1
|
Felicetti T, Sarnari C, Gaito R, Tabarrini O, Manfroni G. Recent Progress toward the Discovery of Small Molecules as Novel Anti-Respiratory Syncytial Virus Agents. J Med Chem 2024; 67:11543-11579. [PMID: 38970494 DOI: 10.1021/acs.jmedchem.4c00630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2024]
Abstract
Respiratory syncytial virus (RSV) stands as the foremost cause of infant hospitalization globally, ranking second only to malaria in terms of infant mortality. Although three vaccines have recently been approved for the prophylaxis of adults aged 60 and above, and pregnant women, there is currently no effective antiviral drug for treating RSV infections. The only preventive measure for infants at high risk of severe RSV disease is passive immunization through monoclonal antibodies. This Perspective offers an overview of the latest advancements in RSV drug discovery of small molecule antivirals, with particular focus on the promising findings from agents targeting the fusion and polymerase proteins. A comprehensive reflection on the current state of RSV research is also given, drawing inspiration from the lessons gleaned from HCV and HIV, while also considering the impact of the recent approval of the three vaccines.
Collapse
Affiliation(s)
- Tommaso Felicetti
- Department of Pharmaceutical Sciences, University of Perugia, Via Del Liceo, 1-06123, Perugia, Italy
| | - Chiara Sarnari
- Department of Pharmaceutical Sciences, University of Perugia, Via Del Liceo, 1-06123, Perugia, Italy
| | - Roberta Gaito
- Department of Pharmaceutical Sciences, University of Perugia, Via Del Liceo, 1-06123, Perugia, Italy
| | - Oriana Tabarrini
- Department of Pharmaceutical Sciences, University of Perugia, Via Del Liceo, 1-06123, Perugia, Italy
| | - Giuseppe Manfroni
- Department of Pharmaceutical Sciences, University of Perugia, Via Del Liceo, 1-06123, Perugia, Italy
| |
Collapse
|
2
|
Langedijk AC, Bont LJ. Respiratory syncytial virus infection and novel interventions. Nat Rev Microbiol 2023; 21:734-749. [PMID: 37438492 DOI: 10.1038/s41579-023-00919-w] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2023] [Indexed: 07/14/2023]
Abstract
The large global burden of respiratory syncytial virus (RSV) respiratory tract infections in young children and older adults has gained increased recognition in recent years. Recent discoveries regarding the neutralization-specific viral epitopes of the pre-fusion RSV glycoprotein have led to a shift from empirical to structure-based design of RSV therapeutics, and controlled human infection model studies have provided early-stage proof of concept for novel RSV monoclonal antibodies, vaccines and antiviral drugs. The world's first vaccines and first monoclonal antibody to prevent RSV among older adults and all infants, respectively, have recently been approved. Large-scale introduction of RSV prophylactics emphasizes the need for active surveillance to understand the global impact of these interventions over time and to timely identify viral mutants that are able to escape novel prophylactics. In this Review, we provide an overview of RSV interventions in clinical development, highlighting global disease burden, seasonality, pathogenesis, and host and viral factors related to RSV immunity.
Collapse
Affiliation(s)
- Annefleur C Langedijk
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Louis J Bont
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands.
- ReSViNET Foundation, Zeist, the Netherlands.
