inert (B) to the permissive (A) chromatin compartment and enables transcription factor binding. Data from non-viral transcription stimuli show that transcription similarly affects cohesin-mediated chromatin contacts within gene bodies. Conversely, inhibition of transcription elongation allows cohesin to accumulate at previously transcribed intragenic CTCF sites and to mediate chromatin looping and compaction. Our data indicate that transcription elongation by RNA polymerase II remodels genome 3D architecture.
- Published
- 2018
15. Detection of clade 2.3.4.4b highly pathogenic H5N1 influenza virus in New York City
- Author
-
Meade, Philip S., primary, Bandawane, Pooja, additional, Bushfield, Kaitlyn, additional, Hoxie, Irene, additional, Azcona, Karla R., additional, Burgos, Daneidy, additional, Choudhury, Sadia, additional, Diaby, Adama, additional, Diallo, Mariama, additional, Gaynor, Kailani, additional, Huang, Aaron, additional, Kante, Kadiatou, additional, Khan, Shehryar N., additional, Kim, William, additional, Ajayi, Paul Kehinde, additional, Roubidoux, Ericka, additional, Nelson, Sasha, additional, McMahon, Rita, additional, Albrecht, Randy A., additional, Krammer, Florian, additional, and Marizzi, Christine, additional
- Published
- 2024
- Full Text
- View/download PDF
16. Seven Opportunities for Effective Biosafety and Biosecurity Governance
- Author
-
Gillum, David R., primary, Schwartz, Antony, additional, Albrecht, Randy A., additional, and Moritz, Rebecca L., additional
- Published
- 2024
- Full Text
- View/download PDF
17. Longitudinal metabolomics of human plasma reveals prognostic markers of COVID-19 disease severity
- Author
-
Sindelar, Miriam, Stancliffe, Ethan, Schwaiger-Haber, Michaela, Anbukumar, Dhanalakshmi S., Adkins-Travis, Kayla, Goss, Charles W., O’Halloran, Jane A., Mudd, Philip A., Liu, Wen-Chun, Albrecht, Randy A., García-Sastre, Adolfo, Shriver, Leah P., and Patti, Gary J.
- Published
- 2021
- Full Text
- View/download PDF
18. Pathophysiology of SARS-CoV-2: the Mount Sinai COVID-19 autopsy experience
- Author
-
Bryce, Clare, Grimes, Zachary, Pujadas, Elisabet, Ahuja, Sadhna, Beasley, Mary Beth, Albrecht, Randy, Hernandez, Tahyna, Stock, Aryeh, Zhao, Zhen, AlRasheed, Mohamed Rizwan, Chen, Joyce, Li, Li, Wang, Diane, Corben, Adriana, Haines, G. Kenneth, III, Westra, William H., Umphlett, Melissa, Gordon, Ronald E., Reidy, Jason, Petersen, Bruce, Salem, Fadi, Fiel, Maria Isabel, El Jamal, Siraj M., Tsankova, Nadejda M., Houldsworth, Jane, Mussa, Zarmeen, Veremis, Brandon, Sordillo, Emilia, Gitman, Melissa R., Nowak, Michael, Brody, Rachel, Harpaz, Noam, Merad, Miriam, Gnjatic, Sacha, Liu, Wen-Chun, Schotsaert, Michael, Miorin, Lisa, Aydillo Gomez, Teresa A., Ramos-Lopez, Irene, Garcia-Sastre, Adolfo, Donnelly, Ryan, Seigler, Patricia, Keys, Calvin, Cameron, Jennifer, Moultrie, Isaiah, Washington, Kae-Lynn, Treatman, Jacquelyn, Sebra, Robert, Jhang, Jeffrey, Firpo, Adolfo, Lednicky, John, Paniz-Mondolfi, Alberto, Cordon-Cardo, Carlos, and Fowkes, Mary E.
- Published
- 2021
- Full Text
- View/download PDF
19. The RNA Exosome Syncs IAV-RNAPII Transcription to Promote Viral Ribogenesis and Infectivity.
- Author
-
McGregor, Michael, Haas, Kelsey, Pefanis, Evangelos, Albrecht, Randy, Pache, Lars, Chanda, Sumit, Jen, Joanna, Ochando, Jordi, Byun, Minji, Basu, Uttiya, García-Sastre, Adolfo, Krogan, Nevan, van Bakel, Harm, Marazzi, Ivan, Rialdi, Alexander, Hultquist, Judd, Jimenez-Morales, David, Peralta, Zuleyma, Campisi, Laura, Fenouil, Romain, Moshkina, Natasha, Wang, Zhen, Laffleur, Brice, and Kaake, Robyn
- Subjects
Influenza virus polymerase ,RNA chimeras ,RNA exosome ,RNA hybrids ,RNA surveillance ,RNAPII elongation ,epigenetics ,host-pathogen interactions ,neurodegeneration ,non-coding RNA ,A549 Cells ,Animals ,Chromatin Immunoprecipitation ,Exoribonucleases ,Exosome Multienzyme Ribonuclease Complex ,Exosomes ,Host-Pathogen Interactions ,Humans ,Influenza A Virus ,H1N1 Subtype ,Influenza A Virus ,H3N2 Subtype ,Influenza ,Human ,Mass Spectrometry ,Mice ,Mutation ,Neurodegenerative Diseases ,RNA Polymerase II ,RNA-Binding Proteins ,Ribosomes ,Transcription ,Genetic - Abstract
The nuclear RNA exosome is an essential multi-subunit complex that controls RNA homeostasis. Congenital mutations in RNA exosome genes are associated with neurodegenerative diseases. Little is known about the role of the RNA exosome in the cellular response to pathogens. Here, using NGS and human and mouse genetics, we show that influenza A virus (IAV) ribogenesis and growth are suppressed by impaired RNA exosome activity. Mechanistically, the nuclear RNA exosome coordinates the initial steps of viral transcription with RNAPII at host promoters. The viral polymerase complex co-opts the nuclear RNA exosome complex and cellular RNAs en route to 3 end degradation. Exosome deficiency uncouples chromatin targeting of the viral polymerase complex and the formation of cellular:viral RNA hybrids, which are essential RNA intermediates that license transcription of antisense genomic viral RNAs. Our results suggest that evolutionary arms races have shaped the cellular RNA quality control machinery.
