1
|
Yamazaki E, Yazawa S, Shimada T, Tamura K, Saga Y, Itamochi M, Inasaki N, Hasegawa S, Morinaga Y, Oishi K, Tani H. Activation of SARS-CoV-2 by trypsin-like proteases in the clinical specimens of patients with COVID-19. Sci Rep 2023; 13:11632. [PMID: 37468582 DOI: 10.1038/s41598-023-38757-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/14/2023] [Indexed: 07/21/2023] Open
Abstract
SARS-CoV-2 enters host cells through the angiotensin converting enzyme 2 (ACE2) receptor and/or transmembrane protease, serine 2 (TMPRSS2). In this study, we investigated whether proteases increased SARS-CoV-2 infectivity using pseudotyped viruses and clinical specimens from patients with COVID-19. First, we investigated how trypsin increased infectivity using the pseudotyped virus. Our findings revealed that trypsin increased infectivity after the virus was adsorbed on the cells, but no increase in infectivity was observed when the virus was treated with trypsin. We examined the effect of trypsin on SARS-CoV-2 infection in clinical specimens and found that the infectivity of the SARS-CoV-2 delta variant increased 36,000-fold after trypsin treatment. By contrast, the infectivity of SARS-CoV-2 omicron variant increased to less than 20-fold in the clinical specimens. Finally, using five clinical specimens containing delta variants, enhancement of viral infectivity was evaluated in the presence of the culture supernatant of several anaerobic bacteria. As a result, viral infectivities of all the clinical specimens containing culture supernatants of Fusobacterium necrophorum were significantly increased from several- to tenfold. Because SARS-CoV-2 infectivity increases in the oral cavity, which may contain anaerobic bacteria, keeping the oral cavities clean may help prevent SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Emiko Yamazaki
- Department of Virology, Toyama Institute of Health, 17-1 Nakataikoyama, Imizu-Shi, Toyama, 939-0363, Japan
- Department of Microbiology, Osaka Institute of Public Health, Osaka, Japan
| | - Shunsuke Yazawa
- Department of Virology, Toyama Institute of Health, 17-1 Nakataikoyama, Imizu-Shi, Toyama, 939-0363, Japan
| | - Takahisa Shimada
- Department of Virology, Toyama Institute of Health, 17-1 Nakataikoyama, Imizu-Shi, Toyama, 939-0363, Japan
| | - Kosuke Tamura
- Department of Research Planning, Toyama Institute of Health, Toyama, Japan
| | - Yumiko Saga
- Department of Virology, Toyama Institute of Health, 17-1 Nakataikoyama, Imizu-Shi, Toyama, 939-0363, Japan
| | - Masae Itamochi
- Department of Virology, Toyama Institute of Health, 17-1 Nakataikoyama, Imizu-Shi, Toyama, 939-0363, Japan
| | - Noriko Inasaki
- Department of Virology, Toyama Institute of Health, 17-1 Nakataikoyama, Imizu-Shi, Toyama, 939-0363, Japan
| | - Sumiyo Hasegawa
- Department of Virology, Toyama Institute of Health, 17-1 Nakataikoyama, Imizu-Shi, Toyama, 939-0363, Japan
| | - Yoshitomo Morinaga
- Department of Microbiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | | | - Hideki Tani
- Department of Virology, Toyama Institute of Health, 17-1 Nakataikoyama, Imizu-Shi, Toyama, 939-0363, Japan.
| |
Collapse
|
2
|
Mohd-Qawiem F, Nawal-Amani AR, Faranieyza-Afiqah F, Yasmin AR, Arshad SS, Norfitriah MS, Nur-Fazila SH. Paramyxoviruses in rodents: A review. Open Vet J 2022; 12:868-876. [PMID: 36650879 PMCID: PMC9805762 DOI: 10.5455/ovj.2022.v12.i6.14] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 10/20/2022] [Indexed: 11/27/2022] Open
Abstract
Paramyxoviruses have been shown to infect a wide range of hosts, including rodents, and humans. Several novel murine paramyxoviruses have been discovered in the last several decades. Although these viruses are unclassified, they are recognized as Beilong virus, Mojiang virus (MojV), and Tailam virus in rats, Jeilongvirus, Nariva, Paju Apodemus paramyxovirus-1 and -2 in mice, and Pentlands paramyxovirus-1, -2, and -3 in squirrels. These paramyxoviruses were reported mainly in China and a few other countries like Australia, the Republic of Korea, Trinidad, and France. In June 2012, it becomes a great concern in China whereby, three miners were reported dead potentially caused by a novel zoonotic MojV, a henipa-like virus isolated from tissue samples of rats from the same cave. Rats are considered to be natural hosts for the MojV from the literature research. The classified paramyxovirus, Sendai virus in rodents is also reviewed. Paramyxoviruses infection in rodents leads to respiratory distress such as necrotizing rhinitis, tracheitis, bronchiolitis, and interstitial pneumonia. Infections caused by paramyxoviruses often spread between species, manifesting disease in spillover hosts, including humans. This review focuses on the paramyxoviruses in rodents, including the epidemiological distributions, transmission and pathogenesis, clinical manifestations, diagnostic methods, and control and prevention of paramyxoviruses infection to provide a better understanding of these highly mutating viruses.