| |
Collapse
|
3
|
Pangesti KNA, Ansari HR, Bayoumi A, Kesson AM, Hill-Cawthorne GA, Abd El Ghany M. Genomic characterization of respiratory syncytial virus genotypes circulating in the paediatric population of Sydney, NSW, Australia. Microb Genom 2023; 9:001095. [PMID: 37656160 PMCID: PMC10569731 DOI: 10.1099/mgen.0.001095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 08/03/2023] [Indexed: 09/02/2023] Open
Abstract
Respiratory syncytial virus (RSV), or human orthopneumovirus, is a major cause of acute lower respiratory infection (ALRI), particularly in young children, causing significant morbidity and mortality. We used pathogen genomics to characterize the population structure and genetic signatures of RSV isolates circulating in children in New South Wales between 2016 and 2018 and to understand the evolutionary dynamics of these strains in the context of publicly available RSV genomes from the region and globally. Whole-genome phylogenetic analysis demonstrated the co-circulation of a few major RSV clades in the paediatric population from Sydney. The whole-genome-based genotypes A23 (RSV-A ON1-like genotype) and B6 (RSV-B BA9-like genotype) were the predominant RSV-A and RSV-B genotypes circulating during the study period, respectively. These genotypes were characterized with high levels of diversity of predicted N- and O-linked glycosylation patterns in both the G and F glycoproteins. Interestingly, a novel 72-nucleotide triplication in the sequence that corresponds to the C-terminal region of the G gene was identified in four of the A23 genotype sequenced in this study. Consistently, the population dynamics analysis demonstrated a continuous increase in the effective population size of A23 and B6 genotypes globally. Further investigations including functional mapping of mutations and identifying the impact of sequence changes on virus fitness are highly required. This study highlights the potential impact of an integrated approach that uses WG-based phylogeny and studying selective pressure events in understanding the emergence and dissemination of RSV genotypes.
Collapse
Affiliation(s)
- Krisna N. A. Pangesti
- School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Hifzur R. Ansari
- King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Ali Bayoumi
- The Westmead Institute for Medical Research, The University of Sydney, Sydney, Australia
| | - Alison M. Kesson
- Department of Infectious Diseases and Microbiology, The Children’s Hospital at Westmead, Sydney, Australia
- Sydney Infectious Diseases Institute, The University of Sydney, Sydney, Australia
- Discipline of Child and Adolescent Health, The University of Sydney, Sydney, Australia
| | - Grant A. Hill-Cawthorne
- School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Moataz Abd El Ghany
- School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- The Westmead Institute for Medical Research, The University of Sydney, Sydney, Australia
- Sydney Infectious Diseases Institute, The University of Sydney, Sydney, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| |
Collapse
|
4
|
Córdova-Dávalos LE, Hernández-Mercado A, Barrón-García CB, Rojas-Martínez A, Jiménez M, Salinas E, Cervantes-García D. Impact of genetic polymorphisms related to innate immune response on respiratory syncytial virus infection in children. Virus Genes 2022; 58:501-514. [PMID: 36085536 PMCID: PMC9462631 DOI: 10.1007/s11262-022-01932-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 08/30/2022] [Indexed: 11/24/2022]
Abstract
Respiratory syncytial virus (RSV) causes lower respiratory tract infections and bronchiolitis, mainly affecting children under 2 years of age and immunocompromised patients. Currently, there are no available vaccines or efficient pharmacological treatments against RSV. In recent years, tremendous efforts have been directed to understand the pathological mechanisms of the disease and generate a vaccine against RSV. Although RSV is highly infectious, not all the patients who get infected develop bronchiolitis and severe disease. Through various sequencing studies, single nucleotide polymorphisms (SNPs) have been discovered in diverse receptors, cytokines, and transcriptional regulators with crucial role in the activation of the innate immune response, which is implicated in the susceptibility to develop or protect from severe forms of the infection. In this review, we highlighted how variations in the key genes affect the development of innate immune response against RSV. This data would provide crucial information about the mechanisms of viral infection, and in the future, could help in generation of new strategies for vaccine development or generation of the pharmacological treatments.