- Published
- 2017
20. A human-airway-on-a-chip for the rapid identification of candidate antiviral therapeutics and prophylactics
- Author
-
Si, Longlong, Bai, Haiqing, Rodas, Melissa, Cao, Wuji, Oh, Crystal Yuri, Jiang, Amanda, Moller, Rasmus, Hoagland, Daisy, Oishi, Kohei, Horiuchi, Shu, Uhl, Skyler, Blanco-Melo, Daniel, Albrecht, Randy A., Liu, Wen-Chun, Jordan, Tristan, Nilsson-Payant, Benjamin E., Golynker, Ilona, Frere, Justin, Logue, James, Haupt, Robert, McGrath, Marisa, Weston, Stuart, Zhang, Tian, Plebani, Roberto, Soong, Mercy, Nurani, Atiq, Kim, Seong Min, Zhu, Danni Y., Benam, Kambez H., Goyal, Girija, Gilpin, Sarah E., Prantil-Baun, Rachelle, Gygi, Steven P., Powers, Rani K., Carlson, Kenneth E., Frieman, Matthew, tenOever, Benjamin R., and Ingber, Donald E.
- Published
- 2021
- Full Text
- View/download PDF
21. Hemagglutinin Stalk Immunity Reduces Influenza Virus Replication and Transmission in Ferrets
- Author
-
Nachbagauer, Raffael, Miller, Matthew S, Hai, Rong, Ryder, Alex B, Rose, John K, Palese, Peter, García-Sastre, Adolfo, Krammer, Florian, and Albrecht, Randy A
- Subjects
Biodefense ,Pneumonia & Influenza ,Immunization ,Prevention ,Emerging Infectious Diseases ,Infectious Diseases ,Influenza ,Vaccine Related ,2.2 Factors relating to the physical environment ,Aetiology ,Infection ,Good Health and Well Being ,Animals ,Disease Models ,Animal ,Disease Transmission ,Infectious ,Ferrets ,Hemagglutinin Glycoproteins ,Influenza Virus ,Influenza A Virus ,H1N1 Subtype ,Influenza Vaccines ,Male ,Orthomyxoviridae Infections ,Viral Load ,Virus Replication ,Biological Sciences ,Agricultural and Veterinary Sciences ,Medical and Health Sciences ,Virology - Abstract
We assessed whether influenza virus hemagglutinin stalk-based immunity protects ferrets against aerosol-transmitted H1N1 influenza virus infection. Immunization of ferrets by a universal influenza virus vaccine strategy based on viral vectors expressing chimeric hemagglutinin constructs induced stalk-specific antibody responses. Stalk-immunized ferrets were cohoused with H1N1-infected ferrets under conditions that permitted virus transmission. Hemagglutinin stalk-immunized ferrets had lower viral titers and delayed or no virus replication at all following natural exposure to influenza virus.
- Published
- 2016
22. Animal models for COVID-19
- Author
-
Muñoz-Fontela, César, Dowling, William E., Funnell, Simon G. P., Gsell, Pierre-S., Riveros-Balta, A. Ximena, Albrecht, Randy A., and Andersen, Hanne
- Subjects
Medical research ,Medicine, Experimental ,Animal models in research -- Usage -- Varieties ,Environmental issues ,Science and technology ,Zoology and wildlife conservation ,World Health Organization -- Research - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the aetiological agent of coronavirus disease 2019 (COVID-19), an emerging respiratory infection caused by the introduction of a novel coronavirus into humans late in 2019 (first detected in Hubei province, China). As of 18 September 2020, SARS-CoV-2 has spread to 215 countries, has infected more than 30 million people and has caused more than 950,000 deaths. As humans do not have pre-existing immunity to SARS-CoV-2, there is an urgent need to develop therapeutic agents and vaccines to mitigate the current pandemic and to prevent the re-emergence of COVID-19. In February 2020, the World Health Organization (WHO) assembled an international panel to develop animal models for COVID-19 to accelerate the testing of vaccines and therapeutic agents. Here we summarize the findings to date and provides relevant information for preclinical testing of vaccine candidates and therapeutic agents for COVID-19. The findings of a World Health Organization expert working group that is developing animal models to test vaccines and therapeutic agents for the treatment of COVID-19, and their relevance for preclinical testing, are reviewed., Author(s): César Muñoz-Fontela [sup.1] [sup.2] , William E. Dowling [sup.3] , Simon G. P. Funnell [sup.4] , Pierre-S. Gsell [sup.5] , A. Ximena Riveros-Balta [sup.5] , Randy A. Albrecht [sup.6] [...]