Collapse
Affiliation(s)
- Firdaus Mohd-Qawiem
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Malaysia
| | - Abdul Rahman Nawal-Amani
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Malaysia
| | - Farzee Faranieyza-Afiqah
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Malaysia
| | - Abd Rahaman Yasmin
- Department of Veterinary Laboratory Diagnostics, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Malaysia
- Laboratory of Vaccines and Immunotherapeutic, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
| | - Siti Suri Arshad
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Malaysia
| | - Mohamed Sohaimi Norfitriah
- Department of Veterinary Laboratory Diagnostics, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Malaysia
- Laboratory of Vaccines and Immunotherapeutic, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
| | - Saulol Hamid Nur-Fazila
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
3
|
Matveeva OV, Shabalina SA. Prospects for Using Expression Patterns of Paramyxovirus Receptors as Biomarkers for Oncolytic Virotherapy. Cancers (Basel) 2020; 12:cancers12123659. [PMID: 33291506 PMCID: PMC7762160 DOI: 10.3390/cancers12123659] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/28/2020] [Accepted: 12/01/2020] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Some non-pathogenic viruses that do not cause serious illness in humans can efficiently target and kill cancer cells and may be considered candidates for cancer treatment with virotherapy. However, many cancer cells are protected from viruses. An important goal of personalized cancer treatment is to identify viruses that can kill a certain type of cancer cells. To this end, researchers investigate expression patterns of cell entry receptors, which viruses use to bind to and enter host cells. We summarized and analyzed the receptor expression patterns of two paramyxoviruses: The non-pathogenic measles and the Sendai viruses. The receptors for these viruses are different and can be proteins or lipids with attached carbohydrates. This review discusses the prospects for using these paramyxovirus receptors as biomarkers for successful personalized virotherapy for certain types of cancer. Abstract The effectiveness of oncolytic virotherapy in cancer treatment depends on several factors, including successful virus delivery to the tumor, ability of the virus to enter the target malignant cell, virus replication, and the release of progeny virions from infected cells. The multi-stage process is influenced by the efficiency with which the virus enters host cells via specific receptors. This review describes natural and artificial receptors for two oncolytic paramyxoviruses, nonpathogenic measles, and Sendai viruses. Cell entry receptors are proteins for measles virus (MV) and sialylated glycans (sialylated glycoproteins or glycolipids/gangliosides) for Sendai virus (SeV). Accumulated published data reviewed here show different levels of expression of cell surface receptors for both viruses in different malignancies. Patients whose tumor cells have low or no expression of receptors for a specific oncolytic virus cannot be successfully treated with the virus. Recent published studies have revealed that an expression signature for immune genes is another important factor that determines the vulnerability of tumor cells to viral infection. In the future, a combination of expression signatures of immune and receptor genes could be used to find a set of oncolytic viruses that are more effective for specific malignancies.
Collapse
Affiliation(s)
- Olga V. Matveeva
- Sendai Viralytics LLC, 23 Nylander Way, Acton, MA 01720, USA
- Correspondence: (O.V.M.); (S.A.S.)
| | - Svetlana A. Shabalina
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
- Correspondence: (O.V.M.); (S.A.S.)
| |
Collapse
|
4
|
Mitra P. Inhibiting fusion with cellular membrane system: therapeutic options to prevent severe acute respiratory syndrome coronavirus-2 infection. Am J Physiol Cell Physiol 2020; 319:C500-C509. [PMID: 32687406 PMCID: PMC7839238 DOI: 10.1152/ajpcell.00260.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV), an enveloped virus with a positive-sense single-stranded RNA genome, facilitates the host cell entry through intricate interactions with proteins and lipids of the cell membrane. The detailed molecular mechanism involves binding to the host cell receptor and fusion at the plasma membrane or after being trafficked to late endosomes under favorable environmental conditions. A crucial event in the process is the proteolytic cleavage of the viral spike protein by the host's endogenous proteases that releases the fusion peptide enabling fusion with the host cellular membrane system. The present review details the mechanism of viral fusion with the host and highlights the therapeutic options that prevent SARS-CoV-2 entry in humans.
Collapse
Affiliation(s)
- Prasenjit Mitra
- Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Hyderabad, Telangana, India
| |
Collapse
|
5
|
Azarm KD, Lee B. Differential Features of Fusion Activation within the Paramyxoviridae. Viruses 2020; 12:v12020161. [PMID: 32019182 PMCID: PMC7077268 DOI: 10.3390/v12020161] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/25/2020] [Accepted: 01/29/2020] [Indexed: 12/13/2022] Open
Abstract
Paramyxovirus (PMV) entry requires the coordinated action of two envelope glycoproteins, the receptor binding protein (RBP) and fusion protein (F). The sequence of events that occurs during the PMV entry process is tightly regulated. This regulation ensures entry will only initiate when the virion is in the vicinity of a target cell membrane. Here, we review recent structural and mechanistic studies to delineate the entry features that are shared and distinct amongst the Paramyxoviridae. In general, we observe overarching distinctions between the protein-using RBPs and the sialic acid- (SA-) using RBPs, including how their stalk domains differentially trigger F. Moreover, through sequence comparisons, we identify greater structural and functional conservation amongst the PMV fusion proteins, as compared to the RBPs. When examining the relative contributions to sequence conservation of the globular head versus stalk domains of the RBP, we observe that, for the protein-using PMVs, the stalk domains exhibit higher conservation and find the opposite trend is true for SA-using PMVs. A better understanding of conserved and distinct features that govern the entry of protein-using versus SA-using PMVs will inform the rational design of broader spectrum therapeutics that impede this process.