Collapse
Affiliation(s)
- Laura Elena Córdova-Dávalos
- Laboratorio de Inmunología, Departamento de Microbiología, Universidad Autónoma de Aguascalientes, 20100, Aguascalientes, México
| | - Alicia Hernández-Mercado
- Laboratorio de Inmunología, Departamento de Microbiología, Universidad Autónoma de Aguascalientes, 20100, Aguascalientes, México
| | - Claudia Berenice Barrón-García
- Laboratorio de Inmunología, Departamento de Microbiología, Universidad Autónoma de Aguascalientes, 20100, Aguascalientes, México
| | - Augusto Rojas-Martínez
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Av. Morones Prieto 3000 Pte, Los Doctores, 64710, Monterrey, Nuevo León, México
| | - Mariela Jiménez
- Laboratorio de Inmunología, Departamento de Microbiología, Universidad Autónoma de Aguascalientes, 20100, Aguascalientes, México
| | - Eva Salinas
- Laboratorio de Inmunología, Departamento de Microbiología, Universidad Autónoma de Aguascalientes, 20100, Aguascalientes, México.
| | - Daniel Cervantes-García
- Laboratorio de Inmunología, Departamento de Microbiología, Universidad Autónoma de Aguascalientes, 20100, Aguascalientes, México. .,Consejo Nacional de Ciencia y Tecnología, 03940, Ciudad de México, México.
| |
Collapse
|
5
|
Virus Hijacks Host Proteins and Machinery for Assembly and Budding, with HIV-1 as an Example. Viruses 2022; 14:v14071528. [PMID: 35891508 PMCID: PMC9318756 DOI: 10.3390/v14071528] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 07/02/2022] [Accepted: 07/06/2022] [Indexed: 11/17/2022] Open
Abstract
Viral assembly and budding are the final steps and key determinants of the virus life cycle and are regulated by virus–host interaction. Several viruses are known to use their late assembly (L) domains to hijack host machinery and cellular adaptors to be used for the requirement of virus replication. The L domains are highly conserved short sequences whose mutation or deletion may lead to the accumulation of immature virions at the plasma membrane. The L domains were firstly identified within retroviral Gag polyprotein and later detected in structural proteins of many other enveloped RNA viruses. Here, we used HIV-1 as an example to describe how the HIV-1 virus hijacks ESCRT membrane fission machinery to facilitate virion assembly and release. We also introduce galectin-3, a chimera type of the galectin family that is up-regulated by HIV-1 during infection and further used to promote HIV-1 assembly and budding via the stabilization of Alix–Gag interaction. It is worth further dissecting the details and finetuning the regulatory mechanism, as well as identifying novel candidates involved in this final step of replication cycle.
Collapse
|
6
|
de Souza Cardoso R, Viana RMM, Vitti BC, Coelho ACL, de Jesus BLS, de Paula Souza J, Pontelli MC, Murakami T, Ventura AM, Ono A, Arruda E. Human Respiratory Syncytial Virus Infection in a Human T Cell Line Is Hampered at Multiple Steps. Viruses 2021; 13:v13020231. [PMID: 33540662 PMCID: PMC7913106 DOI: 10.3390/v13020231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 01/28/2021] [Accepted: 01/28/2021] [Indexed: 11/19/2022] Open
Abstract
Human respiratory syncytial virus (HRSV) is the most frequent cause of severe respiratory disease in children. The main targets of HRSV infection are epithelial cells of the respiratory tract, and the great majority of the studies regarding HRSV infection are done in respiratory cells. Recently, the interest on respiratory virus infection of lymphoid cells has been growing, but details of the interaction of HRSV with lymphoid cells remain unknown. Therefore, this study was done to assess the relationship of HRSV with A3.01 cells, a human CD4+ T cell line. Using flow cytometry and fluorescent focus assay, we found that A3.01 cells are susceptible but virtually not permissive to HRSV infection. Dequenching experiments revealed that the fusion process of HRSV in A3.01 cells was nearly abolished in comparison to HEp-2 cells, an epithelial cell lineage. Quantification of viral RNA by RT-qPCR showed that the replication of HRSV in A3.01 cells was considerably reduced. Western blot and quantitative flow cytometry analyses demonstrated that the production of HRSV proteins in A3.01 was significantly lower than in HEp-2 cells. Additionally, using fluorescence in situ hybridization, we found that the inclusion body-associated granules (IBAGs) were almost absent in HRSV inclusion bodies in A3.01 cells. We also assessed the intracellular trafficking of HRSV proteins and found that HRSV proteins colocalized partially with the secretory pathway in A3.01 cells, but these HRSV proteins and viral filaments were present only scarcely at the plasma membrane. HRSV infection of A3.01 CD4+ T cells is virtually unproductive as compared to HEp-2 cells, as a result of defects at several steps of the viral cycle: Fusion, genome replication, formation of inclusion bodies, recruitment of cellular proteins, virus assembly, and budding.