- Published
- 2020
- Full Text
- View/download PDF
23. Immunologic Characterization of a Rhesus Macaque H1N1 Challenge Model for Candidate Influenza Virus Vaccine Assessment
- Author
-
Skinner, Jason A, Zurawski, Sandra M, Sugimoto, Chie, Vinet-Oliphant, Heather, Vinod, Parvathi, Xue, Yaming, Russell-Lodrigue, Kasi, Albrecht, Randy A, García-Sastre, Adolfo, Salazar, Andres M, Roy, Chad J, Kuroda, Marcelo J, Oh, SangKon, and Zurawski, Gerard
- Subjects
Microbiology ,Biological Sciences ,Rare Diseases ,Vaccine Related ,Emerging Infectious Diseases ,Immunization ,Infectious Diseases ,Influenza ,Pneumonia & Influenza ,Biotechnology ,Prevention ,Biodefense ,Prevention of disease and conditions ,and promotion of well-being ,3.4 Vaccines ,Infection ,Good Health and Well Being ,Animals ,Antibodies ,Viral ,Humans ,Influenza A Virus ,H1N1 Subtype ,Influenza Vaccines ,Influenza ,Human ,Macaca mulatta ,Orthomyxoviridae Infections ,Vaccination ,Immunology - Abstract
Despite the availability of annually formulated vaccines, influenza virus infection remains a worldwide public health burden. Therefore, it is important to develop preclinical challenge models that enable the evaluation of vaccine candidates while elucidating mechanisms of protection. Here, we report that naive rhesus macaques challenged with 2009 pandemic H1N1 (pH1N1) influenza virus do not develop observable clinical symptoms of disease but develop a subclinical biphasic fever on days 1 and 5 to 6 postchallenge. Whole blood microarray analysis further revealed that interferon activity was associated with fever. We then tested whether type I interferon activity in the blood is a correlate of vaccine efficacy. The animals immunized with candidate vaccines carrying hemagglutinin (HA) or nucleoprotein (NP) exhibited significantly reduced interferon activity on days 5 to 6 postchallenge. Supported by cellular and serological data, we conclude that blood interferon activity is a prominent marker that provides a convenient metric of influenza virus vaccine efficacy in the subclinical rhesus macaque model.
- Published
- 2014
24. The immunological potency and therapeutic potential of a prototype dual vaccine against influenza and Alzheimer's disease
- Author
-
Davtyan, Hayk, Ghochikyan, Anahit, Cadagan, Richard, Zamarin, Dmitriy, Petrushina, Irina, Movsesyan, Nina, Martinez-Sobrido, Luis, Albrecht, Randy A, García-Sastre, Adolfo, and Agadjanyan, Michael G
- Abstract
Abstract Background Numerous pre-clinical studies and clinical trials demonstrated that induction of antibodies to the β-amyloid peptide of 42 residues (Aβ42) elicits therapeutic effects in Alzheimer's disease (AD). However, an active vaccination strategy based on full length Aβ42 is currently hampered by elicitation of T cell pathological autoreactivity. We attempt to improve vaccine efficacy by creating a novel chimeric flu vaccine expressing the small immunodominant B cell epitope of Aβ42. We hypothesized that in elderly people with pre-existing memory Th cells specific to influenza this dual vaccine will simultaneously boost anti-influenza immunity and induce production of therapeutically active anti-Aβ antibodies. Methods Plasmid-based reverse genetics system was used for the rescue of recombinant influenza virus containing immunodominant B cell epitopes of Aβ42 (Aβ1-7/10). Results Two chimeric flu viruses expressing either 7 or 10 aa of Aβ42 (flu-Aβ1-7 or flu-Aβ1-10) were generated and tested in mice as conventional inactivated vaccines. We demonstrated that this dual vaccine induced therapeutically potent anti-Aβ antibodies and anti-influenza antibodies in mice. Conclusion We suggest that this strategy might be beneficial for treatment of AD patients as well as for prevention of development of AD pathology in pre-symptomatic individuals while concurrently boosting immunity against influenza.
- Published
- 2011
25. Clinical and Serologic Responses After a Two-dose Series of High-dose Influenza Vaccine in Plasma Cell Disorders: A Prospective, Single-arm Trial
- Author
-
Branagan, Andrew R., Duffy, Eamon, Albrecht, Randy A., Cooper, Dennis L., Seropian, Stuart, Parker, Terri L., Gan, Geliang, Li, Fangyong, Zelterman, Daniel, Boddupalli, Chandra Sekhar, Zhang, Lin, Verma, Rakesh, Ferencz, Thomas M., and Dhodapkar, Madhav V.
- Published
- 2017
- Full Text
- View/download PDF
26. Author Correction: Microbiome disturbance and resilience dynamics of the upper respiratory tract during influenza A virus infection
- Author
-
Kaul, Drishti, Rathnasinghe, Raveen, Ferres, Marcela, Tan, Gene S., Barrera, Aldo, Pickett, Brett E., Methe, Barbara A., Das, Suman R., Budnik, Isolda, Halpin, Rebecca A., Wentworth, David, Schmolke, Mirco, Mena, Ignacio, Albrecht, Randy A., Singh, Indresh, Nelson, Karen E., García-Sastre, Adolfo, Dupont, Chris L., and Medina, Rafael A.
