88 results on '"Manicassamy B"'
Search Results
2. RIG-I mediates recognition of Salmonella RNA in non-phagocytic cells and contributes to early bacterial replication in vivo: P133
- Author
-
Schmolke, M., Patel, J., Miller, J., Aparicio, Sanchez M., Manicassamy, B., Merad, M., and Sastre, Garcia A.
- Published
- 2012
- Full Text
- View/download PDF
3. Pan-viral specificity of IFN-induced genes reveals new roles for cGAS in innate immunity (vol 505, pg 691, 2014)
- Author
-
Schoggins, J.W., MacDuff, D.A., Imanaka, N., Gainey, M.D., Shrestha, B., Eitson, J.L., Mar, K.B., Richardson, R.B., Ratushny, A.V., Litvak, V., Dabelic, R., Manicassamy, B., Aitchison, J.D., Aderem, A., Elliott, R.M., Garcia-Sastre, A., Racaniello, V., Snijder, E.J., Yokoyama, W.M., Diamond, M.S., Virgin, H.W., and Rice, C.M.
- Published
- 2015
4. LA PROTEINA V DEL VIRUS LPMV (LA PIEDAD MICHOACAN MESSICO VIRUS) ANTAGONIZZA LA RISPOSTA DI TIPO I DELL’INTERFERONE LEGANDOSI ALLA PROTEINA STAT2 E PREVENENDONE LA TRASLOCAZIONE
- Author
-
PISANELLI, GIUSEPPE, IOVANE, GIUSEPPE, Manicassamy B., Laurent rolle M., Belicha villanueva A., Morrison J., Garcia Sastre A., S.I.Di.L.V. SOCIETÀ ITALIANA DI DIAGNOSTICA DI LABORATORIO VETERINARIA, S.I.Di.L.V., Pisanelli, Giuseppe, Manicassamy, B., Laurent rolle, M., Belicha villanueva, A., Morrison, J., Iovane, Giuseppe, and Garcia Sastre, A.
- Subjects
La piedad michoacan mexico virus - Abstract
LPMV is the etiologic agent of “blue eye disease”, a disease endemic in Mexico, which mainly affects piglets and is characterized by meningoencephalitis and respiratory distress. The V protein, expressed by most Paramyxoviruses, evade the type I and type II IFN responses by targeting the signaling pathway. In this study we set out to determine if LPMV-V protein possesses IFN signaling antagonist activity, and to identify which signaling components if any are targeted by LPMV-V protein. We demonstrate that LPMV-V protein antagonizes type I but not type II IFN signaling by inhibiting STAT2. Our results indicate that the last 25 amino acids of LPMV-V protein bind endogenous STAT2
- Published
- 2012
5. Oseltamivir Prophylaxis Reduces Inflammation and Facilitates Establishment of Cross-Strain Protective T Cell Memory to Influenza Viruses
- Author
-
Manicassamy, B, Bird, NL, Olson, MR, Hurt, AC, Oshansky, CM, Oh, DY, Reading, PC, Chua, BY, Sun, Y, Tang, L, Handel, A, Jackson, DC, Turner, SJ, Thomas, PG, Kedzierska, K, Manicassamy, B, Bird, NL, Olson, MR, Hurt, AC, Oshansky, CM, Oh, DY, Reading, PC, Chua, BY, Sun, Y, Tang, L, Handel, A, Jackson, DC, Turner, SJ, Thomas, PG, and Kedzierska, K
- Abstract
CD8(+) T cells directed against conserved viral regions elicit broad immunity against distinct influenza viruses, promote rapid virus elimination and enhanced host recovery. The influenza neuraminidase inhibitor, oseltamivir, is prescribed for therapy and prophylaxis, although it remains unclear how the drug impacts disease severity and establishment of effector and memory CD8(+) T cell immunity. We dissected the effects of oseltamivir on viral replication, inflammation, acute CD8(+) T cell responses and the establishment of immunological CD8(+) T cell memory. In mice, ferrets and humans, the effect of osteltamivir on viral titre was relatively modest. However, prophylactic oseltamivir treatment in mice markedly reduced morbidity, innate responses, inflammation and, ultimately, the magnitude of effector CD8(+) T cell responses. Importantly, functional memory CD8(+) T cells established during the drug-reduced effector phase were capable of mounting robust recall responses. Moreover, influenza-specific memory CD4(+) T cells could be also recalled after the secondary challenge, while the antibody levels were unaffected. This provides evidence that long-term memory T cells can be generated during an oseltamivir-interrupted infection. The anti-inflammatory effect of oseltamivir was verified in H1N1-infected patients. Thus, in the case of an unpredicted influenza pandemic, while prophylactic oseltamivir treatment can reduce disease severity, the capacity to generate memory CD8(+) T cells specific for the newly emerged virus is uncompromised. This could prove especially important for any new influenza pandemic which often occurs in separate waves.
- Published
- 2015
6. Glycosylations in the globular head of the hemagglutinin protein modulate the virulence and antigenic properties of the H1N1 influenza viruses
- Author
-
Medina, R A, Stertz, S, Manicassamy, B, Zimmermann, P, Sun, X, Albrecht, R A, Uusi-Kerttula, H, Zagordi, O, Belshe, R B, Frey, S E, Tumpey, T M, García-Sastre, A, University of Zurich, and Medina, R A
- Subjects
10028 Institute of Medical Virology ,570 Life sciences ,biology ,610 Medicine & health ,2700 General Medicine - Published
- 2013
7. P133 RIG-I mediates recognition of Salmonella RNA in non-phagocytic cells and contributes to early bacterial replication in vivo
- Author
-
Schmolke, M., primary, Patel, J., additional, Miller, J., additional, Aparicio, M. Sanchez, additional, Manicassamy, B., additional, Merad, M., additional, and Garcia Sastre, A., additional
- Published
- 2012
- Full Text
- View/download PDF
8. Mutations in the NS1 C-terminal tail do not enhance replication or virulence of the 2009 pandemic H1N1 influenza A virus
- Author
-
Hale, B. G., primary, Steel, J., additional, Manicassamy, B., additional, Medina, R. A., additional, Ye, J., additional, Hickman, D., additional, Lowen, A. C., additional, Perez, D. R., additional, and Garcia-Sastre, A., additional
- Published
- 2010
- Full Text
- View/download PDF
9. A frank statement on the health economics franchise
- Author
-
Manicassamy B Soudarssanane
- Subjects
Economic growth ,Health economics ,National Rural Health Mission ,business.industry ,Statement (logic) ,Public Health, Environmental and Occupational Health ,Public administration ,Alma Ata Declaration ,Editorial ,Health care ,National health policy ,Sociology ,Franchise ,business ,Health policy - Abstract
India has achieved notable progress in health after its commitment to the Alma Ata declaration and especially after the revolutionary steps listed in the National Health Policy 2002(1)—particularly in fund allocations. The latest time-bound National Rural Health Mission (NRHM)(2) has ensured a great surge in taking health care closer to the underprivileged populations. Despite this appreciable progress, commitment in further areas demands urgent attention in a competitive environment.
- Published
- 2008
10. Cross-presenting CD103+ dendritic cells are protected from influenza virus infection.
- Author
-
Helft J, Manicassamy B, Guermonprez P, Hashimoto D, Silvin A, Agudo J, Brown BD, Schmolke M, Miller JC, Leboeuf M, Murphy KM, García-Sastre A, Merad M, Helft, Julie, Manicassamy, Balaji, Guermonprez, Pierre, Hashimoto, Daigo, Silvin, Aymeric, Agudo, Judith, and Brown, Brian D
- Subjects
- *
ANIMAL experimentation , *ANTIGENS , *CELL receptors , *CELLS , *CELLULAR immunity , *CELLULAR signal transduction , *DENDRITIC cells , *DOGS , *INTERFERONS , *LUNGS , *MICE , *PROTEINS , *RECOMBINANT proteins , *RESEARCH funding , *T cells , *VIRAL antigens , *INFLUENZA A virus , *ORTHOMYXOVIRUS infections , *PREVENTION - Abstract
CD8+ cytotoxic T cells are critical for viral clearance from the lungs upon influenza virus infection. The contribution of antigen cross-presentation by DCs to the induction of anti-viral cytotoxic T cells remains controversial. Here, we used a recombinant influenza virus expressing a nonstructural 1-GFP (NS1-GFP) reporter gene to visualize the route of antigen presentation by lung DCs upon viral infection in mice. We found that lung CD103+ DCs were the only subset of cells that carried intact GFP protein to the draining LNs. Strikingly, lung migratory CD103+ DCs were not productively infected by influenza virus and thus were able to induce virus-specific CD8+ T cells through the cross-presentation of antigens from virally infected cells. We also observed that CD103+ DC resistance to infection correlates with an increased anti-viral state in these cells that is dependent on the expression of type I IFN receptor. These results show that efficient cross-priming by migratory lung DCs is coupled to the acquisition of an anti-viral status, which is dependent on the type I IFN signaling pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
11. A Frank Statement on the Health Economics Franchise.
- Author
-
Soudarssanane, Manicassamy B.
- Subjects
- *
MEDICAL economics , *PUBLIC health , *PREVENTIVE medicine , *MEDICAL conferences - Abstract
The article talks about health economics with reference to topics discussed at the 35th Annual National Conference of India Association of Preventive and Social Medicine (IAPSM) in Pondicherry in January 2008. It tackles the areas in public health and preventive medicine which have not yet been explored. It notes the importance of parallel development in medical specialty training. It emphasizes the need to raise awareness about health economics in medical education curriculum.
- Published
- 2008
- Full Text
- View/download PDF
12. Pandemic 2009 H1N1 vaccine protects against 1918 Spanish influenza virus
- Author
-
Medina, R. A., Manicassamy, B., Stertz, S., Seibert, C. W., Hai, R., Belshe, R. B., Frey, S. E., Basler, C. F., Palese, P., and Adolfo Garcia-Sastre
13. Expression of Ebolavirus glycoprotein on the target cells enhances viral entry
- Author
-
Manicassamy Balaji and Rong Lijun
- Subjects
Infectious and parasitic diseases ,RC109-216 - Abstract
Abstract Background Entry of Ebolavirus to the target cells is mediated by the viral glycoprotein GP. The native GP exists as a homotrimer on the virions and contains two subunits, a surface subunit (GP1) that is involved in receptor binding and a transmembrane subunit (GP2) that mediates the virus-host membrane fusion. Previously we showed that over-expression of GP on the target cells blocks GP-mediated viral entry, which is mostly likely due to receptor interference by GP1. Results In this study, using a tetracycline inducible system, we report that low levels of GP expression on the target cells, instead of interfering, specifically enhance GP mediated viral entry. Detailed mapping analysis strongly suggests that the fusion subunit GP2 is primarily responsible for this novel phenomenon, here referred to as trans enhancement. Conclusion Our data suggests that GP2 mediated trans enhancement of virus fusion occurs via a mechanism analogous to eukaryotic membrane fusion processes involving specific trans oligomerization and cooperative interaction of fusion mediators. These findings have important implications in our current understanding of virus entry and superinfection interference.