Collapse
Affiliation(s)
- Ricardo de Souza Cardoso
- Department of Cell and Molecular Biology, School of Medicine of Ribeirao Preto, University of Sao Paulo, São Paulo 14049-900, Brazil; (R.d.S.C.); (R.M.M.V.); (B.C.V.); (A.C.L.C.); (B.L.S.d.J.); (J.d.P.S.); (M.C.P.)
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (T.M.); (A.O.)
| | - Rosa Maria Mendes Viana
- Department of Cell and Molecular Biology, School of Medicine of Ribeirao Preto, University of Sao Paulo, São Paulo 14049-900, Brazil; (R.d.S.C.); (R.M.M.V.); (B.C.V.); (A.C.L.C.); (B.L.S.d.J.); (J.d.P.S.); (M.C.P.)
| | - Brenda Cristina Vitti
- Department of Cell and Molecular Biology, School of Medicine of Ribeirao Preto, University of Sao Paulo, São Paulo 14049-900, Brazil; (R.d.S.C.); (R.M.M.V.); (B.C.V.); (A.C.L.C.); (B.L.S.d.J.); (J.d.P.S.); (M.C.P.)
| | - Ana Carolina Lunardello Coelho
- Department of Cell and Molecular Biology, School of Medicine of Ribeirao Preto, University of Sao Paulo, São Paulo 14049-900, Brazil; (R.d.S.C.); (R.M.M.V.); (B.C.V.); (A.C.L.C.); (B.L.S.d.J.); (J.d.P.S.); (M.C.P.)
| | - Bruna Laís Santos de Jesus
- Department of Cell and Molecular Biology, School of Medicine of Ribeirao Preto, University of Sao Paulo, São Paulo 14049-900, Brazil; (R.d.S.C.); (R.M.M.V.); (B.C.V.); (A.C.L.C.); (B.L.S.d.J.); (J.d.P.S.); (M.C.P.)
| | - Juliano de Paula Souza
- Department of Cell and Molecular Biology, School of Medicine of Ribeirao Preto, University of Sao Paulo, São Paulo 14049-900, Brazil; (R.d.S.C.); (R.M.M.V.); (B.C.V.); (A.C.L.C.); (B.L.S.d.J.); (J.d.P.S.); (M.C.P.)
| | - Marjorie Cornejo Pontelli
- Department of Cell and Molecular Biology, School of Medicine of Ribeirao Preto, University of Sao Paulo, São Paulo 14049-900, Brazil; (R.d.S.C.); (R.M.M.V.); (B.C.V.); (A.C.L.C.); (B.L.S.d.J.); (J.d.P.S.); (M.C.P.)
| | - Tomoyuki Murakami
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (T.M.); (A.O.)
| | - Armando Morais Ventura
- Department of Microbiology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil;
| | - Akira Ono
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (T.M.); (A.O.)
| | - Eurico Arruda
- Department of Cell and Molecular Biology, School of Medicine of Ribeirao Preto, University of Sao Paulo, São Paulo 14049-900, Brazil; (R.d.S.C.); (R.M.M.V.); (B.C.V.); (A.C.L.C.); (B.L.S.d.J.); (J.d.P.S.); (M.C.P.)