- Published
- 2020
- Full Text
- View/download PDF
27. Microbiome disturbance and resilience dynamics of the upper respiratory tract during influenza A virus infection
- Author
-
Kaul, Drishti, Rathnasinghe, Raveen, Ferres, Marcela, Tan, Gene S., Barrera, Aldo, Pickett, Brett E., Methe, Barbara A., Das, Suman R., Budnik, Isolda, Halpin, Rebecca A., Wentworth, David, Schmolke, Mirco, Mena, Ignacio, Albrecht, Randy A., Singh, Indresh, Nelson, Karen E., García-Sastre, Adolfo, Dupont, Chris L., and Medina, Rafael A.
- Published
- 2020
- Full Text
- View/download PDF
28. Pandemic influenza virus vaccines boost hemagglutinin stalk-specific antibody responses in primed adult and pediatric cohorts
- Author
-
Nachbagauer, Raffael, Salaun, Bruno, Stadlbauer, Daniel, Behzadi, Mohammad A., Friel, Damien, Rajabhathor, Arvind, Choi, Angela, Albrecht, Randy A., Debois, Muriel, García-Sastre, Adolfo, Rouxel, Ronan N., Sun, Weina, Palese, Peter, Mallett, Corey P., Innis, Bruce L., Krammer, Florian, and Claeys, Carine
- Published
- 2019
- Full Text
- View/download PDF
29. Phylogenetic landscape of Monkeypox Virus (MPV) during the early outbreak in New York City, 2022.
- Author
-
Patiño, Luz H., Guerra, Susana, Muñoz, Marina, Luna, Nicolas, Farrugia, Keith, van de Guchte, Adriana, Khalil, Zain, Gonzalez-Reiche, Ana Silvia, Hernandez, Matthew M., Banu, Radhika, Shrestha, Paras, Liggayu, Bernadette, Firpo Betancourt, Adolfo, Reich, David, Cordon-Cardo, Carlos, Albrecht, Randy, Pearl, Rebecca, Simon, Viviana, Rooker, Aria, and Sordillo, Emilia Mia
- Published
- 2023
- Full Text
- View/download PDF
30. Mutation L319Q in the PB1 Polymerase Subunit Improves Attenuation of a Candidate Live-Attenuated Influenza A Virus Vaccine
- Author
-
New York Influenza Center of Excellence, National Institute of Allergy and Infectious Diseases (US), Department of Health and Human Services (US), Centers of Excellence for Influenza Research and Surveillance (US), Department of Defense (US), Center for Research for Influenza Pathogenesis (US), Ministerio de Ciencia e Innovación (España), Icahn School of Medicine at Mount Sinai, Nogales, Aitor [0000-0002-2424-7900], Lowen, Anice C. [0000-0002-9829-112X], Chiem, Kevin [0000-0002-3892-5944], García-Sastre, Adolfo [0000-0002-6551-1827], Albrecht, Randy A. [0000-0003-4008-503X], Dewhurst, Stephen [0000-0001-7729-7920], Nogales, Aitor, Steel, John, Liu, Wen-Chun, Lowen, Anice C., Rodriguez, Laura, Chiem, Kevin, Cox, Andrew, García-Sastre, Adolfo, Albrecht, Randy A., Dewhurst, Stephen, Martínez-Sobrido, Luis, New York Influenza Center of Excellence, National Institute of Allergy and Infectious Diseases (US), Department of Health and Human Services (US), Centers of Excellence for Influenza Research and Surveillance (US), Department of Defense (US), Center for Research for Influenza Pathogenesis (US), Ministerio de Ciencia e Innovación (España), Icahn School of Medicine at Mount Sinai, Nogales, Aitor [0000-0002-2424-7900], Lowen, Anice C. [0000-0002-9829-112X], Chiem, Kevin [0000-0002-3892-5944], García-Sastre, Adolfo [0000-0002-6551-1827], Albrecht, Randy A. [0000-0003-4008-503X], Dewhurst, Stephen [0000-0001-7729-7920], Nogales, Aitor, Steel, John, Liu, Wen-Chun, Lowen, Anice C., Rodriguez, Laura, Chiem, Kevin, Cox, Andrew, García-Sastre, Adolfo, Albrecht, Randy A., Dewhurst, Stephen, and Martínez-Sobrido, Luis
- Abstract
Influenza A viruses (IAV) remain emerging threats to human public health. Live-attenuated influenza vaccines (LAIV) are one of the most effective prophylactic options to prevent disease caused by influenza infections. However, licensed LAIV remain restricted for use in 2- to 49-year-old healthy and nonpregnant people. Therefore, development of LAIV with increased safety, immunogenicity, and protective efficacy is highly desired. The U.S.-licensed LAIV is based on the master donor virus (MDV) A/Ann Arbor/6/60 H2N2 backbone, which was generated by adaptation of the virus to growth at low temperatures. Introducing the genetic signature of the U.S. MDV into the backbone of other IAV strains resulted in varying levels of attenuation. While the U.S. MDV mutations conferred an attenuated phenotype to other IAV strains, the same amino acid changes did not significantly attenuate the pandemic A/California/04/09 H1N1 (pH1N1) strain. To attenuate pH1N1, we replaced the conserved leucine at position 319 with glutamine (L319Q) in PB1 and analyzed the in vitro and in vivo properties of pH1N1 viruses containing either PB1 L319Q alone or in combination with the U.S. MDV mutations using two animal models of influenza infection and transmission, ferrets and guinea pigs. Our results demonstrated that L319Q substitution in the pH1N1 PB1 alone or in combination with the mutations of the U.S. MDV resulted in reduced pathogenicity (ferrets) and transmission (guinea pigs), and an enhanced temperature sensitive phenotype. These results demonstrate the feasibility of generating an attenuated MDV based on the backbone of a contemporary pH1N1 IAV strain. IMPORTANCE Vaccination represents the most effective strategy to reduce the impact of seasonal IAV infections. Although LAIV are superior in inducing protection and sterilizing immunity, they are not recommended for many individuals who are at high risk for severe disease. Thus, development of safer and more effective LAIV are needed. A concer
- Published
- 2022
31. Field Research Is Essential to Counter Virological Threats
- Author
-
Runstadler, Jonathan A., primary, Lowen, Anice C., additional, Kayali, Ghazi, additional, Tompkins, S. Mark, additional, Albrecht, Randy A., additional, Fouchier, Ron A. M., additional, Stallknecht, David E., additional, Lakdawala, Seema S., additional, Goodrum, Felicia D., additional, Casadevall, Arturo, additional, Enquist, Lynn W., additional, Alwine, James C., additional, Imperiale, Michael J., additional, Schultz-Cherry, Stacey, additional, and Webby, Richard J., additional