- Published
- 2009
- Full Text
- View/download PDF
14. Dissecting the role of putative CD81 binding regions of E2 in mediating HCV entry: Putative CD81 binding region 1 is not involved in CD81 binding
- Author
-
Uprichard Susan L, Manicassamy Balaji, Rothwangl Katharina B, and Rong Lijun
- Subjects
Infectious and parasitic diseases ,RC109-216 - Abstract
Abstract Background Hepatitis C virus (HCV) encodes two transmembrane glycoproteins E1 and E2 which form a heterodimer. E1 is believed to mediate fusion while E2 has been shown to bind cellular receptors including CD81. In this study, alanine substitutions in E2 were generated within putative CD81 binding regions to define residues critical for viral entry. The effect of each mutation was tested by challenging susceptible cell lines with mutant HCV E1E2 pseudotyped viruses generated using a lentiviral system (HCVpp). In addition to assaying infectivity, producer cell expression and HCVpp incorporation of HCV E1 and E2 proteins, CD81 binding profiles, and E1E2 association of mutants were examined. Results Based on these characteristics, mutants either displayed wt characteristics (high infectivity [≥ 50% of wt HCVpp], CD81 binding, E1E2 expression, association, and incorporation into viral particles and proper conformation) or segregated into 4 distinct low infectivity (≤ 50% of wt HCVpp) mutant phenotypes: (I) CD81 binding deficient (despite wt E1E2 expression, incorporation and association and proper conformation); (II) CD81 binding competent, but lack of E1 detection on the viral particle, (despite adequate E1E2 expression in producer cell lysates and proper conformation); (III) CD81 binding competent, with adequate E1E2 expression, incorporation, association, and proper E2 conformation (i.e. no defect identified to explain the reduced infectivity observed); (IV) CD81 binding deficient due to disruption of E2 mutant protein conformation. Conclusion Although most alanine substitutions within the putative CD81 binding region 1 (amino acids 474–492) displayed greatly reduced HCVpp infectivity, they retained soluble CD81 binding, proper E2 conformation, E1E2 association and incorporation into HCVpp suggesting that region 1 of E2 does not mediate binding to CD81. In contrast, conformationally correct E2 mutants (Y527 and W529) within the second putative CD81 binding region (amino acids 522–551) disrupted binding of E2 to CD81-GST, suggesting that region 2 is critical to CD81 binding. Likewise, all conformationally intact mutants within the third putative CD81 binding region (amino acids 612–619), except L615A, were important for E2 binding to CD81-GST. This region is highly conserved across genotypes, underlining its importance in mediating viral entry.
- Published
- 2008
- Full Text
- View/download PDF
15. Conserved role of spike S2 domain N-glycosylation across beta-coronavirus family.
- Author
-
Yang Q, Kelkar A, Manicassamy B, and Neelamegham S
- Abstract
Besides acting as an immunological shield, the N-glycans of SARS-CoV-2 are also critical for viral life cycle. As the S2 subunit of spike is highly conserved across beta-coronaviruses, we determined the functional significance of the five 'stem N-glycans' located in S2 between N1098-N1194. Studies were performed with 31 Asn-to-Gln mutants, beta-coronavirus virus-like particles and single-cycle viral replicons. Deletions of stem N-glycans enhanced S1 shedding from trimeric spike, reduced ACE2 binding and abolished syncytia formation. When three or more N-glycans were deleted, spike expression on cell surface and incorporation into virions was both reduced. Viral entry function was progressively lost upon deleting the N1098 glycan in combination with additional glycosite modifications. In addition to SARS-CoV-2, deleting stem N-glycans in SARS-CoV and MERS-CoV spike also prevented viral entry into target cells. These data suggest multiple functional roles for the stem N-glycans, and evolutionarily conserved properties for these complex carbohydrates across human beta-coronaviruses., Competing Interests: Declaration of interests The authors declare no completing financial interests.
- Published
- 2024
- Full Text
- View/download PDF
16. A viral assembly inhibitor blocks SARS-CoV-2 replication in airway epithelial cells.
- Author
-
Du L, Deiter F, Bouzidi MS, Billaud JN, Simmons G, Dabral P, Selvarajah S, Lingappa AF, Michon M, Yu SF, Paulvannan K, Manicassamy B, Lingappa VR, Boushey H, Greenland JR, and Pillai SK
- Subjects
- Humans, Virus Assembly drug effects, COVID-19 virology, COVID-19 Drug Treatment, SARS-CoV-2 drug effects, SARS-CoV-2 physiology, Virus Replication drug effects, Epithelial Cells virology, Epithelial Cells drug effects, Epithelial Cells metabolism, Antiviral Agents pharmacology
- Abstract
The ongoing evolution of SARS-CoV-2 to evade vaccines and therapeutics underlines the need for innovative therapies with high genetic barriers to resistance. Therefore, there is pronounced interest in identifying new pharmacological targets in the SARS-CoV-2 viral life cycle. The small molecule PAV-104, identified through a cell-free protein synthesis and assembly screen, was recently shown to target host protein assembly machinery in a manner specific to viral assembly. In this study, we investigate the capacity of PAV-104 to inhibit SARS-CoV-2 replication in human airway epithelial cells (AECs). We show that PAV-104 inhibits >99% of infection with diverse SARS-CoV-2 variants in immortalized AECs, and in primary human AECs cultured at the air-liquid interface (ALI) to represent the lung microenvironment in vivo. Our data demonstrate that PAV-104 inhibits SARS-CoV-2 production without affecting viral entry, mRNA transcription, or protein synthesis. PAV-104 interacts with SARS-CoV-2 nucleocapsid (N) and interferes with its oligomerization, blocking particle assembly. Transcriptomic analysis reveals that PAV-104 reverses SARS-CoV-2 induction of the type-I interferon response and the maturation of nucleoprotein signaling pathway known to support coronavirus replication. Our findings suggest that PAV-104 is a promising therapeutic candidate for COVID-19 with a mechanism of action that is distinct from existing clinical management approaches., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
17. An orally active entry inhibitor of influenza A viruses protects mice and synergizes with oseltamivir and baloxavir marboxil.
- Author
-
Gaisina I, Li P, Du R, Cui Q, Dong M, Zhang C, Manicassamy B, Caffrey M, Moore T, Cooper L, and Rong L
- Subjects
- Animals, Mice, Oseltamivir pharmacology, Oseltamivir therapeutic use, Antiviral Agents therapeutic use, Oxazines pharmacology, Oxazines therapeutic use, Pyridines, Influenza A virus, Thiepins pharmacology, Thiepins therapeutic use, Dibenzothiepins, Morpholines, Pyridones, Triazines
- Abstract
Seasonal or pandemic illness caused by influenza A viruses (IAVs) is a major public health concern due to the high morbidity and notable mortality. Although there are several approved drugs targeting different mechanisms, the emergence of drug resistance calls for new drug candidates that can be used alone or in combinations. Small-molecule IAV entry inhibitor, ING-1466, binds to hemagglutinin (HA) and blocks HA-mediated viral infection. Here, we show that this inhibitor demonstrates preventive and therapeutic effects in a mouse model of IAV with substantial improvement in the survival rate. When administered orally it elicits a therapeutic effect in mice, even after the well-established infection. Moreover, the combination of ING-1466 with oseltamivir phosphate or baloxavir marboxil enhances the therapeutic effect in a synergistic manner. Overall, ING-1466 has excellent oral bioavailability and in vitro absorption, distribution, metabolism, excretion, and toxicity profile, suggesting that it can be developed for monotherapy or combination therapy for the treatment of IAV infections.
- Published
- 2024
- Full Text
- View/download PDF
18. Avian influenza A viruses exhibit plasticity in sialylglycoconjugate receptor usage in human lung cells.
- Author
-
Liang C-Y, Huang I, Han J, Sownthirarajan B, Kulhankova K, Murray NB, Taherzadeh M, Archer-Hartmann S, Pepi L, Manivasagam S, Plung J, Sturtz M, Yu Y, Vogel OA, Kandasamy M, Gourronc FA, Klingelhutz AJ, Choudhury B, Rong L, Perez JT, Azadi P, McCray PB Jr, Neelamegham S, and Manicassamy B
- Subjects
- Animals, Humans, Carrier Proteins metabolism, Glycoconjugates metabolism, N-Acetylneuraminic Acid metabolism, Polysaccharides metabolism, Sugars metabolism, Influenza in Birds metabolism, Receptors, Virus metabolism, Influenza A virus metabolism, Influenza, Human, Lung virology, Receptors, Cell Surface metabolism
- Abstract
Importance: It is well known that influenza A viruses (IAV) initiate host cell infection by binding to sialic acid, a sugar molecule present at the ends of various sugar chains called glycoconjugates. These sugar chains can vary in chain length, structure, and composition. However, it remains unknown if IAV strains preferentially bind to sialic acid on specific glycoconjugate type(s) for host cell infection. Here, we utilized CRISPR gene editing to abolish sialic acid on different glycoconjugate types in human lung cells, and evaluated human versus avian IAV infections. Our studies show that both human and avian IAV strains can infect human lung cells by utilizing any of the three major sialic acid-containing glycoconjugate types, specifically N-glycans, O-glycans, and glycolipids. Interestingly, simultaneous elimination of sialic acid on all three major glycoconjugate types in human lung cells dramatically decreased human IAV infection, yet had little effect on avian IAV infection. These studies show that avian IAV strains effectively utilize other less prevalent glycoconjugates for infection, whereas human IAV strains rely on a limited repertoire of glycoconjugate types. The remarkable ability of avian IAV strains to utilize diverse glycoconjugate types may allow for easy transmission into new host species., Competing Interests: The authors declare no conflict of interest.