- Correspondence:
| |
Collapse
|
7
|
Hu M, Bogoyevitch MA, Jans DA. Impact of Respiratory Syncytial Virus Infection on Host Functions: Implications for Antiviral Strategies. Physiol Rev 2020; 100:1527-1594. [PMID: 32216549 DOI: 10.1152/physrev.00030.2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Respiratory syncytial virus (RSV) is one of the leading causes of viral respiratory tract infection in infants, the elderly, and the immunocompromised worldwide, causing more deaths each year than influenza. Years of research into RSV since its discovery over 60 yr ago have elucidated detailed mechanisms of the host-pathogen interface. RSV infection elicits widespread transcriptomic and proteomic changes, which both mediate the host innate and adaptive immune responses to infection, and reflect RSV's ability to circumvent the host stress responses, including stress granule formation, endoplasmic reticulum stress, oxidative stress, and programmed cell death. The combination of these events can severely impact on human lungs, resulting in airway remodeling and pathophysiology. The RSV membrane envelope glycoproteins (fusion F and attachment G), matrix (M) and nonstructural (NS) 1 and 2 proteins play key roles in modulating host cell functions to promote the infectious cycle. This review presents a comprehensive overview of how RSV impacts the host response to infection and how detailed knowledge of the mechanisms thereof can inform the development of new approaches to develop RSV vaccines and therapeutics.
Collapse
Affiliation(s)
- MengJie Hu
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria, Australia; and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| | - Marie A Bogoyevitch
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria, Australia; and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| | - David A Jans
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria, Australia; and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
8
|
Robinson M, Schor S, Barouch-Bentov R, Einav S. Viral journeys on the intracellular highways. Cell Mol Life Sci 2018; 75:3693-3714. [PMID: 30043139 PMCID: PMC6151136 DOI: 10.1007/s00018-018-2882-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/01/2018] [Accepted: 07/19/2018] [Indexed: 12/24/2022]
Abstract
Viruses are obligate intracellular pathogens that are dependent on cellular machineries for their replication. Recent technological breakthroughs have facilitated reliable identification of host factors required for viral infections and better characterization of the virus-host interplay. While these studies have revealed cellular machineries that are uniquely required by individual viruses, accumulating data also indicate the presence of broadly required mechanisms. Among these overlapping cellular functions are components of intracellular membrane trafficking pathways. Here, we review recent discoveries focused on how viruses exploit intracellular membrane trafficking pathways to promote various stages of their life cycle, with an emphasis on cellular factors that are usurped by a broad range of viruses. We describe broadly required components of the endocytic and secretory pathways, the Endosomal Sorting Complexes Required for Transport pathway, and the autophagy pathway. Identification of such overlapping host functions offers new opportunities to develop broad-spectrum host-targeted antiviral strategies.
Collapse
Affiliation(s)
- Makeda Robinson
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Lane Building, Rm L127, Stanford, CA, 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Stanford Schor
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Lane Building, Rm L127, Stanford, CA, 94305, USA
| | - Rina Barouch-Bentov
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Lane Building, Rm L127, Stanford, CA, 94305, USA
| | - Shirit Einav
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Lane Building, Rm L127, Stanford, CA, 94305, USA.