- Published
- 2023
- Full Text
- View/download PDF
32. Proteomic and genetic analyses of influenza A viruses identify pan-viral host targets.
- Author
-
Haas, Kelsey, Haas, Kelsey, McGregor, Michael, Polacco, Benjamin, Kim, Eun-Young, Nguyen, Thong, Newton, Billy, Urbanowski, Matthew, Kim, Heejin, Williams, Michael, Rezelj, Veronica, Hardy, Alexandra, Stevenson, Erica, Sukerman, Ellie, Kim, Tiffany, Penugonda, Sudhir, Moreno, Elena, Braberg, Hannes, Zhou, Yuan, Metreveli, Giorgi, Harjai, Bhavya, Tummino, Tia, Melnyk, James, Shoichet, Brian, Shriver, Leah, Johnson, Jeffrey, Shaw, Megan, Chanda, Sumit, Roden, Dan, Carter, Tonia, Kottyan, Leah, Chisholm, Rex, Pacheco, Jennifer, Smith, Maureen, Schrodi, Steven, Albrecht, Randy, Vignuzzi, Marco, Zuliani-Alvarez, Lorena, Swaney, Danielle, Eckhardt, Manon, Wolinsky, Steven, White, Kris, Hultquist, Judd, Batra, Jyoti, Pache, Lars, Martin-Sancho, Laura, Carlson-Stevermer, Jared, Jureka, Alexander, Basler, Christopher, García-Sastre, Adolfo, Krogan, Nevan, Shokat, Kevan, Kaake, Robyn, Soucheray, Margaret, Bouhaddou, Mehdi, Fossati, Andrea, Haas, Kelsey, Haas, Kelsey, McGregor, Michael, Polacco, Benjamin, Kim, Eun-Young, Nguyen, Thong, Newton, Billy, Urbanowski, Matthew, Kim, Heejin, Williams, Michael, Rezelj, Veronica, Hardy, Alexandra, Stevenson, Erica, Sukerman, Ellie, Kim, Tiffany, Penugonda, Sudhir, Moreno, Elena, Braberg, Hannes, Zhou, Yuan, Metreveli, Giorgi, Harjai, Bhavya, Tummino, Tia, Melnyk, James, Shoichet, Brian, Shriver, Leah, Johnson, Jeffrey, Shaw, Megan, Chanda, Sumit, Roden, Dan, Carter, Tonia, Kottyan, Leah, Chisholm, Rex, Pacheco, Jennifer, Smith, Maureen, Schrodi, Steven, Albrecht, Randy, Vignuzzi, Marco, Zuliani-Alvarez, Lorena, Swaney, Danielle, Eckhardt, Manon, Wolinsky, Steven, White, Kris, Hultquist, Judd, Batra, Jyoti, Pache, Lars, Martin-Sancho, Laura, Carlson-Stevermer, Jared, Jureka, Alexander, Basler, Christopher, García-Sastre, Adolfo, Krogan, Nevan, Shokat, Kevan, Kaake, Robyn, Soucheray, Margaret, Bouhaddou, Mehdi, and Fossati, Andrea
- Abstract
Influenza A Virus (IAV) is a recurring respiratory virus with limited availability of antiviral therapies. Understanding host proteins essential for IAV infection can identify targets for alternative host-directed therapies (HDTs). Using affinity purification-mass spectrometry and global phosphoproteomic and protein abundance analyses using three IAV strains (pH1N1, H3N2, H5N1) in three human cell types (A549, NHBE, THP-1), we map 332 IAV-human protein-protein interactions and identify 13 IAV-modulated kinases. Whole exome sequencing of patients who experienced severe influenza reveals several genes, including scaffold protein AHNAK, with predicted loss-of-function variants that are also identified in our proteomic analyses. Of our identified host factors, 54 significantly alter IAV infection upon siRNA knockdown, and two factors, AHNAK and coatomer subunit COPB1, are also essential for productive infection by SARS-CoV-2. Finally, 16 compounds targeting our identified host factors suppress IAV replication, with two targeting CDK2 and FLT3 showing pan-antiviral activity across influenza and coronavirus families. This study provides a comprehensive network model of IAV infection in human cells, identifying functional host targets for pan-viral HDT.