- Published
- 2023
- Full Text
- View/download PDF
19. Virology under the Microscope-a Call for Rational Discourse.
- Author
-
Goodrum F, Lowen AC, Lakdawala S, Alwine J, Casadevall A, Imperiale MJ, Atwood W, Avgousti D, Baines J, Banfield B, Banks L, Bhaduri-McIntosh S, Bhattacharya D, Blanco-Melo D, Bloom D, Boon A, Boulant S, Brandt C, Broadbent A, Brooke C, Cameron C, Campos S, Caposio P, Chan G, Cliffe A, Coffin J, Collins K, Damania B, Daugherty M, Debbink K, DeCaprio J, Dermody T, Dikeakos J, DiMaio D, Dinglasan R, Duprex WP, Dutch R, Elde N, Emerman M, Enquist L, Fane B, Fernandez-Sesma A, Flenniken M, Frappier L, Frieman M, Frueh K, Gack M, Gaglia M, Gallagher T, Galloway D, García-Sastre A, Geballe A, Glaunsinger B, Goff S, Greninger A, Hancock M, Harris E, Heaton N, Heise M, Heldwein E, Hogue B, Horner S, Hutchinson E, Hyser J, Jackson W, Kalejta R, Kamil J, Karst S, Kirchhoff F, Knipe D, Kowalik T, Lagunoff M, Laimins L, Langlois R, Lauring A, Lee B, Leib D, Liu SL, Longnecker R, Lopez C, Luftig M, Lund J, Manicassamy B, McFadden G, McIntosh M, Mehle A, Miller WA, Mohr I, Moody C, Moorman N, Moscona A, Mounce B, Munger J, Münger K, Murphy E, Naghavi M, Nelson J, Neufeldt C, Nikolich J, O'Connor C, Ono A, Orenstein W, Ornelles D, Ou JH, Parker J, Parrish C, Pekosz A, Pellett P, Pfeiffer J, Plemper R, Polyak S, Purdy J, Pyeon D, Quinones-Mateu M, Renne R, Rice C, Schoggins J, Roller R, Russell C, Sandri-Goldin R, Sapp M, Schang L, Schmid S, Schultz-Cherry S, Semler B, Shenk T, Silvestri G, Simon V, Smith G, Smith J, Spindler K, Stanifer M, Subbarao K, Sundquist W, Suthar M, Sutton T, Tai A, Tarakanova V, tenOever B, Tibbetts S, Tompkins S, Toth Z, van Doorslaer K, Vignuzzi M, Wallace N, Walsh D, Weekes M, Weinberg J, Weitzman M, Weller S, Whelan S, White E, Williams B, Wobus C, Wong S, and Yurochko A
- Subjects
- Humans, SARS-CoV-2, Pandemics prevention & control, Antiviral Agents, COVID-19 prevention & control, Viruses
- Abstract
Viruses have brought humanity many challenges: respiratory infection, cancer, neurological impairment and immunosuppression to name a few. Virology research over the last 60+ years has responded to reduce this disease burden with vaccines and antivirals. Despite this long history, the COVID-19 pandemic has brought unprecedented attention to the field of virology. Some of this attention is focused on concern about the safe conduct of research with human pathogens. A small but vocal group of individuals has seized upon these concerns - conflating legitimate questions about safely conducting virus-related research with uncertainties over the origins of SARS-CoV-2. The result has fueled public confusion and, in many instances, ill-informed condemnation of virology. With this article, we seek to promote a return to rational discourse. We explain the use of gain-of-function approaches in science, discuss the possible origins of SARS-CoV-2 and outline current regulatory structures that provide oversight for virological research in the United States. By offering our expertise, we - a broad group of working virologists - seek to aid policy makers in navigating these controversial issues. Balanced, evidence-based discourse is essential to addressing public concern while maintaining and expanding much-needed research in virology.
- Published
- 2023
- Full Text
- View/download PDF
20. Synthesis, Optimization, and Structure-Activity Relationships of Imidazo[1,2- a ]pyrimidines as Inhibitors of Group 2 Influenza A Viruses.
- Author
-
Alqarni S, Cooper L, Galvan Achi J, Bott R, Sali VK, Brown A, Santarsiero BD, Krunic A, Manicassamy B, Peet NP, Zhang P, Thatcher GRJ, Gaisina IN, Rong L, and Moore TW
- Subjects
- Humans, Oseltamivir, Hemagglutinin Glycoproteins, Influenza Virus metabolism, Hemagglutinins, Structure-Activity Relationship, Pyrimidines pharmacology, Antiviral Agents pharmacology, Antiviral Agents chemistry, Influenza A virus metabolism, Influenza, Human drug therapy
- Abstract
The influenza A virus (IAV) is a highly contagious virus that causes pandemics and seasonal epidemics, which are major public health issues. Current anti-influenza therapeutics are limited partly due to the continuous emergence of drug-resistant IAV strains; thus, there is an unmet need to develop novel anti-influenza therapies. Here, we present a novel imidazo[1,2- a ]pyrimidine scaffold that targets group 2 IAV entry. We have explored three different regions of the lead compound, and we have developed a series of small molecules that have nanomolar activity against oseltamivir-sensitive and -resistant forms of group 2 IAVs. These small molecules target hemagglutinin (HA), which mediates the viral entry process. Mapping a known small-molecule-binding cavity of the HA structure with resistant mutants suggests that these molecules bind to that cavity and block HA-mediated membrane fusion.
- Published
- 2022
- Full Text
- View/download PDF
21. Expanding the tolerance of segmented Influenza A Virus genome using a balance compensation strategy.
- Author
-
Zhao X, Lin X, Li P, Chen Z, Zhang C, Manicassamy B, Rong L, Cui Q, and Du R
- Subjects
- Animals, Humans, Mice, Virus Replication genetics, Influenza A virus genetics, Influenza, Human genetics
- Abstract
Reporter viruses provide powerful tools for both basic and applied virology studies, however, the creation and exploitation of reporter influenza A viruses (IAVs) have been hindered by the limited tolerance of the segmented genome to exogenous modifications. Interestingly, our previous study has demonstrated the underlying mechanism that foreign insertions reduce the replication/transcription capacity of the modified segment, impairing the delicate balance among the multiple segments during IAV infection. In the present study, we developed a "balance compensation" strategy by incorporating additional compensatory mutations during initial construction of recombinant IAVs to expand the tolerance of IAV genome. As a proof of concept, promoter-enhancing mutations were introduced within the modified segment to rectify the segments imbalance of a reporter influenza PR8-NS-Gluc virus, while directed optimization of the recombinant IAV was successfully achieved. Further, we generated recombinant IAVs expressing a much larger firefly luciferase (Fluc) by coupling with a much stronger compensatory enhancement, and established robust Fluc-based live-imaging mouse models of IAV infection. Our strategy feasibly expands the tolerance for foreign gene insertions in the segmented IAV genome, which opens up better opportunities to develop more versatile reporter IAVs as well as live attenuated influenza virus-based vaccines for other important human pathogens., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2022
- Full Text
- View/download PDF
22. Development of a Single-Cycle Infectious SARS-CoV-2 Virus Replicon Particle System for Use in Biosafety Level 2 Laboratories.
- Author
-
Malicoat J, Manivasagam S, Zuñiga S, Sola I, McCabe D, Rong L, Perlman S, Enjuanes L, and Manicassamy B
- Subjects
- COVID-19 virology, Cell Culture Techniques, Cell Line, Containment of Biohazards standards, Genes, Reporter, Humans, Laboratories standards, Viral Proteins genetics, Virus Replication, Genetic Engineering, Recombination, Genetic, Replicon, SARS-CoV-2 genetics
- Abstract
Research activities with infectious severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are currently permitted only under biosafety level 3 (BSL3) containment. Here, we report the development of a single-cycle infectious SARS-CoV-2 virus replicon particle (VRP) system with a luciferase and green fluorescent protein (GFP) dual reporter that can be safely handled in BSL2 laboratories to study SARS-CoV-2 biology. The spike (S) gene of SARS-CoV-2 encodes the envelope glycoprotein, which is essential for mediating infection of new host cells. Through deletion and replacement of this essential S gene with a luciferase and GFP dual reporter, we have generated a conditional SARS-CoV-2 mutant (ΔS-VRP) that produces infectious particles only in cells expressing a viral envelope glycoprotein of choice. Interestingly, we observed more efficient production of infectious particles in cells expressing vesicular stomatitis virus (VSV) glycoprotein G [ΔS-VRP(G)] than in cells expressing other viral glycoproteins, including S. We confirmed that infection from ΔS-VRP(G) is limited to a single round and can be neutralized by anti-VSV serum. In our studies with ΔS-VRP(G), we observed robust expression of both luciferase and GFP reporters in various human and murine cell types, demonstrating that a broad variety of cells can support intracellular replication of SARS-CoV-2. In addition, treatment of ΔS-VRP(G)-infected cells with either of the anti-CoV drugs remdesivir (nucleoside analog) and GC376 (CoV 3CL protease inhibitor) resulted in a robust decrease in both luciferase and GFP expression in a drug dose- and cell-type-dependent manner. Taken together, our findings show that we have developed a single-cycle infectious SARS-CoV-2 VRP system that serves as a versatile platform to study SARS-CoV-2 intracellular biology and to perform high-throughput screening of antiviral drugs under BSL2 containment. IMPORTANCE Due to the highly contagious nature of SARS-CoV-2 and the lack of immunity in the human population, research on SARS-CoV-2 has been restricted to biosafety level 3 laboratories. This has greatly limited participation of the broader scientific community in SARS-CoV-2 research and thus has hindered the development of vaccines and antiviral drugs. By deleting the essential spike gene in the viral genome, we have developed a conditional mutant of SARS-CoV-2 with luciferase and fluorescent reporters, which can be safely used under biosafety level 2 conditions. Our single-cycle infectious SARS-CoV-2 virus replicon system can serve as a versatile platform to study SARS-CoV-2 intracellular biology and to perform high-throughput screening of antiviral drugs under BSL2 containment.
- Published
- 2022
- Full Text
- View/download PDF
23. Generation of potent cellular and humoral immunity against SARS-CoV-2 antigens via conjugation to a polymeric glyco-adjuvant.