Collapse
|
6
|
Abstract
The distribution pattern of host proteases and their cleavage specificity for viral fusion glycoproteins are key determinants for viral tissue tropism and pathogenicity. The discovery of this protease-dependent virus tropism and pathogenicity has been triggered by the leading studies of the host-induced or -controlled modification of viruses by Homma et al. in 1970s. With the introduction of advanced protein analysis method, the observations by Homma et al. have been clearly explained by the cleavage activation of viral fusion glycoproteins by proteases. The molecular biological features of viruses, which show distinct protease specificity or dependency, have been also revealed by newly introduced nucleotide and molecular analysis method. Highly pathogenic avian influenza viruses (HPAIVs) have multi-basic cleavage motif in the hemagglutinin (HA) protein and are activated proteolytically by furin. Furin is ubiquitously expressed in eukaryotic cells and thereby HPAIVs have the potential to cause a systemic infection in infected animals. On the other hand, the HA cleavage site of low pathogenic avian influenza viruses (LPAIVs) and seasonal human influenza viruses is mono-basic and thus not recognized by furin. They are likely cleaved by protease(s) localized in specific organs or tissues. However, the protease(s), which cleaves mono-basic HA in vivo, has long been undetermined, although many proteases have been shown as candidates. Finally, recent studies using gene knocked out mice revealed that TMPRSS2, a member of type II transmembrane serine proteases, is responsible for the cleavage of influenza viruses with a mono-basic HA in vivo. A subsequent study further demonstrated that TMPRSS2 contributes to replication and pathology of emerging SARS- and MERS coronaviruses in vivo.
Collapse
|
7
|
Matveeva OV, Kochneva GV, Zainutdinov SS, Ilyinskaya GV, Chumakov PM. Oncolytic Paramyxoviruses: Mechanism of Action, Preclinical and Clinical Studies. Mol Biol 2018. [DOI: 10.1134/s002689331803010x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
8
|
Šantak M, Slović A, Ljubin-Sternak S, Mlinarić Galinović G, Forčić D. Genetic diversity among human parainfluenza virus type 2 isolated in Croatia between 2011 and 2014. J Med Virol 2016; 88:1733-41. [PMID: 27004845 DOI: 10.1002/jmv.24532] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2016] [Indexed: 11/09/2022]
Abstract
The dynamics and evolution of the human parainfluenza virus type 2 (HPIV2) in Croatia, and also globally, are largely unknown. Most HPIV2 infections are treated symptomatically outside the hospital setting. Thus, the diagnosis is missing making it difficult to follow the genetic variation and evolution of the HPIV2. This study explores hospitalized HPIV2 cases in Croatia during 4-year period (2011-2014). Most cases in this period were reported in October or November (68.75%) and most of patients were under 2 years of age (81.25%). For molecular analyses, we used the F and HN gene sequences and showed that although both regions are equally suitable for phylogenetic analyses it would be advantageous to use regions longer than 2 kb for HPIV2 analyses of isolates which are spatially and temporally closely related. We show here that the dominant cluster in this area was cluster G3 while only one strain isolated in this period was positioned in the distant cluster G1a. Further monitoring of the HPIV2 will determine whether cluster G3 will remain dominant or it will be overruled by cluster G1a. This will be important for the surveillance of virus circulation in population and significance of the viral infection. J. Med. Virol. 88:1733-1741, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Maja Šantak
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Zagreb, Croatia
| | - Anamarija Slović
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Zagreb, Croatia
| | - Sunčanica Ljubin-Sternak
- Teaching Institute of Public Health "Dr. Andrija Štampar", Zagreb, Croatia.,School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Gordana Mlinarić Galinović
- School of Medicine, University of Zagreb, Zagreb, Croatia.,Croatian National Institute of Public Health, Zagreb, Croatia
| | - Dubravko Forčić
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
9
|
A serpin shapes the extracellular environment to prevent influenza A virus maturation. Cell 2015; 160:631-643. [PMID: 25679759 PMCID: PMC4328142 DOI: 10.1016/j.cell.2015.01.040] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 10/22/2014] [Accepted: 01/13/2015] [Indexed: 01/23/2023]
Abstract
Interferon-stimulated genes (ISGs) act in concert to provide a tight barrier against viruses. Recent studies have shed light on the contribution of individual ISG effectors to the antiviral state, but most have examined those acting on early, intracellular stages of the viral life cycle. Here, we applied an image-based screen to identify ISGs inhibiting late stages of influenza A virus (IAV) infection. We unraveled a directly antiviral function for the gene SERPINE1, encoding plasminogen activator inhibitor 1 (PAI-1). By targeting extracellular airway proteases, PAI-1 inhibits IAV glycoprotein cleavage, thereby reducing infectivity of progeny viruses. This was biologically relevant for IAV restriction in vivo. Further, partial PAI-1 deficiency, attributable to a polymorphism in human SERPINE1, conferred increased susceptibility to IAV in vitro. Together, our findings reveal that manipulating the extracellular environment to inhibit the last step in a virus life cycle is an important mechanism of the antiviral response. SERPINE1/PAI-1 was identified as an unconventional ISG that acts extracellularly PAI-1 inhibits influenza A virus (IAV) spread by inhibiting glycoprotein cleavage Endogenous PAI-1 blocks IAV spread in human and murine cells, ex vivo and in vivo PAI-1 potentially inhibits other viruses requiring extracellular maturation
Collapse
|
10
|