- Published
- 2023
33. Field Research Is Essential to Counter Virological Threats
- Author
-
Runstadler, Jonathan A., Lowen, Anice C., Kayali, Ghazi, Tompkins, S. Mark, Albrecht, Randy A., Fouchier, Ron A.M., Stallknecht, David E., Lakdawala, Seema S., Goodrum, Felicia D., Casadevall, Arturo, Enquist, Lynn W., Alwine, James C., Imperiale, Michael J., Schultz-Cherry, Stacey, Webby, Richard J., Runstadler, Jonathan A., Lowen, Anice C., Kayali, Ghazi, Tompkins, S. Mark, Albrecht, Randy A., Fouchier, Ron A.M., Stallknecht, David E., Lakdawala, Seema S., Goodrum, Felicia D., Casadevall, Arturo, Enquist, Lynn W., Alwine, James C., Imperiale, Michael J., Schultz-Cherry, Stacey, and Webby, Richard J.
- Abstract
The interface between humans and wildlife is changing and, with it, the potential for pathogen introduction into humans has increased. Avian influenza is a prominent example, with an ongoing outbreak showing the unprecedented expansion of both geographic and host ranges. Research in the field is essential to understand this and other zoonotic threats. Only by monitoring dynamic viral populations and defining their biology in situ can we gather the information needed to ensure effective pandemic preparation.
- Published
- 2023
34. Active opioid use does not attenuate the humoral responses to inactivated influenza vaccine
- Author
-
Moroz, Ekaterina, Albrecht, Randy A., Aden, Brandon, Beeder, Ann Bordwine, Yuan, Jianda, García-Sastre, Adolfo, Edlin, Brian R., and Salvatore, Mirella
- Published
- 2016
- Full Text
- View/download PDF
35. Analyses of Cellular Immune Responses in Ferrets Following Influenza Virus Infection
- Author
-
DiPiazza, Anthony T., primary, Richards, Katherine A., additional, Liu, Wen-Chun, additional, Albrecht, Randy A., additional, and Sant, Andrea J., additional
- Published
- 2018
- Full Text
- View/download PDF
36. Assessment of Influenza Virus Hemagglutinin Stalk-Specific Antibody Responses
- Author
-
Liu, Wen-Chun, primary, Nachbagauer, Raffael, additional, Krammer, Florian, additional, and Albrecht, Randy A., additional
- Published
- 2018
- Full Text
- View/download PDF
37. Defining the antibody cross-reactome directed against the influenza virus surface glycoproteins
- Author
-
Nachbagauer, Raffael, Choi, Angela, Hirsh, Ariana, Margine, Irina, Iida, Sayaka, Barrera, Aldo, Ferres, Marcela, Albrecht, Randy A, García-Sastre, Adolfo, Bouvier, Nicole M, Ito, Kimihito, Medina, Rafael A, Palese, Peter, and Krammer, Florian
- Published
- 2017
- Full Text
- View/download PDF
38. Life-threatening influenza and impaired interferon amplification in human IRF7 deficiency
- Author
-
Ciancanelli, Michael J., Huang, Sarah X. L., Luthra, Priya, Garner, Hannah, Itan, Yuval, Volpi, Stefano, Lafaille, Fabien G., Trouillet, Céline, Schmolke, Mirco, Albrecht, Randy A., Israelsson, Elisabeth, Lim, Hye Kyung, Casadio, Melina, Hermesh, Tamar, Lorenzo, Lazaro, Leung, Lawrence W., Pedergnana, Vincent, Boisson, Bertrand, Okada, Satoshi, Picard, Capucine, Ringuier, Benedicte, Troussier, Françoise, Chaussabel, Damien, Abel, Laurent, Pellier, Isabelle, Notarangelo, Luigi D., García-Sastre, Adolfo, Basler, Christopher F., Geissmann, Frédéric, Zhang, Shen-Ying, Snoeck, Hans-Willem, and Casanova, Jean-Laurent
- Published
- 2015
39. Distinct Patterns of B-Cell Activation and Priming by Natural Influenza Virus Infection Versus Inactivated Influenza Vaccination
- Author
-
He, Xiao-Song, Holmes, Tyson H., Sanyal, Mrinmoy, Albrecht, Randy A., García-Sastre, Adolfo, Dekker, Cornelia L., Davis, Mark M., and Greenberg, Harry B.
- Published
- 2015
40. A dual vaccine against influenza & Alzheimer's disease failed to enhance anti-β-amyloid antibody responses in mice with pre-existing virus specific memory
- Author
-
Davtyan, Hayk, Ghochikyan, Anahit, Hovakimyan, Armine, Davtyan, Arpine, Cadagan, Richard, Marleau, Annette M., Albrecht, Randy A., García-Sastre, Adolfo, and Agadjanyan, Michael G.