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
9
|
An overview of process intensification and thermo stabilization for upscaling of Peste des petits ruminants vaccines in view of global control and eradication. Virusdisease 2018; 29:285-296. [PMID: 30159362 DOI: 10.1007/s13337-018-0455-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 05/03/2018] [Indexed: 12/22/2022] Open
Abstract
Peste des petits ruminants (PPR) has been recognized as a globally distributed disease affecting the small ruminant population. The disease results in severe economic losses mainly to small land holders and low input farming systems. The control of PPR is mainly achieved through vaccination with available live attenuated vaccines. The thermo labile nature of PPR virus poses a major constraint in production of quality vaccines which often results in vaccine failures. The lack of quality vaccine production jeopardize the wide vaccination coverage especially in countries with poor infrastructure due to which PPR persists endemically. The vaccine production system may require augmentation to attain consistent and quality vaccines through efforts of process intensification integrated with suitable stabilizer formulations with appropriate freeze drying cycles for improved thermo tolerance. Manufacturing of live attenuated PPR vaccines during batch cultures might introduce defective interfering particles (DIPs) as a result of high multiplicity of infection (MOI) of inoculums, which has a huge impact on virus dynamics and yield. Accumulation of DIPs adversely affects the quality of the manufactured vaccines which can be avoided through use of appropriate MOI of virus inoculums and quality control of working seed viruses. Therefore, adherence to critical manufacturing standard operating procedures in vaccine production and ongoing efforts on development of thermo tolerant vaccine will help a long way in PPR control and eradication programme globally. The present review focuses on the way forward to achieve the objectives of quality vaccine production and easy upscaling to help the global PPR control and eradication by mass vaccination as an important tool.
Collapse
|
10
|
RSV glycoprotein and genomic RNA dynamics reveal filament assembly prior to the plasma membrane. Nat Commun 2017; 8:667. [PMID: 28939853 PMCID: PMC5610308 DOI: 10.1038/s41467-017-00732-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 07/20/2017] [Indexed: 11/25/2022] Open
Abstract
The human respiratory syncytial virus G protein plays an important role in the entry and assembly of filamentous virions. Here, we report the use of fluorescently labeled soybean agglutinin to selectively label the respiratory syncytial virus G protein in living cells without disrupting respiratory syncytial virus infectivity or filament formation and allowing for interrogations of respiratory syncytial virus virion assembly. Using this approach, we discovered that plasma membrane-bound respiratory syncytial virus G rapidly recycles from the membrane via clathrin-mediated endocytosis. This event is then followed by the dynamic formation of filamentous and branched respiratory syncytial virus particles, and assembly with genomic ribonucleoproteins and caveolae-associated vesicles prior to re-insertion into the plasma membrane. We demonstrate that these processes are halted by the disruption of microtubules and inhibition of molecular motors. Collectively, our results show that for respiratory syncytial virus assembly, viral filaments are produced and loaded with genomic RNA prior to insertion into the plasma membrane. Assembly of filamentous RSV particles is incompletely understood due to a lack of techniques suitable for live-cell imaging. Here Vanover et al. use labeled soybean agglutinin to selectively label RSV G protein and show how filamentous RSV assembly, initiated in the cytoplasm, uses G protein recycled from the plasma membrane.
Collapse
|
11
|
Ludwig A, Nguyen TH, Leong D, Ravi LI, Tan BH, Sandin S, Sugrue RJ. Caveolae provide a specialized membrane environment for respiratory syncytial virus assembly. J Cell Sci 2017; 130:1037-1050. [PMID: 28154158 PMCID: PMC5358342 DOI: 10.1242/jcs.198853] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/26/2017] [Indexed: 12/20/2022] Open
Abstract
Respiratory syncytial virus (RSV) is an enveloped virus that assembles into filamentous virus particles on the surface of infected cells. Morphogenesis of RSV is dependent upon cholesterol-rich (lipid raft) membrane microdomains, but the specific role of individual raft molecules in RSV assembly is not well defined. Here, we show that RSV morphogenesis occurs within caveolar membranes and that both caveolin-1 and cavin-1 (also known as PTRF), the two major structural and functional components of caveolae, are actively recruited to and incorporated into the RSV envelope. The recruitment of caveolae occurred just prior to the initiation of RSV filament assembly, and was dependent upon an intact actin network as well as a direct physical interaction between caveolin-1 and the viral G protein. Moreover, cavin-1 protein levels were significantly increased in RSV-infected cells, leading to a virus-induced change in the stoichiometry and biophysical properties of the caveolar coat complex. Our data indicate that RSV exploits caveolae for its assembly, and we propose that the incorporation of caveolae into the virus contributes to defining the biological properties of the RSV envelope.