Matveeva OV, Kochneva GV, Netesov SV, Onikienko SB, Chumakov PM. Mechanisms of Oncolysis by Paramyxovirus Sendai. Acta Naturae 2015; 7:6-16. [PMID: 26085940 PMCID: PMC4463408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Some viral strains of the Paramyxoviridae family may be used as anti-tumor agents. Oncolytic paramyxoviruses include attenuated strains of the measles virus, Newcastle disease virus, and Sendai virus. These viral strains, and the Sendai virus in particular, can preferentially induce the death of malignant, rather than normal, cells. The death of cancer cells results from both direct killing by the virus and through virus-induced activation of anticancer immunity. Sialic-acid-containing glycoproteins that are overexpressed in cancer cells serve as receptors for some oncolytic paramyxoviruses and ensure preferential interaction of paramyxoviruses with malignant cells. Frequent genetic defects in interferon and apoptotic response systems that are common to cancer cells ensure better susceptibility of malignant cells to viruses. The Sendai virus as a Paramyxovirus is capable of inducing the formation of syncytia, multinuclear cell structures which promote viral infection spread within a tumor without virus exposure to host neutralizing antibodies. As a result, the Sendai virus can cause mass killing of malignant cells and tumor destruction. Oncolytic paramyxoviruses can also promote the immune-mediated elimination of malignant cells. In particular, they are powerful inducers of interferon and other cytokynes promoting antitumor activity of various cell components of the immune response, such as dendritic and natural killer cells, as well as cytotoxic T lymphocytes. Taken together these mechanisms explain the impressive oncolytic activity of paramyxoviruses that hold promise as future, efficient anticancer therapeutics.
Collapse
Affiliation(s)
- O. V. Matveeva
- Biopolymer Design, 23 Nylander Way, Acton, Massachusetts, United States
| | - G. V. Kochneva
- Center of Virology and Biotechnology “Vector”, Koltsovo, Novosibirsk Region, Russia
| | | | - S. B. Onikienko
- Department of Military Field Therapy, Kirov Military Medical Academy, St. Petersburg, Russia
| | - P. M. Chumakov
- Engelhardt Institute of Molecular Biology, Moscow, Russia
| |
Collapse
|
11
|
TMPRSS2 is an activating protease for respiratory parainfluenza viruses. J Virol 2013; 87:11930-5. [PMID: 23966399 DOI: 10.1128/jvi.01490-13] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Here, we show that human parainfluenza viruses and Sendai virus (SeV), like other respiratory viruses, use TMPRSS2 for their activation. The membrane fusion proteins of respiratory viruses often possess serine and glutamine residues at the P2 and P3 positions, respectively, but these residues were not critical for cleavage by TMPRSS2. However, mutations of these residues affected SeV growth in specific epithelial cell lines, suggesting the importance of these residues for SeV replication in epithelia.
Collapse
|
12
|
Abstract
The influenza virus (IV) is still of great importance as it poses an immanent threat to humans and animals. Among the three IV-types (A, B, and C) influenza A viruses are clinically the most important being responsible for severe epidemics in humans and domestic animals. Aerosol droplets transmit the virus that causes a respiratory disease in humans that can lead to severe pneumonia and ultimately death. The high mutation rate combined with the high replication rate allows the virus to rapidly adapt to changes in the environment. Thereby, IV escape the existing immunity and become resistant to drugs targeting the virus. This causes annual epidemics and demands for new compositions of the yearly vaccines. Furthermore, due to the nature of their segmented genome, IV can recombine segments. This can eventually lead to the generation of a virus with the ability to replicate in humans and with novel antigenic properties that can be the cause of a pandemic outbreak. For its propagation the virus binds to the target cells and enters the cell to replicate its genome. Newly produced viral proteins and genomes are packaged at the cell membrane where progeny virions are released. As all viruses IV depends on cellular functions and factors for their own propagation, and therefore intensively interact with the cells. This dependency opens new possibilities for anti-viral strategies.
Collapse
|
13
|
Salti SM, Hammelev EM, Grewal JL, Reddy ST, Zemple SJ, Grossman WJ, Grayson MH, Verbsky JW. Granzyme B regulates antiviral CD8+ T cell responses. THE JOURNAL OF IMMUNOLOGY 2011; 187:6301-9. [PMID: 22084442 DOI: 10.4049/jimmunol.1100891] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CTLs and NK cells use the perforin/granzyme cytotoxic pathway to kill virally infected cells and tumors. Human regulatory T cells also express functional granzymes and perforin and can induce autologous target cell death in vitro. Perforin-deficient mice die of excessive immune responses after viral challenges, implicating a potential role for this pathway in immune regulation. To further investigate the role of granzyme B in immune regulation in response to viral infections, we characterized the immune response in wild-type, granzyme B-deficient, and perforin-deficient mice infected with Sendai virus. Interestingly, granzyme B-deficient mice, and to a lesser extent perforin-deficient mice, exhibited a significant increase in the number of Ag-specific CD8(+) T cells in the lungs and draining lymph nodes of virally infected animals. This increase was not the result of failure in viral clearance because viral titers in granzyme B-deficient mice were similar to wild-type mice and significantly less than perforin-deficient mice. Regulatory T cells from WT mice expressed high levels of granzyme B in response to infection, and depletion of regulatory T cells from these mice resulted in an increase in the number of Ag-specific CD8(+) T cells, similar to that observed in granzyme B-deficient mice. Furthermore, granzyme B-deficient regulatory T cells displayed defective suppression of CD8(+) T cell proliferation in vitro. Taken together, these results suggest a role for granzyme B in the regulatory T cell compartment in immune regulation to viral infections.