- Published
- 2014
- Full Text
- View/download PDF
41. Evaluation and validation of an RT‐PCR assay for specific detection of monkeypox virus (MPXV)
- Author
-
Paniz‐Mondolfi, Alberto, primary, Guerra, Susana, additional, Muñoz, Marina, additional, Luna, Nicolas, additional, Hernandez, Matthew M., additional, Patino, Luz H., additional, Reidy, Jason, additional, Banu, Radhika, additional, Shrestha, Paras, additional, Liggayu, Bernadette, additional, Umeaku, Audrey, additional, Chen, Feng, additional, Cao, Liyong, additional, Patel, Armi, additional, Hanna, Ayman, additional, Li, Sunny, additional, Look, Andy, additional, Pagani, Nina, additional, Albrecht, Randy, additional, Pearl, Rebecca, additional, Garcia‐Sastre, Adolfo, additional, Bogunovic, Dusan, additional, Palacios, Gustavo, additional, Bonnier, Lucia, additional, Cera, Freddy, additional, Lopez, Heidi, additional, Calderon, Yvette, additional, Eiting, Erick, additional, Mullen, Karr, additional, Shin, Sangyoon Jason, additional, Lugo, Luz Amarilis, additional, Urbina, Antonio E., additional, Starks, Carlotta, additional, Koo, Tonny, additional, Uychiat, Patricia, additional, Look, Avery, additional, van Bakel, Harm, additional, Gonzalez‐Reiche, Ana, additional, Betancourt, Adolfo Firpo, additional, Reich, David, additional, Cordon‐Cardo, Carlos, additional, Simon, Viviana, additional, Sordillo, Emilia M., additional, and Ramírez, Juan David, additional
- Published
- 2022
- Full Text
- View/download PDF
42. A variant NS1 protein from H5N2 avian influenza virus suppresses PKR activation and promotes replication and virulence in mammals
- Author
-
Chung, Yun-Ting, primary, Kuan, Chih-Ying, additional, Liao, Guan-Ru, additional, Albrecht, Randy A., additional, Tseng, Yeu-Yang, additional, Hsu, Yu-Chen, additional, Ou, Shan-Chia, additional, and Hsu, Wei-Li, additional
- Published
- 2022
- Full Text
- View/download PDF
43. The origin of the PB1 segment of swine influenza A virus subtype H1N2 determines viral pathogenicity in mice
- Author
-
Metreveli, Giorgi, Gao, Qinshan, Mena, Ignacio, Schmolke, Mirco, Berg, Mikael, Albrecht, Randy A., and García-Sastre, Adolfo
- Published
- 2014
- Full Text
- View/download PDF
44. One-shot vaccination with an insect cell-derived low-dose influenza A H7 virus-like particle preparation protects mice against H7N9 challenge
- Author
-
Klausberger, Miriam, Wilde, Monika, Palmberger, Dieter, Hai, Rong, Albrecht, Randy A., Margine, Irina, Hirsh, Ariana, García-Sastre, Adolfo, Grabherr, Reingard, and Krammer, Florian
- Published
- 2014
- Full Text
- View/download PDF
45. Distinct Cross-reactive B-Cell Responses to Live Attenuated and Inactivated Influenza Vaccines
- Author
-
Sasaki, Sanae, Holmes, Tyson H., Albrecht, Randy A., García-Sastre, Adolfo, Dekker, Cornelia L., He, Xiao-Song, and Greenberg, Harry B.
- Published
- 2014
46. Differences in Antibody Responses Between Trivalent Inactivated Influenza Vaccine and Live Attenuated Influenza Vaccine Correlate With the Kinetics and Magnitude of Interferon Signaling in Children
- Author
-
Cao, Raquel G., Suarez, Nicolas M., Obermoser, Gerlinde, Lopez, Santiago M. C., Flano, Emilio, Mertz, Sara E., Albrecht, Randy A., Garcia-Sastre, Adolfo, Mejias, Asuncion, Xu, Hui, Qin, Huanying, Blankenship, Derek, Palucka, Karolina, Pascual, Virginia, and Ramilo, Octavio
- Published
- 2014
47. H7N9 influenza virus neutralizing antibodies that possess few somatic mutations
- Author
-
Thornburg, Natalie J., Zhang, Heng, Bangaru, Sandhya, Sapparapu, Gopal, Kose, Nurgun, Lampley, Rebecca M., Bombardi, Robin G., Yu, Yingchun, Graham, Stephen, Branchizio, Andre, Yoder, Sandra M., Rock, Michael T., Creech, C. Buddy, Edwards, Kathryn M., Lee, David, Li, Sheng, Wilson, Ian A., Garcia-Sastre, Adolfo, Albrecht, Randy A., and Crowe, Jr., James E.
- Subjects
Gene mutations -- Identification and classification -- Health aspects ,Influenza -- Genetic aspects -- Care and treatment ,Monoclonal antibodies -- Testing ,Health care industry - Abstract
Avian H7N9 influenza viruses are group 2 influenza A viruses that have been identified as the etiologic agent for a current major outbreak that began in China in 2013 and may pose a pandemic threat. Here, we examined the human H7-reactive antibody response in 75 recipients of a monovalent inactivated A/Shanghai/02/2013 H7N9 vaccine. After 2 doses of vaccine, the majority of donors had memory B cells that secreted IgGs specific for H7 HA, with dominant responses against single HA subtypes, although frequencies of H7-reactive B cells ranged widely between donors. We isolated 12 naturally occurring mAbs with low half-maximal effective concentrations for binding, 5 of which possessed neutralizing and HA-inhibiting activities. The 5 neutralizing mAbs exhibited narrow breadth of reactivity with influenza H7 strains. Epitope-mapping studies using neutralization escape mutant analysis, deuterium exchange mass spectrometry, and x-ray crystallography revealed that these neutralizing mAbs bind near the receptor-binding pocket on HA. All 5 neutralizing mAbs possessed low numbers of somatic mutations, suggesting the clones arose from naive B cells. The most potent mAb, H7.167, was tested as a prophylactic treatment in a mouse intranasal virus challenge study, and systemic administration of the mAb markedly reduced viral lung titers., Introduction Influenza type A viruses comprise a phylogenetically and antigenically diverse group of viruses that infect both human and animal populations. Influenza is a common cause of yearly epidemics and, [...]