- Author
-
Gray LT, Raczy MM, Briquez PS, Marchell TM, Alpar AT, Wallace RP, Volpatti LR, Sasso MS, Cao S, Nguyen M, Mansurov A, Budina E, Watkins EA, Solanki A, Mitrousis N, Reda JW, Yu SS, Tremain AC, Wang R, Nicolaescu V, Furlong K, Dvorkin S, Manicassamy B, Randall G, Wilson DS, Kwissa M, Swartz MA, and Hubbell JA
- Subjects
- Adjuvants, Immunologic, Aged, Animals, Antibodies, Neutralizing, Antibodies, Viral, CD8-Positive T-Lymphocytes, COVID-19 Vaccines, Humans, Immunity, Cellular, Mice, SARS-CoV-2, COVID-19, Immunity, Humoral
- Abstract
The SARS-CoV-2 virus has caused an unprecedented global crisis, and curtailing its spread requires an effective vaccine which elicits a diverse and robust immune response. We have previously shown that vaccines made of a polymeric glyco-adjuvant conjugated to an antigen were effective in triggering such a response in other disease models and hypothesized that the technology could be adapted to create an effective vaccine against SARS-CoV-2. The core of the vaccine platform is the copolymer p(Man-TLR7), composed of monomers with pendant mannose or a toll-like receptor 7 (TLR7) agonist. Thus, p(Man-TLR7) is designed to target relevant antigen-presenting cells (APCs) via mannose-binding receptors and then activate TLR7 upon endocytosis. The p(Man-TLR7) construct is amenable to conjugation to protein antigens such as the Spike protein of SARS-CoV-2, yielding Spike-p(Man-TLR7). Here, we demonstrate Spike-p(Man-TLR7) vaccination elicits robust antigen-specific cellular and humoral responses in mice. In adult and elderly wild-type mice, vaccination with Spike-p(Man-TLR7) generates high and long-lasting titers of anti-Spike IgGs, with neutralizing titers exceeding levels in convalescent human serum. Interestingly, adsorbing Spike-p(Man-TLR7) to the depot-forming adjuvant alum amplified the broadly neutralizing humoral responses to levels matching those in mice vaccinated with formulations based off of clinically-approved adjuvants. Additionally, we observed an increase in germinal center B cells, antigen-specific antibody secreting cells, activated T follicular helper cells, and polyfunctional Th1-cytokine producing CD4
+ and CD8+ T cells. We conclude that Spike-p(Man-TLR7) is an attractive, next-generation subunit vaccine candidate, capable of inducing durable and robust antibody and T cell responses., (Copyright © 2021 The Authors. Published by Elsevier Ltd.. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
24. Rab11a mediates cell-cell spread and reassortment of influenza A virus genomes via tunneling nanotubes.
- Author
-
Ganti K, Han J, Manicassamy B, and Lowen AC
- Subjects
- A549 Cells, Animals, Dogs, Humans, Influenza A virus genetics, Influenza, Human genetics, Madin Darby Canine Kidney Cells, Nanotubes, Cell Communication, Influenza A virus pathogenicity, Influenza, Human virology, rab GTP-Binding Proteins metabolism
- Abstract
Influenza A virus [IAV] genomes comprise eight negative strand RNAs packaged into virions in the form of viral ribonucleoproteins [vRNPs]. Rab11a plays a crucial role in the transport of vRNPs from the nucleus to the plasma membrane via microtubules, allowing assembly and virus production. Here, we identify a novel function for Rab11a in the inter-cellular transport of IAV vRNPs using tunneling nanotubes [TNTs]as molecular highways. TNTs are F-Actin rich tubules that link the cytoplasm of nearby cells. In IAV-infected cells, Rab11a was visualized together with vRNPs in these actin-rich intercellular connections. To better examine viral spread via TNTs, we devised an infection system in which conventional, virion-mediated, spread was not possible. Namely, we generated HA-deficient reporter viruses which are unable to produce progeny virions but whose genomes can be replicated and trafficked. In this system, vRNP transfer to neighboring cells was observed and this transfer was found to be dependent on both actin and Rab11a. Generation of infectious virus via TNT transfer was confirmed using donor cells infected with HA-deficient virus and recipient cells stably expressing HA protein. Mixing donor cells infected with genetically distinct IAVs furthermore revealed the potential for Rab11a and TNTs to serve as a conduit for genome mixing and reassortment in IAV infections. These data therefore reveal a novel role for Rab11a in the IAV life cycle, which could have significant implications for within-host spread, genome reassortment and immune evasion., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
25. Polymersomes Decorated with the SARS-CoV-2 Spike Protein Receptor-Binding Domain Elicit Robust Humoral and Cellular Immunity.
- Author
-
Volpatti LR, Wallace RP, Cao S, Raczy MM, Wang R, Gray LT, Alpar AT, Briquez PS, Mitrousis N, Marchell TM, Sasso MS, Nguyen M, Mansurov A, Budina E, Solanki A, Watkins EA, Schnorenberg MR, Tremain AC, Reda JW, Nicolaescu V, Furlong K, Dvorkin S, Yu SS, Manicassamy B, LaBelle JL, Tirrell MV, Randall G, Kwissa M, Swartz MA, and Hubbell JA
- Abstract
The COVID-19 pandemic underscores the need for rapid, safe, and effective vaccines. In contrast to some traditional vaccines, nanoparticle-based subunit vaccines are particularly efficient in trafficking antigens to lymph nodes, where they induce potent immune cell activation. Here, we developed a strategy to decorate the surface of oxidation-sensitive polymersomes with multiple copies of the SARS-CoV-2 spike protein receptor-binding domain (RBD) to mimic the physical form of a virus particle. We evaluated the vaccination efficacy of these surface-decorated polymersomes (RBD
surf ) in mice compared to RBD-encapsulated polymersomes (RBDencap ) and unformulated RBD (RBDfree ), using monophosphoryl-lipid-A-encapsulated polymersomes (MPLA PS) as an adjuvant. While all three groups produced high titers of RBD-specific IgG, only RBDsurf elicited a neutralizing antibody response to SARS-CoV-2 comparable to that of human convalescent plasma. Moreover, RBDsurf was the only group to significantly increase the proportion of RBD-specific germinal center B cells in the vaccination-site draining lymph nodes. Both RBDsurf and RBDencap drove similarly robust CD4+ and CD8+ T cell responses that produced multiple Th1-type cytokines. We conclude that a multivalent surface display of spike RBD on polymersomes promotes a potent neutralizing antibody response to SARS-CoV-2, while both antigen formulations promote robust T cell immunity., Competing Interests: The authors declare the following competing financial interest(s): M.A.S. and J.A.H. have patents related to the polymersome technology and interests in LantaBio, which has licensed those patents., (© 2021 The Authors. Published by American Chemical Society.)- Published
- 2021
- Full Text
- View/download PDF
26. Host factor Rab11a is critical for efficient assembly of influenza A virus genomic segments.
- Author
-
Han J, Ganti K, Sali VK, Twigg C, Zhang Y, Manivasagam S, Liang CY, Vogel OA, Huang I, Emmanuel SN, Plung J, Radoshevich L, Perez JT, Lowen AC, and Manicassamy B
- Subjects
- A549 Cells, HEK293 Cells, Humans, Influenza A virus isolation & purification, Influenza, Human genetics, Ribonucleoproteins genetics, Viral Proteins genetics, Virus Replication, rab GTP-Binding Proteins genetics, Genome, Viral, Influenza A virus genetics, Influenza, Human virology, Ribonucleoproteins metabolism, Viral Proteins metabolism, Virus Assembly, rab GTP-Binding Proteins metabolism
- Abstract
It is well documented that influenza A viruses selectively package 8 distinct viral ribonucleoprotein complexes (vRNPs) into each virion; however, the role of host factors in genome assembly is not completely understood. To evaluate the significance of cellular factors in genome assembly, we generated a reporter virus carrying a tetracysteine tag in the NP gene (NP-Tc virus) and assessed the dynamics of vRNP localization with cellular components by fluorescence microscopy. At early time points, vRNP complexes were preferentially exported to the MTOC; subsequently, vRNPs associated on vesicles positive for cellular factor Rab11a and formed distinct vRNP bundles that trafficked to the plasma membrane on microtubule networks. In Rab11a deficient cells, however, vRNP bundles were smaller in the cytoplasm with less co-localization between different vRNP segments. Furthermore, Rab11a deficiency increased the production of non-infectious particles with higher RNA copy number to PFU ratios, indicative of defects in specific genome assembly. These results indicate that Rab11a+ vesicles serve as hubs for the congregation of vRNP complexes and enable specific genome assembly through vRNP:vRNP interactions, revealing the importance of Rab11a as a critical host factor for influenza A virus genome assembly., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
27. Polymersomes decorated with SARS-CoV-2 spike protein receptor binding domain elicit robust humoral and cellular immunity.
- Author
-
Volpatti LR, Wallace RP, Cao S, Raczy MM, Wang R, Gray LT, Alpar AT, Briquez PS, Mitrousis N, Marchell TM, Sasso MS, Nguyen M, Mansurov A, Budina E, Solanki A, Watkins EA, Schnorenberg MR, Tremain AC, Reda JW, Nicolaescu V, Furlong K, Dvorkin S, Yu SS, Manicassamy B, LaBelle JL, Tirrell MV, Randall G, Kwissa M, Swartz MA, and Hubbell JA
- Abstract
A diverse portfolio of SARS-CoV-2 vaccine candidates is needed to combat the evolving COVID-19 pandemic. Here, we developed a subunit nanovaccine by conjugating SARS-CoV-2 Spike protein receptor binding domain (RBD) to the surface of oxidation-sensitive polymersomes. We evaluated the humoral and cellular responses of mice immunized with these surface-decorated polymersomes (RBD
surf ) compared to RBD-encapsulated polymersomes (RBDencap ) and unformulated RBD (RBDfree ), using monophosphoryl lipid A-encapsulated polymersomes (MPLA PS) as an adjuvant. While all three groups produced high titers of RBD-specific IgG, only RBDsurf elicited a neutralizing antibody response to SARS-CoV-2 comparable to that of human convalescent plasma. Moreover, RBDsurf was the only group to significantly increase the proportion of RBD-specific germinal center B cells in the vaccination-site draining lymph nodes. Both RBDsurf and RBDencap drove similarly robust CD4+ and CD8+ T cell responses that produced multiple Th1-type cytokines. We conclude that multivalent surface display of Spike RBD on polymersomes promotes a potent neutralizing antibody response to SARS-CoV-2, while both antigen formulations promote robust T cell immunity., Competing Interests: COMPETING INTERESTS M.A.S. and J.A.H. have patents related to the polymersome technology and interests in LantaBio, which has licensed those patents.- Published
- 2021
- Full Text
- View/download PDF
28. The p150 Isoform of ADAR1 Blocks Sustained RLR signaling and Apoptosis during Influenza Virus Infection.
- Author
-
Vogel OA, Han J, Liang CY, Manicassamy S, Perez JT, and Manicassamy B
- Subjects
- A549 Cells, Adenosine Deaminase genetics, DEAD Box Protein 58 genetics, HEK293 Cells, Humans, Influenza, Human genetics, Isoenzymes genetics, Isoenzymes metabolism, RNA-Binding Proteins genetics, Receptors, Immunologic, Adenosine Deaminase metabolism, Apoptosis, DEAD Box Protein 58 metabolism, Influenza A virus physiology, Influenza, Human metabolism, RNA-Binding Proteins metabolism, Signal Transduction, Virus Replication
- Abstract
Signaling through retinoic acid inducible gene I (RIG-I) like receptors (RLRs) is tightly regulated, with activation occurring upon sensing of viral nucleic acids, and suppression mediated by negative regulators. Under homeostatic conditions aberrant activation of melanoma differentiation-associated protein-5 (MDA5) is prevented through editing of endogenous dsRNA by RNA editing enzyme Adenosine Deaminase Acting on RNA (ADAR1). In addition, ADAR1 is postulated to play pro-viral and antiviral roles during viral infections that are dependent or independent of RNA editing activity. Here, we investigated the importance of ADAR1 isoforms in modulating influenza A virus (IAV) replication and revealed the opposing roles for ADAR1 isoforms, with the nuclear p110 isoform restricting versus the cytoplasmic p150 isoform promoting IAV replication. Importantly, we demonstrate that p150 is critical for preventing sustained RIG-I signaling, as p150 deficient cells showed increased IFN-β expression and apoptosis during IAV infection, independent of RNA editing activity. Taken together, the p150 isoform of ADAR1 is important for preventing sustained RIG-I induced IFN-β expression and apoptosis during viral infection., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