Collapse
Affiliation(s)
- Suzan M Salti
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Illumination of parainfluenza virus infection and transmission in living animals reveals a tissue-specific dichotomy. PLoS Pathog 2011; 7:e1002134. [PMID: 21750677 PMCID: PMC3131265 DOI: 10.1371/journal.ppat.1002134] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Accepted: 05/07/2011] [Indexed: 11/19/2022] Open
Abstract
The parainfluenza viruses (PIVs) are highly contagious respiratory paramyxoviruses and a leading cause of lower respiratory tract (LRT) disease. Since no vaccines or antivirals exist, non-pharmaceutical interventions are the only means of control for these pathogens. Here we used bioluminescence imaging to visualize the spatial and temporal progression of murine PIV1 (Sendai virus) infection in living mice after intranasal inoculation or exposure by contact. A non-attenuated luciferase reporter virus (rSeV-luc(M-F*)) that expressed high levels of luciferase yet was phenotypically similar to wild-type Sendai virus in vitro and in vivo was generated to allow visualization. After direct intranasal inoculation, we unexpectedly observed that the upper respiratory tract (URT) and trachea supported robust infection under conditions that result in little infection or pathology in the lungs including a low inoculum of virus, an attenuated virus, and strains of mice genetically resistant to lung infection. The high permissivity of the URT and trachea to infection resulted in 100% transmission to naïve contact recipients, even after low-dose (70 PFU) inoculation of genetically resistant BALB/c donor mice. The timing of transmission was consistent with the timing of high viral titers in the URT and trachea of donor animals but was independent of the levels of infection in the lungs of donors. The data therefore reveals a disconnect between transmissibility, which is associated with infection in the URT, and pathogenesis, which arises from infection in the lungs and the immune response. Natural infection after transmission was universally robust in the URT and trachea yet limited in the lungs, inducing protective immunity without weight loss even in genetically susceptible 129/SvJ mice. Overall, these results reveal a dichotomy between PIV infection in the URT and trachea versus the lungs and define a new model for studies of pathogenesis, development of live virus vaccines, and testing of antiviral therapies. Human parainfluenza viruses (HPIVs) are a leading cause of pediatric hospitalization for lower respiratory tract infection, yet it is unknown why primary infection typically induces immunity without causing severe pathology. To study the determinants of PIV spread within the respiratory tracts of living animals, we developed a model for non-invasive imaging of living mice infected with Sendai virus, the murine counterpart of HPIV1. This system allowed us to measure the temporal and spatial dynamics of paramyxovirus infection throughout the respiratory tracts of living animals after direct inoculation or transmission. We found that the upper respiratory tract and trachea were highly permissive to infection, even under conditions that limit lower respiratory infection and pathogenesis. The timing of transmission coincided with high virus growth in the upper respiratory tracts and trachea of donor mice independent of the extent of infection in the lungs. After transmission, infection spread preferentially in the upper respiratory tract and trachea, inducing protective immunity without weight loss. Our work reveals a disconnect between Sendai virus transmissibility and pathogenicity, and the experimental model developed here will be instrumental in studying PIV pathogenesis.
Collapse
|
15
|
Kido H, Ishidoh K. Nobuhiko Katunuma: an outstanding scientist in the field of proteolysis and warm-hearted 'Kendo Fighter' biochemist. J Biochem 2011; 148:527-31. [PMID: 20980477 DOI: 10.1093/jb/mvq109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Professor Nobuhiko Katunuma is well known for his outstanding contribution to the understanding of proteolysis in general and cysteine proteinases and their inhibitors in mammals. In fact, he is a world pioneer in the field. In 1963, he started his highly successful scientific career as a Professor at the Institute for Enzyme Research, the University of Tokushima. During the initial 30 years of his career, he was interested in vitamin B6 metabolism and discovered the acceleration of turnover rates of pyridoxal enzyme in apoprotein formation. After this period, his interest expanded to lysosomal cystein proteinases and their endogenous inhibitors. After determining the crystal structure of human cathepsin B, he generated a series of chemically synthesized specific inhibitors of cathepsins. These inhibitors are currently used throughout the world and some of them have been applied therapeutically in various diseases. During his career and even at present, Professor Katunuma has been studying Biochemistry in Medicine and also practicing to become a 'Kendo sword fencing Fighter'.