Collapse
Affiliation(s)
- Alexander Ludwig
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | - Tra Huong Nguyen
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | - Daniel Leong
- Detection and Diagnostics Laboratory, DSO National Laboratories, 27 Medical Drive, Singapore 117510
| | - Laxmi Iyer Ravi
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | - Boon Huan Tan
- Detection and Diagnostics Laboratory, DSO National Laboratories, 27 Medical Drive, Singapore 117510
| | - Sara Sandin
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | - Richard J Sugrue
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| |
Collapse
|
12
|
Zheng LL, Li CM, Zhen SJ, Li YF, Huang CZ. His-tag based in situ labelling of progeny viruses for real-time single virus tracking in living cells. NANOSCALE 2016; 8:18635-18639. [PMID: 27805216 DOI: 10.1039/c6nr05806j] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Tracking virus infection events in live cells is useful for understanding the mechanism of virus infection, and fluorescent labelling is a critical step. Herein a noninvasive strategy for labelling viruses with His-tags was developed by in situ modifying the cell surface proteins with polypeptides containing His-tags during progeny virus assembly. The His-tagged viruses were further conjugated with Ni2+-nitrilotriacetate complex modified quantum dots, and retained their infectivity for real-time single virus tracking in living cells.
Collapse
Affiliation(s)
- Lin Ling Zheng
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China.
| | - Chun Mei Li
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China.
| | - Shu Jun Zhen
- Chongqing Key Laboratory of Biomedical Analysis (Southwest University), Chongqing Science & Technology Commission, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China
| | - Yuan Fang Li
- Chongqing Key Laboratory of Biomedical Analysis (Southwest University), Chongqing Science & Technology Commission, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China
| | - Cheng Zhi Huang
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China. and Chongqing Key Laboratory of Biomedical Analysis (Southwest University), Chongqing Science & Technology Commission, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China
| |
Collapse
|
13
|
Pattern and associated risk factors of caprine pneumonia complex in Nigeria. ASIAN PACIFIC JOURNAL OF TROPICAL DISEASE 2016. [DOI: 10.1016/s2222-1808(15)61009-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
14
|
Huong TN, Iyer Ravi L, Tan BH, Sugrue RJ. Evidence for a biphasic mode of respiratory syncytial virus transmission in permissive HEp2 cell monolayers. Virol J 2016; 13:12. [PMID: 26790623 PMCID: PMC4719537 DOI: 10.1186/s12985-016-0467-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/12/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND During respiratory syncytial virus (RSV) infection filamentous virus particles are formed on the cell surface. Although the virus infectivity remains cell-associated, low levels of cell-free virus is detected during advanced infection. It is currently unclear if this cell-free virus infectivity is due to a low-efficiency specific cell-release mechanism, or if it arises due to mechanical breakage following virus-induced cell damage at the advanced stage of infection. Understanding the origin of this cell-free virus is a prerequisite for understanding the mechanism of RSV transmission in permissive cells. In this study we describe a detailed examination of RSV transmission in permissive HEp2 cell monolayers. METHODS HEp2 cell monolayers were infected with RSV using a multiplicity of infection of 0.0002, and the course of infection monitored over 5 days. The progression of the virus infection within the cell monolayers was performed using bright-field microscopy to visualise the cell monolayer and immunofluorescence microscopy to detect virus-infected cells. The cell-associated and cell-free virus infectivity were determined by virus plaque assay, and the virus-induced cell cytotoxicity determined by measuring cell membrane permeability and cellular DNA fragmentation. RESULTS At 2 days-post infection (dpi), large clusters of virus-infected cells could be detected indicating localised transmission in the cell monolayer, and during this stage we failed to detect either cell-free virus or cell cytotoxicity. At 3 dpi the presence of much larger infected cell clusters correlated with the begining of virus-induced changes in cell permeability. The presence of cell-free virus correlated with continued increase in cell permeability and cytotoxicity at 4 and 5 dpi. At 5 dpi extensive cell damage, syncytial formation, and increased cellular DNA fragmentation was noted. However, even at 5 dpi the cell-free virus constituted less than 1 % of the total virus infectivity. CONCLUSIONS Our data supports a model of RSV transmission that initially involves the localised cell-to-cell spread of virus particles within the HEp2 cell monolayer. However, low levels of cell free-virus infectivity was observed at the advanced stages of infection, which correlated with a general loss in cell monolayer integrity due to virus-induced cytotoxicity.