29. Structure of avian influenza hemagglutinin in complex with a small molecule entry inhibitor.
- Author
-
Antanasijevic A, Durst MA, Cheng H, Gaisina IN, Perez JT, Manicassamy B, Rong L, Lavie A, and Caffrey M
- Subjects
- Animals, Antiviral Agents pharmacology, Binding Sites drug effects, Birds virology, Crystallography, X-Ray methods, Hemagglutinin Glycoproteins, Influenza Virus chemistry, Hemagglutinin Glycoproteins, Influenza Virus metabolism, Hemagglutinins metabolism, Humans, Influenza A Virus, H5N1 Subtype chemistry, Influenza A Virus, H5N1 Subtype drug effects, Influenza in Birds metabolism, Influenza, Human metabolism, Models, Molecular, Orthomyxoviridae Infections, Virus Internalization drug effects, Antiviral Agents chemistry, Hemagglutinins ultrastructure
- Abstract
HA plays a critical role in influenza infection and, thus HA is a potential target for antivirals. Recently, our laboratories have described a novel fusion inhibitor, termed CBS1117, with EC
50 ∼3 μM against group 1 HA. In this work, we characterize the binding properties of CBS1117 to avian H5 HA by x-ray crystallography, NMR, and mutagenesis. The x-ray structure of the complex shows that the compound binds near the HA fusion peptide, a region that plays a critical role in HA-mediated fusion. NMR studies demonstrate binding of CBS1117 to H5 HA in solution and show extensive hydrophobic contacts between the compound and HA surface. Mutagenesis studies further support the location of the compound binding site proximal to the HA fusion peptide and identify additional amino acids that are important to compound binding. Together, this work gives new insights into the CBS1117 mechanism of action and can be exploited to further optimize this compound and better understand the group specific activity of small-molecule inhibitors of HA-mediated entry., (© 2020 Antanasijevic et al.)- Published
- 2020
- Full Text
- View/download PDF
30. Identification of entry inhibitors with 4-aminopiperidine scaffold targeting group 1 influenza A virus.
- Author
-
Hussein AFA, Cheng H, Tundup S, Antanasijevic A, Varhegyi E, Perez J, AbdulRahman EM, Elenany MG, Helal S, Caffrey M, Peet N, Manicassamy B, and Rong L
- Subjects
- A549 Cells, Humans, Influenza A Virus, H1N1 Subtype drug effects, Influenza A Virus, H1N1 Subtype physiology, Influenza A Virus, H5N1 Subtype drug effects, Influenza A Virus, H5N1 Subtype physiology, Influenza A virus physiology, Small Molecule Libraries pharmacology, Structure-Activity Relationship, Virus Replication drug effects, Antiviral Agents pharmacology, Influenza A virus drug effects, Piperidines pharmacology, Virus Internalization drug effects
- Abstract
Influenza A viruses (IAVs) cause seasonal flu and occasionally pandemics. The current therapeutics against IAVs target two viral proteins - neuraminidase (NA) and M2 ion-channel protein. However, M2 ion channel inhibitors (amantadine and rimantadine) are no longer recommended by CDC for use due to the emergence of high level of antiviral resistance among the circulating influenza viruses, and resistant strains to NA inhibitors (oseltamivir and zanamivir) have also been reported. Therefore, development of novel anti-influenza therapies is urgently needed. As one of the viral surface glycoproteins, hemagglutinin (HA) mediates critical virus entry steps including virus binding to host cells and virus-host membrane fusion, which makes it a potential target for anti-influenza drug development. In this study, we report the identification of compound CBS1116 with a 4-aminopiperidine scaffold from a chemical library screen as an entry inhibitor specifically targeting two group 1 influenza A viruses, A/Puerto Rico/8/34 (H1N1) and recombinant low pathogenic avian H5N1 virus (A/Vietnam/1203/04, VN04
Low ). Mechanism of action studies show that CBS1116 interferes with the HA-mediated fusion process. Further structure activity relationship study generated a more potent compound CBS1117 which has a 50% inhibitory concentration of 70 nM and a selectivity index of ~4000 against A/Puerto Rico/8/34 (H1N1) infection in human lung epithelial cell line (A549)., (Copyright © 2020 Elsevier B.V. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
31. Suppression of Cytotoxic T Cell Functions and Decreased Levels of Tissue-Resident Memory T Cells during H5N1 Infection.
- Author
-
Kandasamy M, Furlong K, Perez JT, Manicassamy S, and Manicassamy B
- Subjects
- Animals, Birds, Humans, Immunity, Innate immunology, Immunologic Memory immunology, Influenza A Virus, H1N1 Subtype immunology, Influenza A Virus, H3N2 Subtype immunology, Influenza A Virus, H5N1 Subtype metabolism, Influenza A virus physiology, Influenza, Human metabolism, Lung virology, Mice, Orthomyxoviridae Infections virology, Virus Replication genetics, Influenza A Virus, H5N1 Subtype genetics, Influenza, Human immunology, T-Lymphocytes, Cytotoxic immunology
- Abstract
Seasonal influenza virus infections cause mild illness in healthy adults, as timely viral clearance is mediated by the functions of cytotoxic T cells. However, avian H5N1 influenza virus infections can result in prolonged and fatal illness across all age groups, which has been attributed to the overt and uncontrolled activation of host immune responses. Here, we investigate how excessive innate immune responses to H5N1 impair subsequent adaptive T cell responses in the lungs. Using recombinant H1N1 and H5N1 strains sharing 6 internal genes, we demonstrate that H5N1 (2:6) infection in mice causes higher stimulation and increased migration of lung dendritic cells to the draining lymph nodes, resulting in greater numbers of virus-specific T cells in the lungs. Despite robust T cell responses in the lungs, H5N1 (2:6)-infected mice showed inefficient and delayed viral clearance compared with H1N1-infected mice. In addition, we observed higher levels of inhibitory signals, including increased PD-1 and interleukin-10 (IL-10) expression by cytotoxic T cells in H5N1 (2:6)-infected mice, suggesting that delayed viral clearance of H5N1 (2:6) was due to the suppression of T cell functions in vivo Importantly, H5N1 (2:6)-infected mice displayed decreased numbers of tissue-resident memory T cells compared with H1N1-infected mice; however, despite the decreased number of tissue-resident memory T cells, H5N1 (2:6) was protected against a heterologous challenge from H3N2 virus (X31). Taken together, our study provides mechanistic insight for the prolonged viral replication and protracted illness observed in H5N1-infected patients. IMPORTANCE Influenza viruses cause upper respiratory tract infections in humans. In healthy adults, seasonal influenza virus infections result in mild disease. Occasionally, influenza viruses endemic in domestic birds can cause severe and fatal disease even in healthy individuals. In avian influenza virus-infected patients, the host immune system is activated in an uncontrolled manner and is unable to control infection in a timely fashion. In this study, we investigated why the immune system fails to effectively control a modified form of avian influenza virus. Our studies show that T cell functions important for clearing virally infected cells are impaired by higher negative regulatory signals during modified avian influenza virus infection. In addition, memory T cell numbers were decreased in modified avian influenza virus-infected mice. Our studies provide a possible mechanism for the severe and prolonged disease associated with avian influenza virus infections in humans., (Copyright © 2020 Kandasamy et al.)
- Published
- 2020
- Full Text
- View/download PDF
32. Optimization of 4-Aminopiperidines as Inhibitors of Influenza A Viral Entry That Are Synergistic with Oseltamivir.
- Author
-
Gaisina IN, Peet NP, Cheng H, Li P, Du R, Cui Q, Furlong K, Manicassamy B, Caffrey M, Thatcher GRJ, and Rong L
- Subjects
- Animals, Antiviral Agents chemical synthesis, Antiviral Agents metabolism, Binding Sites, Dogs, Drug Synergism, Hemagglutinins, Viral chemistry, Hemagglutinins, Viral metabolism, Humans, Influenza A Virus, H1N1 Subtype chemistry, Influenza A Virus, H5N1 Subtype chemistry, Madin Darby Canine Kidney Cells, Mice, Microbial Sensitivity Tests, Microsomes, Liver metabolism, Molecular Docking Simulation, Molecular Structure, Piperidines chemical synthesis, Piperidines metabolism, Protein Binding, Small Molecule Libraries chemical synthesis, Small Molecule Libraries pharmacology, Structure-Activity Relationship, Antiviral Agents pharmacology, Influenza A Virus, H1N1 Subtype drug effects, Influenza A Virus, H5N1 Subtype drug effects, Oseltamivir pharmacology, Piperidines pharmacology, Virus Internalization drug effects
- Abstract
Vaccination is the most prevalent prophylactic means for controlling seasonal influenza infections. However, an effective vaccine usually takes at least 6 months to develop for the circulating strains. Therefore, new therapeutic options are needed for the acute treatment of influenza infections to control this virus and prevent epidemics/pandemics from developing. We have discovered fast-acting, orally bioavailable acylated 4-aminopiperidines with an effective mechanism of action targeting viral hemagglutinin (HA). Our data show that these compounds are potent entry inhibitors of influenza A viruses. We present docking studies that suggest an HA binding site for these inhibitors on H5N1. Compound 16 displayed a significant decrease of viral titer when evaluated in the infectious assays with influenza virus H1N1 (A/Puerto Rico/8/1934) or H5N1 (A/Vietnam/1203/2004) strains and the oseltamivir-resistant strain with the most common H274Y mutation. In addition, compound 16 showed significant synergistic activity with oseltamivir in vitro.
- Published
- 2020
- Full Text
- View/download PDF
33. Varying Inoculum Dose to Assess the Roles of the Immune Response and Target Cell Depletion by the Pathogen in Control of Acute Viral Infections.
- Author
-
Moore JR, Ahmed H, Manicassamy B, Garcia-Sastre A, Handel A, and Antia R
- Subjects
- Adaptive Immunity, Animals, Disease Models, Animal, Host Microbial Interactions immunology, Humans, Immunity, Innate, Mathematical Concepts, Mice, Orthomyxoviridae Infections immunology, Orthomyxoviridae Infections virology, Viral Load, Models, Immunological, Virus Diseases immunology, Virus Diseases virology
- Abstract
It is difficult to determine whether an immune response or target cell depletion by the infectious agent is most responsible for the control of acute primary infection. Both mechanisms can explain the basic dynamics of an acute infection-exponential growth of the pathogen followed by control and clearance-and can also be represented by many different differential equation models. Consequently, traditional model comparison techniques using time series data can be ambiguous or inconclusive. We propose that varying the inoculum dose and measuring the subsequent infectious load can rule out target cell depletion by the pathogen as the main control mechanism. Infectious load can be any measure that is proportional to the number of infected cells, such as viraemia. We show that a twofold or greater change in infectious load is unlikely when target cell depletion controls infection, regardless of the model details. Analyzing previously published data from mice infected with influenza, we find the proportion of lung epithelial cells infected was 21-fold greater (95% confidence interval 14-32) in the highest dose group than in the lowest. This provides evidence in favor of an alternative to target cell depletion, such as innate immunity, in controlling influenza infections in this experimental system. Data from other experimental animal models of acute primary infection have a similar pattern.