Collapse
Affiliation(s)
- Hiroshi Kido
- Division of Enzyme Chemistry, Institute for Enzyme Research, The University of Tokushima, Kuramotocho 3-18-15, Tokushima 770-8503, Japan.
| | | |
Collapse
|
16
|
Cytopathogenesis of Sendai virus in well-differentiated primary pediatric bronchial epithelial cells. J Virol 2010; 84:11718-28. [PMID: 20810726 DOI: 10.1128/jvi.00798-10] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Sendai virus (SeV) is a murine respiratory virus of considerable interest as a gene therapy or vaccine vector, as it is considered nonpathogenic in humans. However, little is known about its interaction with the human respiratory tract. To address this, we developed a model of respiratory virus infection based on well-differentiated primary pediatric bronchial epithelial cells (WD-PBECs). These physiologically authentic cultures are comprised of polarized pseudostratified multilayered epithelium containing ciliated, goblet, and basal cells and intact tight junctions. To facilitate our studies, we rescued a replication-competent recombinant SeV expressing enhanced green fluorescent protein (rSeV/eGFP). rSeV/eGFP infected WD-PBECs efficiently and progressively and was restricted to ciliated and nonciliated cells, not goblet cells, on the apical surface. Considerable cytopathology was evident in the rSeV/eGFP-infected cultures postinfection. This manifested itself by ciliostasis, cell sloughing, apoptosis, and extensive degeneration of WD-PBEC cultures. Syncytia were also evident, along with significant basolateral secretion of proinflammatory chemokines, including IP-10, RANTES, tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), interleukin 6 (IL-6), and IL-8. Such deleterious responses are difficult to reconcile with a lack of pathogenesis in humans and suggest that caution may be required in exploiting replication-competent SeV as a vaccine vector. Alternatively, such robust responses might constitute appropriate normal host responses to viral infection and be a prerequisite for the induction of efficient immune responses.
Collapse
|
17
|
Hermesh T, Moltedo B, Moran TM, López CB. Antiviral instruction of bone marrow leukocytes during respiratory viral infections. Cell Host Microbe 2010; 7:343-53. [PMID: 20478536 PMCID: PMC2874206 DOI: 10.1016/j.chom.2010.04.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Revised: 02/04/2010] [Accepted: 03/26/2010] [Indexed: 12/19/2022]
Abstract
Respiratory viral infections trigger a robust inflammatory response in the lung, producing cytokines, chemokines, and growth factors that promote infiltration of effector leukocytes. Whereas the role of chemokines and infiltrating leukocytes in antiviral immunity is well studied, the effect that lung cytokines have on leukocytes in distal hematopoietic and lymphoid tissues and their role in antiviral immunity is unknown. We show that, during infection with influenza or Sendai virus, the lung communicates with the sterile bone marrow, the primary site of hematopoiesis, through type I interferons. While in the bone marrow, leukocytes exposed to type I interferons activate an antiviral transcriptional program and become resistant to infection with different viruses. The protected bone marrow leukocytes are capable of migrating to the infected lung and contribute to virus clearance. These findings show that appropriate instruction of cells during their development in the bone marrow is needed for effective control of infection.
Collapse
Affiliation(s)
- Tamar Hermesh
- Department of Microbiology and Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| | - Bruno Moltedo
- Department of Microbiology and Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| | - Thomas M. Moran
- Department of Microbiology and Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| | - Carolina B. López
- Department of Microbiology and Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| |
Collapse
|
18
|
Residues in the heptad repeat a region of the fusion protein modulate the virulence of Sendai virus in mice. J Virol 2009; 84:810-21. [PMID: 19906935 DOI: 10.1128/jvi.01990-09] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
While the molecular basis of fusion (F) protein refolding during membrane fusion has been studied extensively in vitro, little is known about the biological significance of membrane fusion activity in parainfluenza virus replication and pathogenesis in vivo. Two recombinant Sendai viruses, F-L179V and F-K180Q, were generated that contain F protein mutations in the heptad repeat A region of the ectodomain, a region of the protein known to regulate F protein activation. In vitro, the F-L179V virus caused increased syncytium formation (cell-cell membrane fusion) yet had a rate of replication and levels of F protein expression and cleavage similar to wild-type virus. The F-K180Q virus had a reduced replication rate along with reduced levels of F protein expression, cleavage, and fusogenicity. In DBA/2 mice, the hyperfusogenic F-L179V virus induced greater morbidity and mortality than wild-type virus, while the attenuated F-K180Q virus was much less pathogenic. During the first week of infection, virus replication and inflammation in the lungs were similar for wild-type and F-L179V viruses. After approximately 1 week of infection, the clearance of F-L179V virus was delayed, and more extensive interstitial inflammation and necrosis were observed in the lungs, affecting entire lobes of the lungs and having significantly greater numbers of syncytial cell masses in alveolar spaces on day 10. On the other hand, the slower-growing F-K180Q virus caused much less extensive inflammation than wild-type virus, presumably due to its reduced replication rate, and did not cause observable syncytium formation in the lungs. Overall, the results show that residues in the heptad repeat A region of the F protein modulate the virulence of Sendai virus in mice by influencing both the spread and clearance of the virus and the extent and severity of inflammation. An understanding of how the F protein contributes to infection and inflammation in vivo may assist in the development of antiviral therapies against respiratory paramyxoviruses.