Collapse
Affiliation(s)
- Tra Nguyen Huong
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore.
| | - Laxmi Iyer Ravi
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore.
| | - Boon Huan Tan
- Detection and Diagnostics Laboratory, DSO National Laboratories, 27 Medical Drive, Singapore, 117510, Singapore.
| | - Richard J Sugrue
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore.
| |
Collapse
|
15
|
Kumar N, Maherchandani S, Kashyap SK, Singh SV, Sharma S, Chaubey KK, Ly H. Peste des petits ruminants virus infection of small ruminants: a comprehensive review. Viruses 2014; 6:2287-327. [PMID: 24915458 PMCID: PMC4074929 DOI: 10.3390/v6062287] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 05/26/2014] [Accepted: 05/28/2014] [Indexed: 12/14/2022] Open
Abstract
Peste des petits ruminants (PPR) is caused by a Morbillivirus that belongs to the family Paramyxoviridae. PPR is an acute, highly contagious and fatal disease primarily affecting goats and sheep, whereas cattle undergo sub-clinical infection. With morbidity and mortality rates that can be as high as 90%, PPR is classified as an OIE (Office International des Epizooties)-listed disease. Considering the importance of sheep and goats in the livelihood of the poor and marginal farmers in Africa and South Asia, PPR is an important concern for food security and poverty alleviation. PPR virus (PPRV) and rinderpest virus (RPV) are closely related Morbilliviruses. Rinderpest has been globally eradicated by mass vaccination. Though a live attenuated vaccine is available against PPR for immunoprophylaxis, due to its instability in subtropical climate (thermo-sensitivity), unavailability of required doses and insufficient coverage (herd immunity), the disease control program has not been a great success. Further, emerging evidence of poor cross neutralization between vaccine strain and PPRV strains currently circulating in the field has raised concerns about the protective efficacy of the existing PPR vaccines. This review summarizes the recent advancement in PPRV replication, its pathogenesis, immune response to vaccine and disease control. Attempts have also been made to highlight the current trends in understanding the host susceptibility and resistance to PPR.
Collapse
Affiliation(s)
- Naveen Kumar
- Virology Laboratory, Division of Animal Health, Central Institute for Research on Goats, Makhdoom, P.O. Farah, Mathura, UP 281122, India.
| | - Sunil Maherchandani
- Department of Veterinary Microbiology and Biotechnology, Rajasthan University of Veterinary and Animal Sciences, Bikaner, Rajasthan 334001, India.
| | - Sudhir Kumar Kashyap
- Department of Veterinary Microbiology and Biotechnology, Rajasthan University of Veterinary and Animal Sciences, Bikaner, Rajasthan 334001, India.
| | - Shoor Vir Singh
- Virology Laboratory, Division of Animal Health, Central Institute for Research on Goats, Makhdoom, P.O. Farah, Mathura, UP 281122, India.
| | - Shalini Sharma
- Department of Veterinary Physiology and Biochemistry, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana 125004, India.
| | - Kundan Kumar Chaubey
- Virology Laboratory, Division of Animal Health, Central Institute for Research on Goats, Makhdoom, P.O. Farah, Mathura, UP 281122, India.
| | - Hinh Ly
- Veterinary and Biomedical Sciences Department, University of Minnesota, 1988 Fitch Ave., Ste 295, Saint Paul, MN 55108, USA.
| |
Collapse
|