- Published
- 2020
- Full Text
- View/download PDF
34. Broadly Protective Strategies Against Influenza Viruses: Universal Vaccines and Therapeutics.
- Author
-
Vogel OA and Manicassamy B
- Abstract
Influenza virus is a respiratory pathogen that can cause disease in humans, with symptoms ranging from mild to life-threatening. The vast majority of influenza virus infections in humans are observed during seasonal epidemics and occasional pandemics. Given the substantial public health burden associated with influenza virus infection, yearly vaccination is recommended for protection against seasonal influenza viruses. Despite vigilant surveillance for new variants and careful selection of seasonal vaccine strains, the efficacy of seasonal vaccines can vary widely from year to year. This often results in lowered protection within the population, regardless of vaccination status. In order to broaden the protection afforded by seasonal influenza vaccines, the National Institute of Allergy and Infectious Diseases (NIAID) has deemed the development of a universal influenza virus vaccine to be a priority in influenza virus vaccine research. This universal vaccine would provide protection against all influenza virus strains, eliminating the need for the yearly reformulations of seasonal influenza vaccines. In addition to universal influenza vaccine efforts, substantial progress has been made in developing novel influenza virus therapeutics that utilize broadly neutralizing antibodies to provide protection against influenza virus infection and to mitigate disease outcomes during infection. In this review, we discuss various approaches toward the goal of improving influenza virus vaccine efficacy through a universal influenza virus vaccine. We also address the novel methods of discovery and utilization of broadly neutralizing antibodies to improve influenza disease outcomes., (Copyright © 2020 Vogel and Manicassamy.)
- Published
- 2020
- Full Text
- View/download PDF
35. A Simple and Robust Approach for Evaluation of Antivirals Using a Recombinant Influenza Virus Expressing Gaussia Luciferase.
- Author
-
Li P, Cui Q, Wang L, Zhao X, Zhang Y, Manicassamy B, Yang Y, Rong L, and Du R
- Subjects
- Animals, Dogs, Genes, Reporter, HEK293 Cells, Humans, Influenza A virus genetics, Influenza A virus growth & development, Luciferases genetics, Lung virology, Madin Darby Canine Kidney Cells, Mice, Orthomyxoviridae Infections drug therapy, Orthomyxoviridae Infections virology, Treatment Outcome, Antiviral Agents isolation & purification, Antiviral Agents pharmacology, Drug Evaluation, Preclinical methods, Influenza A virus drug effects, Luciferases analysis, Staining and Labeling methods
- Abstract
Influenza A virus (IAV) causes seasonal epidemics and occasional but devastating pandemics, which are major public health concerns. Because the effectiveness of seasonal vaccines is highly variable and the currently available drugs are limited in their efficacy because of the emergence of drug resistance, there is an urgent need to develop novel antivirals. In this study, we characterized a recombinant IAV-carrying Gaussia luciferase (Gluc) gene and determined its potential as a tool for evaluating therapeutics. We demonstrated that this recombinant IAV is replication-competent in tissue culture and pathogenic in mice, although it is slightly attenuated compared to the parental virus. Luciferase expression correlated well with virus propagation both in vitro and in vivo, providing a simple measure for viral replication in tissue culture and in mouse lungs. To demonstrate the utility of this virus, ribavirin and oseltamivir phosphate were used to treat the IAV-infected cells and mice, and we observed the dose-dependent inhibition of viral replication by a luciferase assay. Moreover, the decreased luciferase expression in the infected lungs could predict the protective efficacy of antiviral interventions as early as day 2 post virus challenge. In summary, this study provides a new and quantitative approach to evaluate antivirals against IAV., Competing Interests: The authors declare no conflict of interest.
- Published
- 2018
- Full Text
- View/download PDF
36. Genome-wide CRISPR/Cas9 Screen Identifies Host Factors Essential for Influenza Virus Replication.
- Author
-
Han J, Perez JT, Chen C, Li Y, Benitez A, Kandasamy M, Lee Y, Andrade J, tenOever B, and Manicassamy B
- Subjects
- A549 Cells, Gene Library, Genome, Human, Humans, Influenza A Virus, H5N1 Subtype genetics, Lentivirus genetics, Nucleotide Transport Proteins genetics, Nucleotide Transport Proteins metabolism, Virus Internalization, Virus Replication, CRISPR-Cas Systems genetics, Gene Knockout Techniques methods, Influenza A Virus, H5N1 Subtype physiology
- Abstract
The emergence of influenza A viruses (IAVs) from zoonotic reservoirs poses a great threat to human health. As seasonal vaccines are ineffective against zoonotic strains, and newly transmitted viruses can quickly acquire drug resistance, there remains a need for host-directed therapeutics against IAVs. Here, we performed a genome-scale CRISPR/Cas9 knockout screen in human lung epithelial cells with a human isolate of an avian H5N1 strain. Several genes involved in sialic acid biosynthesis and related glycosylation pathways were highly enriched post-H5N1 selection, including SLC35A1, a sialic acid transporter essential for IAV receptor expression and thus viral entry. Importantly, we have identified capicua (CIC) as a negative regulator of cell-intrinsic immunity, as loss of CIC resulted in heightened antiviral responses and restricted replication of multiple viruses. Therefore, our study demonstrates that the CRISPR/Cas9 system can be utilized for the discovery of host factors critical for the replication of intracellular pathogens., (Copyright © 2018 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
37. Broad Spectrum Inhibitor of Influenza A and B Viruses Targeting the Viral Nucleoprotein.
- Author
-
White KM, Abreu P Jr, Wang H, De Jesus PD, Manicassamy B, García-Sastre A, Chanda SK, DeVita RJ, and Shaw ML
- Subjects
- Animals, Cell Line, Drug Synergism, Gene Expression Regulation, Viral drug effects, Humans, Influenza A virus physiology, Betainfluenzavirus physiology, Microbial Sensitivity Tests, Molecular Structure, Oseltamivir pharmacology, Protein Aggregates drug effects, Protein Binding, Virus Internalization drug effects, Virus Replication drug effects, Antiviral Agents pharmacology, Influenza A virus drug effects, Betainfluenzavirus drug effects, Viral Core Proteins antagonists & inhibitors
- Abstract
S119 was a top hit from an ultrahigh throughput screen performed to identify novel inhibitors of influenza virus replication. It showed a potent antiviral effect (50% inhibitory concentration, IC
50 = 20 nM) and no detectable cytotoxicity (50% cytotoxic concentration, CC50 > 500 μM) to yield a selectivity index greater than 25 000. Upon investigation, we found that S119 selected for resistant viruses carrying mutations in the viral nucleoprotein (NP). These resistance mutations highlight a likely S119 binding site overlapping with but not identical to that found for the compound nucleozin. Mechanism of action studies revealed that S119 affects both the oligomerization state and cellular localization of the NP protein which has an impact on viral transcription, replication, and protein expression. Through a hit-to-lead structure-activity relationship (SAR) study, we found an analog of S119, named S119-8, which had increased breadth of inhibition against influenza A and B viruses accompanied by only a small loss in potency. Finally, in vitro viral inhibition assays showed a synergistic relationship between S119-8 and oseltamivir when they were combined, indicating the potential for future drug cocktails.- Published
- 2018
- Full Text
- View/download PDF
38. Influenza A virus infection impacts systemic microbiota dynamics and causes quantitative enteric dysbiosis.
- Author
-
Yildiz S, Mazel-Sanchez B, Kandasamy M, Manicassamy B, and Schmolke M
- Subjects
- Animals, Bacteria genetics, Bacteria isolation & purification, DNA, Bacterial genetics, DNA, Ribosomal genetics, Disease Models, Animal, Dysbiosis microbiology, Female, Gastrointestinal Microbiome, Influenza A virus pathogenicity, Mice, Sequence Analysis, DNA, Bacteria classification, Orthomyxoviridae Infections microbiology, Paneth Cells microbiology, RNA, Ribosomal, 16S genetics
- Abstract
Background: Microbiota integrity is essential for a growing number of physiological processes. Consequently, disruption of microbiota homeostasis correlates with a variety of pathological states. Importantly, commensal microbiota provide a shield against invading bacterial pathogens, probably by direct competition. The impact of viral infections on host microbiota composition and dynamics is poorly understood. Influenza A viruses (IAV) are common respiratory pathogens causing acute infections. Here, we show dynamic changes in respiratory and intestinal microbiota over the course of a sublethal IAV infection in a mouse model., Results: Using a combination of 16S rRNA gene-specific next generation sequencing and qPCR as well as culturing of bacterial organ content, we found body site-specific and transient microbiota responses. In the lower respiratory tract, we observed only minor qualitative changes in microbiota composition. No quantitative impact on bacterial colonization after IAV infection was detectable, despite a robust antimicrobial host response and increased sensitivity to bacterial super infection. In contrast, in the intestine, IAV induced robust depletion of bacterial content, disruption of mucus layer integrity, and higher levels of antimicrobial peptides in Paneth cells. As a functional consequence of IAV-mediated microbiota depletion, we demonstrated that the small intestine is rendered more susceptible to bacterial pathogen invasion, in a Salmonella typhimurium super infection model., Conclusion: We show for the first time the consequences of IAV infection for lower respiratory tract and intestinal microbiobiota in a qualitative and quantitative fashion. The discrepancy of relative 16S rRNA gene next-generation sequencing (NGS) and normalized 16S rRNA gene-specific qPCR stresses the importance of combining qualitative and quantitative approaches to correctly analyze composition of organ associated microbial communities. The transiently induced dysbiosis underlines the overall stability of microbial communities to effects of acute infection. However, during a short-time window, specific ecological niches might lose their microbiota shield and remain vulnerable to bacterial invasion.