- Published
- 2016
- Full Text
- View/download PDF
48. Mutation L319Q in the PB1 Polymerase Subunit Improves Attenuation of a Candidate Live-Attenuated Influenza A Virus Vaccine
- Author
-
Nogales, Aitor, primary, Steel, John, additional, Liu, Wen-Chun, additional, Lowen, Anice C., additional, Rodriguez, Laura, additional, Chiem, Kevin, additional, Cox, Andrew, additional, García-Sastre, Adolfo, additional, Albrecht, Randy A., additional, Dewhurst, Stephen, additional, and Martínez-Sobrido, Luis, additional
- Published
- 2022
- Full Text
- View/download PDF
49. A point mutation in the polymerase protein PB2 allows a reassortant H9N2 influenza isolate of wild-bird origin to replicate in human cells
- Author
-
Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Division of Comparative Medicine, Hussein, Islam, Ma, Eric Jinglong, Hill, Nichola, Meixell, Brandt W, Lindberg, Mark, Albrecht, Randy A, Bahl, Justin, Runstadler, Jonathan, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Division of Comparative Medicine, Hussein, Islam, Ma, Eric Jinglong, Hill, Nichola, Meixell, Brandt W, Lindberg, Mark, Albrecht, Randy A, Bahl, Justin, and Runstadler, Jonathan
- Abstract
© 2016. H9N2 influenza A viruses are on the list of potentially pandemic subtypes. Therefore, it is important to understand how genomic reassortment and genetic polymorphisms affect phenotypes of H9N2 viruses circulating in the wild bird reservoir. A comparative genetic analysis of North American H9N2 isolates of wild bird origin identified a naturally occurring reassortant virus containing gene segments derived from both North American and Eurasian lineage ancestors. The PB2 segment of this virus encodes 10 amino acid changes that distinguish it from other H9 strains circulating in North America. G590S, one of the 10 amino acid substitutions observed, was present in ~12% of H9 viruses worldwide. This mutation combined with R591 has been reported as a marker of pathogenicity for human pandemic 2009 H1N1 viruses. Screening by polymerase reporter assay of all the natural polymorphisms at these two positions identified G590/K591 and S590/K591 as the most active, with the highest polymerase activity recorded for the SK polymorphism. Rescued viruses containing these two polymorphic combinations replicated more efficiently in MDCK cells and they were the only ones tested that were capable of establishing productive infection in NHBE cells. A global analysis of all PB2 sequences identified the K591 signature in six viral HA/NA subtypes isolated from several hosts in seven geographic locations. Interestingly, introducing the K591 mutation into the PB2 of a human-adapted H3N2 virus did not affect its polymerase activity. Our findings demonstrate that a single point mutation in the PB2 of a low pathogenic H9N2 isolate could have a significant effect on viral phenotype and increase its propensity to infect mammals. However, this effect is not universal, warranting caution in interpreting point mutations without considering protein sequence context.
- Published
- 2022
50. Advances and gaps in SARS-CoV-2 infection models
- Author
-
Munoz-Fontela, César, Widerspick, Lina, Albrecht, Randy A., Beer, Martin, Carroll, Miles W., de Wit, Emmie, Diamond, Michael S., Dowling, William E., Funnell, Simon G.P., Garcia-Sastre, Adolfo, Gerhards, N.M., Klaassen-de Jong, M.C., Munster, Vincent J., Neyts, Johan, Perlman, Stanley, Reed, Douglas S., Richt, Juergen A., Riveros-Balta, Ximena, Roy, Chad J., Salguero, Francisco J., Schotsaert, Michael, Schwartz, Lauren M., Seder, Robert A., Segalés, Joaquim, Vasan, Seshadri S., Henao-Restrepo, Ana Mariá, Barouch, Dan H., Munoz-Fontela, César, Widerspick, Lina, Albrecht, Randy A., Beer, Martin, Carroll, Miles W., de Wit, Emmie, Diamond, Michael S., Dowling, William E., Funnell, Simon G.P., Garcia-Sastre, Adolfo, Gerhards, N.M., Klaassen-de Jong, M.C., Munster, Vincent J., Neyts, Johan, Perlman, Stanley, Reed, Douglas S., Richt, Juergen A., Riveros-Balta, Ximena, Roy, Chad J., Salguero, Francisco J., Schotsaert, Michael, Schwartz, Lauren M., Seder, Robert A., Segalés, Joaquim, Vasan, Seshadri S., Henao-Restrepo, Ana Mariá, and Barouch, Dan H.
- Abstract
The global response to Coronavirus Disease 2019 (COVID-19) is now facing new challenges such as vaccine inequity and the emergence of SARS-CoV-2 variants of concern (VOCs). Preclinical models of disease, in particular animal models, are essential to investigate VOC pathogenesis, vaccine correlates of protection and postexposure therapies. Here, we provide an update from the World Health Organization (WHO) COVID-19 modeling expert group (WHO-COM) assembled by WHO, regarding advances in preclinical models. In particular, we discuss how animal model research is playing a key role to evaluate VOC virulence, transmission and immune escape, and how animal models are being refined to recapitulate COVID-19 demographic variables such as comorbidities and age.
- Published
- 2022
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.