Collapse
|
19
|
Nipah virus fusion protein: influence of cleavage site mutations on the cleavability by cathepsin L, trypsin and furin. Virus Res 2009; 145:300-6. [PMID: 19665506 PMCID: PMC7126315 DOI: 10.1016/j.virusres.2009.07.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2009] [Revised: 07/28/2009] [Accepted: 07/29/2009] [Indexed: 11/22/2022]
Abstract
Nipah virus (NiV), a highly pathogenic member of the Paramyxoviridae which originated from bats, encodes for a fusion (F) protein which is proteolytically processed within endosomes by cathepsin L. We show here that sequence requirements for NiV F activation differ markedly from other para- or orthomyxoviral fusion proteins. In contrast to other viral fusion proteins with monobasic cleavage sites, processing of NiV F proteins with one single basic amino acid in the cleavage peptide by exogenous trypsin is very inefficient, and introduction of a consensus sequence for furin does not result in cleavage by this ubiquitous protease. In contrast, a multibasic cleavage peptide in the NiV F protein completely impairs proteolytic processing and the generation of biological activity.
Collapse
|
20
|
A new paramyxovirus, Tianjin strain, isolated from common cotton-eared marmoset: genome characterization and structural protein sequence analysis. Arch Virol 2008; 153:1715-23. [PMID: 18696006 DOI: 10.1007/s00705-008-0184-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2008] [Accepted: 08/01/2008] [Indexed: 10/21/2022]
Abstract
The complete RNA genome sequence of Tianjin strain, isolated from marmoset, has been determined. The genome was 15,384 nucleotides (nt) in length, which was identical to that of Sendai virus (SeV), and consisted of six genes in the order 3'-NP-P/C-M-F-HN-L-5'. The gene junctions contained highly conserved transcription start and stop signal sequences and trinucleotide intergenic regions (IGR) similar to those of SeV. There was also an overlapping open reading frame and a conserved AnGn site (AAAAAGGG) in the P mRNA. Phylogenetic analysis based on protein sequences from other members of the family Paramyxoviridae showed that Tianjin strain belonged to the genus Respirovirus and was most closely related to SeV. Pairwise comparisons of amino acid identities and phylogenetic analysis with homologous sequences of known SeVs demonstrated that Tianjin strain represented a new evolutionary lineage of SeV.
Collapse
|
21
|
Shi LY, Li M, Li XM, Yuan LJ, Wang Q. Bioinformatic analysis of structural proteins of paramyxovirus Tianjin strain. Virol Sin 2008. [DOI: 10.1007/s12250-008-2947-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
22
|
Kosai K, Seki M, Yanagihara K, Nakamura S, Kurihara S, Izumikawa K, Kakeya H, Yamamoto Y, Tashiro T, Kohno S. Gabexate mesilate suppresses influenza pneumonia in mice through inhibition of cytokines. J Int Med Res 2008; 36:322-8. [PMID: 18380943 DOI: 10.1177/147323000803600215] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Gabexate mesilate is a synthetic protease inhibitor that is effective for acute pancreatitis. The effect of gabexate mesilate in influenza pneumonia in mice was investigated by examining the changes in pulmonary inflammatory cytokines and chemokines. Pathological changes in the lungs of treated mice were extremely mild, compared with changes in infected, untreated mice. Intrapulmonary levels of interleukin-6 and macrophage inflammatory protein-2 decreased in treated mice compared with untreated mice, despite similar viral titres in the lungs. Survival terms for treated and untreated groups were similar. These data indicate that gabexate mesilate has beneficial effects on influenza pneumonia, which may be due to the modulation of inflammatory cytokine/chemokine responses.
Collapse
Affiliation(s)
- K Kosai
- Department of Molecular Microbiology and Immunology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki University School of Medicine, Nagasaki, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Nipah virus (NiV) is a highly pathogenic paramyxovirus, which emerged in 1998 from fruit bats in Malaysia and caused an outbreak of severe respiratory disease in pigs and fatal encephalitis in humans with high mortality rates. In contrast to most paramyxoviruses, NiV can infect a large variety of mammalian species. Due to this broad host range, its zoonotic potential, its high pathogenicity for humans, and the lack of effective vaccines or therapeutics, NiV was classified as a biosafety level 4 pathogen. This article provides an overview of the molecular characteristics of NiV focusing on the structure, functions, and unique biological properties of the two NiV surface glycoproteins, the receptor-binding G protein, and the fusion protein F. Since viral glycoproteins are major determinants for cell tropism and virus spread, a detailed knowledge of these proteins can help to understand the molecular basis of viral pathogenicity.