- Published
- 2018
- Full Text
- View/download PDF
39. Development of Potential Small Molecule Therapeutics for Treatment of Ebola Virus Disease.
- Author
-
Schafer A, Cheng H, Lee C, Du R, Han J, Perez J, Peet N, Manicassamy B, and Rong L
- Subjects
- Animals, Hemorrhagic Fever, Ebola etiology, Humans, Transcription, Genetic drug effects, Virus Internalization drug effects, Virus Release drug effects, Virus Replication drug effects, Antiviral Agents therapeutic use, Drug Development methods, Ebolavirus drug effects, Ebolavirus genetics, Ebolavirus growth & development, Hemorrhagic Fever, Ebola drug therapy
- Abstract
Ebola virus has caused 26 outbreaks in 10 different countries since its identification in 1976, making it one of the deadliest emerging viral pathogens. The most recent outbreak in West Africa from 2014-16 was the deadliest yet and culminated in 11,310 deaths out of 28,616 confirmed cases. Currently, there are no FDA-approved therapeutics or vaccines to treat Ebola virus infections. The slow development of effective vaccines combined with the severity of past outbreaks emphasizes the need to accelerate research into understanding the virus lifecycle and the development of therapeutics for post exposure treatment. Here we present a summary of the major findings on the Ebola virus replication cycle and the therapeutic approaches explored to treat this devastating disease. The major focus of this review is on small molecule inhibitors., (Copyright© Bentham Science Publishers; For any queries, please email at epub@benthamscience.net.)
- Published
- 2018
- Full Text
- View/download PDF
40. Influenza Virus: Small Molecule Therapeutics and Mechanisms of Antiviral Resistance.
- Author
-
Han J, Perez J, Schafer A, Cheng H, Peet N, Rong L, and Manicassamy B
- Subjects
- Animals, Child, Enzyme Inhibitors therapeutic use, Humans, Viral Proteins antagonists & inhibitors, Antiviral Agents therapeutic use, Drug Resistance, Viral drug effects, Influenza, Human drug therapy, Orthomyxoviridae drug effects
- Abstract
Background: Influenza viruses cause severe upper respiratory illness in children and the elderly during seasonal epidemics. Influenza viruses from zoonotic reservoirs can also cause pandemics with significant loss of life in all age groups. Although vaccination is one of the most effective methods to protect against seasonal epidemics, seasonal vaccines vary in efficacy, can be ineffective in the elderly population, and do not provide protection against novel strains. Small molecule therapeutics are a critical part of our antiviral strategies to control influenza virus epidemics and pandemics as well as to ameliorate disease in elderly and immunocompromised individuals., Objective: This review aims to summarize the existing antiviral strategies for combating influenza viruses, the mechanisms of antiviral resistance for available drugs, and novel therapeutics currently in development., Methods: We systematically evaluated and synthesized the published scientific literature for mechanistic detail into therapeutic strategies against influenza viruses., Results: Current IAV strains have developed resistance to neuraminidase inhibitors and nearly complete resistance to M2 ion channel inhibitors, exacerbated by sub-therapeutic dosing used for treatment and chemoprophylaxis. New tactics include novel therapeutics targeting host components and combination therapy, which show potential for fighting influenza virus disease while minimizing viral resistance., Conclusion: Antiviral drugs are crucial for controlling influenza virus disease burden, but their efficacy is limited by human misuse and the capacity of influenza viruses to circumvent antiviral barriers. To relieve the public health hardship of influenza virus, emerging therapies must be selected for their capacity to impede not only influenza virus disease, but also the development of antiviral resistance., (Copyright© Bentham Science Publishers; For any queries, please email at epub@benthamscience.net.)
- Published
- 2018
- Full Text
- View/download PDF
41. Igβ ubiquitination activates PI3K signals required for endosomal sorting.
- Author
-
Veselits M, Tanaka A, Chen Y, Hamel K, Mandal M, Kandasamy M, Manicassamy B, O'Neill SK, Wilson P, Sciammas R, and Clark MR
- Subjects
- Animals, B-Lymphocytes metabolism, Endocytosis, Histocompatibility Antigens Class II metabolism, Immunity, Humoral, Male, Mice, Inbred C57BL, Phosphatidylinositol Phosphates metabolism, Receptors, Antigen, B-Cell metabolism, Toll-Like Receptors metabolism, Ubiquitin metabolism, CD79 Antigens metabolism, Endosomes metabolism, Phosphatidylinositol 3-Kinase metabolism, Signal Transduction, Ubiquitination
- Abstract
A wealth of in vitro data has demonstrated a central role for receptor ubiquitination in endocytic sorting. However, how receptor ubiquitination functions in vivo is poorly understood. Herein, we report that ablation of B cell antigen receptor ubiquitination in vivo uncouples the receptor from CD19 phosphorylation and phosphatidylinositol 3-kinase (PI3K) signals. These signals are necessary and sufficient for accumulating phosphatidylinositol (3,4,5)-trisphosphate (PIP
3 ) on B cell receptor-containing early endosomes and proper sorting into the MHC class II antigen-presenting compartment (MIIC). Surprisingly, MIIC targeting is dispensable for T cell-dependent immunity. Rather, it is critical for activating endosomal toll-like receptors and antiviral humoral immunity. These findings demonstrate a novel mechanism of receptor endosomal signaling required for specific peripheral immune responses., (© 2017 Veselits et al.)- Published
- 2017
- Full Text
- View/download PDF
42. Endothelial cell tropism is a determinant of H5N1 pathogenesis in mammalian species.
- Author
-
Tundup S, Kandasamy M, Perez JT, Mena N, Steel J, Nagy T, Albrecht RA, and Manicassamy B
- Subjects
- Animals, Blotting, Western, Disease Models, Animal, Female, Ferrets, Flow Cytometry, HEK293 Cells, Humans, Male, Mice, Mice, Inbred C57BL, Real-Time Polymerase Chain Reaction, Virulence, Endothelial Cells virology, Influenza A Virus, H5N1 Subtype pathogenicity, Influenza A Virus, H5N1 Subtype physiology, Orthomyxoviridae Infections pathology, Viral Tropism physiology
- Abstract
The cellular and molecular mechanisms underpinning the unusually high virulence of highly pathogenic avian influenza H5N1 viruses in mammalian species remains unknown. Here, we investigated if the cell tropism of H5N1 virus is a determinant of enhanced virulence in mammalian species. We engineered H5N1 viruses with restricted cell tropism through the exploitation of cell type-specific microRNA expression by incorporating microRNA target sites into the viral genome. Restriction of H5N1 replication in endothelial cells via miR-126 ameliorated disease symptoms, prevented systemic viral spread and limited mortality, despite showing similar levels of peak viral replication in the lungs as compared to control virus-infected mice. Similarly, restriction of H5N1 replication in endothelial cells resulted in ameliorated disease symptoms and decreased viral spread in ferrets. Our studies demonstrate that H5N1 infection of endothelial cells results in excessive production of cytokines and reduces endothelial barrier integrity in the lungs, which culminates in vascular leakage and viral pneumonia. Importantly, our studies suggest a need for a combinational therapy that targets viral components, suppresses host immune responses, and improves endothelial barrier integrity for the treatment of highly pathogenic H5N1 virus infections.
- Published
- 2017
- Full Text
- View/download PDF
43. Stabilization and Improvement of a Promising Influenza Antiviral: Making a PAIN PAINless.
- Author
-
Antanasijevic A, Hafeman NJ, Tundup S, Kingsley C, Mishra RK, Rong L, Manicassamy B, Wardrop D, and Caffrey M
- Subjects
- Antioxidants chemistry, Antioxidants pharmacology, Antiviral Agents pharmacology, Hemagglutinin Glycoproteins, Influenza Virus genetics, Hemagglutinin Glycoproteins, Influenza Virus metabolism, Humans, Hydroquinones pharmacology, Influenza A virus drug effects, Influenza A virus genetics, Influenza A virus metabolism, Molecular Dynamics Simulation, Structure-Activity Relationship, Antiviral Agents chemistry, Hydroquinones chemistry, Influenza, Human virology
- Abstract
The viral envelope protein hemagglutinin (HA) plays a critical role in influenza entry and thus is an attractive target for novel therapeutics. The small molecule tert-butylhydroquinone (TBHQ) has previously been shown to bind to HA and inhibit HA-mediated entry with low micromolar potency. However, enthusiasm for the use of TBHQ has diminished due to the compound's antioxidant properties. In this work we show that the antioxidant properties of TBHQ are not responsible for the inhibition of HA-mediated entry. In addition, we have performed a structure-activity relationship (SAR) analysis of TBHQ derivatives. We find that the most promising compound, 3-tert-butyl-4-methoxyphenol, exhibits enhanced potency (IC
50 = 0.6 μM), decreased toxicity (CC50 = 340 μM), and increased stability (t1/2 > 48 h). Finally, we have characterized the binding properties of 3-tert-butyl-4-methoxyphenol using NMR and molecular dynamics to guide future efforts for chemical optimization.- Published
- 2016
- Full Text
- View/download PDF
44. RIG-I Signaling Is Critical for Efficient Polyfunctional T Cell Responses during Influenza Virus Infection.
- Author
-
Kandasamy M, Suryawanshi A, Tundup S, Perez JT, Schmolke M, Manicassamy S, and Manicassamy B
- Subjects
- Adaptor Proteins, Signal Transducing immunology, Animals, Coculture Techniques, Dendritic Cells immunology, Disease Models, Animal, Flow Cytometry, Influenza A virus immunology, Interferon Type I biosynthesis, Interferon Type I immunology, Lymphocyte Activation immunology, Membrane Proteins metabolism, Mice, Mice, Knockout, Nerve Tissue Proteins metabolism, Polymerase Chain Reaction, Receptors, Cell Surface, T-Lymphocytes metabolism, Membrane Proteins immunology, Nerve Tissue Proteins immunology, Orthomyxoviridae Infections immunology, Signal Transduction immunology, T-Lymphocytes immunology
- Abstract
Retinoic acid inducible gene-I (RIG-I) is an innate RNA sensor that recognizes the influenza A virus (IAV) RNA genome and activates antiviral host responses. Here, we demonstrate that RIG-I signaling plays a crucial role in restricting IAV tropism and regulating host immune responses. Mice deficient in the RIG-I-MAVS pathway show defects in migratory dendritic cell (DC) activation, viral antigen presentation, and priming of CD8+ and CD4+ T cell responses during IAV infection. These defects result in decreased frequency of polyfunctional effector T cells and lowered protection against heterologous IAV challenge. In addition, our data show that RIG-I activation is essential for protecting epithelial cells and hematopoietic cells from IAV infection. These diverse effects of RIG-I signaling are likely imparted by the actions of type I interferon (IFN), as addition of exogenous type I IFN is sufficient to overcome the defects in antigen presentation by RIG-I deficient BMDC. Moreover, the in vivo T cell defects in RIG-I deficient mice can be overcome by the activation of MDA5 -MAVS via poly I:C treatment. Taken together, these findings demonstrate that RIG-I signaling through MAVS is critical for determining the quality of polyfunctional T cell responses against IAV and for providing protection against subsequent infection from heterologous or novel pandemic IAV strains.
- Published
- 2016
- Full Text
- View/download PDF
45. Homeostatic PPARα Signaling Limits Inflammatory Responses to Commensal Microbiota in the Intestine.
- Author
-
Manoharan I, Suryawanshi A, Hong Y, Ranganathan P, Shanmugam A, Ahmad S, Swafford D, Manicassamy B, Ramesh G, Koni PA, Thangaraju M, and Manicassamy S
- Subjects
- Animals, Cells, Cultured, Homeodomain Proteins immunology, Inflammation immunology, Mice, Mice, Inbred C57BL, Mice, Knockout, PPAR alpha deficiency, Gastrointestinal Microbiome immunology, Homeostasis, Inflammation prevention & control, PPAR alpha metabolism, Signal Transduction
- Abstract
Dietary lipids and their metabolites activate members of the peroxisome proliferative-activated receptor (PPAR) family of transcription factors and are critical for colonic health. The PPARα isoform plays a vital role in regulating inflammation in various disease settings, but its role in intestinal inflammation, commensal homeostasis, and mucosal immunity in the gut are unclear. In this study, we demonstrate that the PPARα pathway in innate immune cells orchestrates gut mucosal immunity and commensal homeostasis by regulating the expression of IL-22 and the antimicrobial peptides RegIIIβ, RegIIIγ, and calprotectin. Additionally, the PPARα pathway is critical for imparting regulatory phenotype in intestinal macrophages. PPARα deficiency in mice led to commensal dysbiosis in the gut, resulting in a microbiota-dependent increase in the expression of inflammatory cytokines and enhanced susceptibility to intestinal inflammation. Pharmacological activation of this pathway decreased the expression of inflammatory cytokines and ameliorated colonic inflammation. Taken together, these findings identify a new important innate immune function for the PPARα signaling pathway in regulating intestinal inflammation, mucosal immunity, and commensal homeostasis. Thus, the manipulation of the PPARα pathway could provide novel opportunities for enhancing mucosal immunity and treating intestinal inflammation., (Copyright © 2016 by The American Association of Immunologists, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
46. La Piedad Michoacán Mexico Virus V protein antagonizes type I interferon response by binding STAT2 protein and preventing STATs nuclear translocation.
- Author
-
Pisanelli G, Laurent-Rolle M, Manicassamy B, Belicha-Villanueva A, Morrison J, Lozano-Dubernard B, Castro-Peralta F, Iovane G, and García-Sastre A
- Subjects
- Animals, Cell Line, Humans, Protein Binding, Protein Transport, Swine, Interferon Type I antagonists & inhibitors, Rubulavirus immunology, STAT2 Transcription Factor metabolism, Viral Proteins metabolism
- Abstract
La Piedad Michoacán Mexico Virus (LPMV) is a member of the Rubulavirus genus within the Paramyxoviridae family. LPMV is the etiologic agent of "blue eye disease", causing a significant disease burden in swine in Mexico with long-term implications for the agricultural industry. This virus mainly affects piglets and is characterized by meningoencephalitis and respiratory distress. It also affects adult pigs, causing reduced fertility and abortions in females, and orchitis and epididymitis in males. Viruses of the Paramyxoviridae family evade the innate immune response by targeting components of the interferon (IFN) signaling pathway. The V protein, expressed by most paramyxoviruses, is a well-characterized IFN signaling antagonist. Until now, there were no reports on the role of the LPMV-V protein in inhibiting the IFN response. In this study we demonstrate that LPMV-V protein antagonizes type I but not type II IFN signaling by binding STAT2, a component of the type I IFN cascade. Our results indicate that the last 18 amino acids of LPMV-V protein are required for binding to STAT2 in human and swine cells. While LPMV-V protein does not affect the protein levels of STAT1 or STAT2, it does prevent the IFN-induced phosphorylation and nuclear translocation of STAT1 and STAT2 thereby inhibiting cellular responses to IFN α/β., (Copyright © 2015 Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
47. Deletion of LRP5 and LRP6 in dendritic cells enhances antitumor immunity.
- Author
-
Hong Y, Manoharan I, Suryawanshi A, Shanmugam A, Swafford D, Ahmad S, Chinnadurai R, Manicassamy B, He Y, Mellor AL, Thangaraju M, Munn DH, and Manicassamy S
- Abstract
The tumor microenvironment (TME) contains high levels of the Wnt family of ligands, and aberrant Wnt-signaling occurs in many tumors. Past studies have been directed toward how the Wnt signaling cascade regulates cancer development, progression and metastasis. However, its effects on host antitumor immunity remain unknown. In this report, we show that Wnts in the TME condition dendritic cells (DCs) to a regulatory state and suppress host antitumor immunity. DC-specific deletion of Wnt co-receptors low-density lipoprotein receptor-related protein 5 and 6 (LRP5/6) in mice markedly delayed tumor growth and enhanced host antitumor immunity. Mechanistically, loss of LRP5/6-mediated signaling in DCs resulted in enhanced effector T cell differentiation and decreased regulatory T cell differentiation. This was due to increased production of pro-inflammatory cytokines and decreased production of IL-10, TGF-β1 and retinoic acid (RA). Likewise, pharmacological inhibition of the Wnts' interaction with its cognate co-receptors LRP5/6 and Frizzled (Fzd) receptors had similar effects on tumor growth and effector T cell responses. Moreover, blocking Wnt-signaling in DCs resulted in enhanced capture of tumor-associated antigens and efficient cross-priming of CD8
+ T cells. Hence, blocking the Wnt pathway represents a potential therapeutic to overcome tumor-mediated immune suppression and augment antitumor immunity.- Published
- 2015
- Full Text
- View/download PDF
48. Meta- and Orthogonal Integration of Influenza "OMICs" Data Defines a Role for UBR4 in Virus Budding.
- Author
-
Tripathi S, Pohl MO, Zhou Y, Rodriguez-Frandsen A, Wang G, Stein DA, Moulton HM, DeJesus P, Che J, Mulder LC, Yángüez E, Andenmatten D, Pache L, Manicassamy B, Albrecht RA, Gonzalez MG, Nguyen Q, Brass A, Elledge S, White M, Shapira S, Hacohen N, Karlas A, Meyer TF, Shales M, Gatorano A, Johnson JR, Jang G, Johnson T, Verschueren E, Sanders D, Krogan N, Shaw M, König R, Stertz S, García-Sastre A, and Chanda SK
- Subjects
- Animals, Cell Line, Computational Biology, Flow Cytometry, Humans, Immunoprecipitation, Mice, Inbred BALB C, Microscopy, Fluorescence, Protein Binding, Protein Interaction Maps, Protein Transport, Ubiquitin-Protein Ligases, Calmodulin-Binding Proteins metabolism, Cytoskeletal Proteins metabolism, Host-Pathogen Interactions, Influenza A virus physiology, Viral Matrix Proteins metabolism, Virus Release
- Abstract
Several systems-level datasets designed to dissect host-pathogen interactions during influenza A infection have been reported. However, apparent discordance among these data has hampered their full utility toward advancing mechanistic and therapeutic knowledge. To collectively reconcile these datasets, we performed a meta-analysis of data from eight published RNAi screens and integrated these data with three protein interaction datasets, including one generated within the context of this study. Further integration of these data with global virus-host interaction analyses revealed a functionally validated biochemical landscape of the influenza-host interface, which can be queried through a simplified and customizable web portal (http://www.metascape.org/IAV). Follow-up studies revealed that the putative ubiquitin ligase UBR4 associates with the viral M2 protein and promotes apical transport of viral proteins. Taken together, the integrative analysis of influenza OMICs datasets illuminates a viral-host network of high-confidence human proteins that are essential for influenza A virus replication., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
49. Corrigendum: Pan-viral specificity of IFN-induced genes reveals new roles for cGAS in innate immunity.
- Author
-
Schoggins JW, MacDuff DA, Imanaka N, Gainey MD, Shrestha B, Eitson JL, Mar KB, Richardson RB, Ratushny AV, Litvak V, Dabelic R, Manicassamy B, Aitchison JD, Aderem A, Elliott RM, García-Sastre A, Racaniello V, Snijder EJ, Yokoyama WM, Diamond MS, Virgin HW, and Rice CM
- Published
- 2015
- Full Text
- View/download PDF
50. Canonical wnt signaling in dendritic cells regulates Th1/Th17 responses and suppresses autoimmune neuroinflammation.
- Author
-
Suryawanshi A, Manoharan I, Hong Y, Swafford D, Majumdar T, Taketo MM, Manicassamy B, Koni PA, Thangaraju M, Sun Z, Mellor AL, Munn DH, and Manicassamy S
- Subjects
- Animals, Cell Differentiation, Cytokines metabolism, Encephalomyelitis, Autoimmune, Experimental immunology, Encephalomyelitis, Autoimmune, Experimental metabolism, Encephalomyelitis, Autoimmune, Experimental pathology, Gene Deletion, Gene Knockout Techniques, Inflammation Mediators metabolism, Interleukin-10 metabolism, Low Density Lipoprotein Receptor-Related Protein-5 genetics, Low Density Lipoprotein Receptor-Related Protein-6 genetics, Male, Mice, Mice, Transgenic, Signal Transduction, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, Th1 Cells cytology, Th1 Cells metabolism, Th17 Cells cytology, Th17 Cells metabolism, Wnt3A Protein metabolism, beta Catenin metabolism, Dendritic Cells immunology, Dendritic Cells metabolism, Th1 Cells immunology, Th17 Cells immunology, Transforming Growth Factor beta metabolism, Wnt Signaling Pathway drug effects
- Abstract
Breakdown in immunological tolerance to self-Ags or uncontrolled inflammation results in autoimmune disorders. Dendritic cells (DCs) play an important role in regulating the balance between inflammatory and regulatory responses in the periphery. However, factors in the tissue microenvironment and the signaling networks critical for programming DCs to control chronic inflammation and promote tolerance are unknown. In this study, we show that wnt ligand-mediated activation of β-catenin signaling in DCs is critical for promoting tolerance and limiting neuroinflammation. DC-specific deletion of key upstream (lipoprotein receptor-related protein [LRP]5/6) or downstream (β-catenin) mediators of canonical wnt signaling in mice exacerbated experimental autoimmune encephalomyelitis pathology. Mechanistically, loss of LRP5/6-β-catenin-mediated signaling in DCs led to an increased Th1/Th17 cell differentiation but reduced regulatory T cell response. This was due to increased production of proinflammatory cytokines and decreased production of anti-inflammatory cytokines such as IL-10 and IL-27 by DCs lacking LRP5/6-β-catenin signaling. Consistent with these findings, pharmacological activation of canonical wnt/β-catenin signaling delayed experimental autoimmune encephalomyelitis onset and diminished CNS pathology. Thus, the activation of canonical wnt signaling in DCs limits effector T cell responses and represents a potential therapeutic approach to control autoimmune neuroinflammation., (Copyright © 2015 by The American Association of Immunologists, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.