Collapse
Affiliation(s)
- Sandra Diederich
- Philipps-Universität Marburg, Institut für Virologie, Hans-Meerwein-Str 2, Marburg, Germany
| | | |
Collapse
|
24
|
Springfeld C, von Messling V, Frenzke M, Ungerechts G, Buchholz CJ, Cattaneo R. Oncolytic efficacy and enhanced safety of measles virus activated by tumor-secreted matrix metalloproteinases. Cancer Res 2006; 66:7694-700. [PMID: 16885371 DOI: 10.1158/0008-5472.can-06-0538] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancer cells secrete matrix metalloproteinases (MMP) that degrade the extracellular matrix and are responsible for some hallmarks of malignant cancer. Many viruses, including a few currently used in oncolytic virotherapy clinical trials, depend on intracellular proteases to process their proteins and activate their particles. We show here for measles virus (MV) that particle activation can be made dependent of proteases secreted by cancer cells. The MV depends on the intracellular protease furin to process and activate its envelope fusion (F) protein. To make F protein activation cancer cell specific, we introduced hexameric sequences recognized by an MMP and identified the mutant proteins most effective in fusing MMP-expressing human fibrosarcoma cells (HT1080). We showed that an MMP inhibitor interferes with syncytia formation elicited by mutant F proteins and confirmed MMP-dependent cleavage by Edman degradation sequence analysis. We generated recombinant MVs expressing the modified F proteins in place of furin-activated F. These viruses spread only in cells secreting MMP. In nude mice, an MMP-activated MV retarded HT1080 xenograft growth as efficiently as the furin-activated MV vaccine strain. In MV-susceptible mice, the furin-activated virus caused lethal encephalitis upon intracerebral inoculation, whereas the MMP-activated did not. Thus, MV particle activation can be made dependent of proteases secreted by cancer cells, enhancing safety. This study opens the perspective of combining targeting at the particle activation, receptor recognition, and selective replication levels to improve the therapeutic index of MV and other viruses in ongoing clinical trials of oncolysis.
Collapse
Affiliation(s)
- Christoph Springfeld
- Molecular Medicine Program and Virology and Gene Therapy Track, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55902, USA
| | | | | | | | | | | |
Collapse
|
25
|
Torashima T, Yamada N, Itoh M, Yamamoto A, Hirai H. Exposure of lentiviral vectors to subneutral pH shifts the tropism from Purkinje cell to Bergmann glia. Eur J Neurosci 2006; 24:371-80. [PMID: 16836635 DOI: 10.1111/j.1460-9568.2006.04927.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Cerebellar Purkinje cells play an important role in cerebellar function; lesions of Purkinje cells result in the disruption of motor coordination and motor learning. Although selective gene delivery to Purkinje cells would be a powerful technique for the study of pathophysiology in the cerebellum, a method for such a delivery has not yet been established. Here we employed human immunodeficiency virus-derived lentiviral vectors pseudotyped with vesicular stomatitis virus glycoprotein to transduce Purkinje cells and examined factors that critically affect the viral tropism for Purkinje cells. Viral vectors encoding GFP were generated using different protocols, and were then injected into the mouse cerebellum. At 7 days and 2 months post-transduction, the relative proportions of transduced Purkinje cells were determined. Lentiviral vectors harvested from a medium of pH 7.2 preferentially transduced Purkinje cells (about half of the transduced cells). In contrast, when the viral vector was harvested from medium of <or= pH 7.0, only 12-26% of transduced cells were identified as Purkinje cells and 68-77% as Bergmann glia. A similar decrease in the efficiency of transduction for Purkinje cells, depending on the pH of the medium at the viral harvest, was observed in dissociated cell cultures. These results indicate that lentivector tropism for Purkinje cells is extremely sensitive to pH: a subtle decrease in the pH of the medium at the harvest shifts viral tropism strikingly towards Bergmann glia.
Collapse
Affiliation(s)
- Takashi Torashima
- Innovative Brain Science Project, Advanced Science Research Center, Kanazawa University, Kanazawa 920-8640, Japan
| | | | | | | | | |
Collapse
|
26
|
Yamada H, Le QT, Kousaka A, Higashi Y, Tsukane M, Kido H. Sendai virus infection up-regulates trypsin I and matrix metalloproteinase-9, triggering viral multiplication and matrix degradation in rat lungs and lung L2 cells. Arch Virol 2006; 151:2529-37. [PMID: 16816895 DOI: 10.1007/s00705-006-0807-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2006] [Accepted: 05/16/2006] [Indexed: 11/29/2022]
Abstract
To elucidate the virus-host cell interaction, we analyzed quantitatively the expression of various cellular proteases and tumor necrosis factor-alpha (TNF-alpha) after Sendai virus infection in rat lungs and lung L2 cells. After infection, TNF-alpha mRNA levels increased rapidly to a peak on day one, and then trypsin I and matrix metalloproteinase (MMP)-9, but not MMP-2, were significantly up-regulated with a peak on day 2 in vivo. These up-regulations were confirmed in L2 cells. Up-regulation of proMMP-9 and its active convertase trypsin I seems to synergistically enhance virus multiplication and the destruction of lung matrix, resulting in the progression of pneumonia.
Collapse
Affiliation(s)
- H Yamada
- Division of Enzyme Chemistry, Institute for Enzyme Research, The University of Tokushima, Tokushima, Japan
| | | | | | | | | | | |
Collapse
|
27
|
Abstract
Hendra virus and Nipah virus are highly pathogenic paramyxoviruses that have recently emerged from flying foxes to cause serious disease outbreaks in humans and livestock in Australia, Malaysia, Singapore and Bangladesh. Their unique genetic constitution, high virulence and wide host range set them apart from other paramyxoviruses. These features led to their classification into the new genus Henipavirus within the family Paramyxoviridae and to their designation as Biosafety Level 4 pathogens. This review provides an overview of henipaviruses and the types of infection they cause, and describes how studies on the structure and function of henipavirus proteins expressed from cloned genes have provided insights into the unique biological properties of these emerging human pathogens.
Collapse
Affiliation(s)
- Bryan T Eaton
- Australian Animal Health Laboratory, Commonwealth Scientific and Industrial Research Organization, 5 Portarlington Road, Geelong, Victoria 3220, Australia.
| | | | | | | |
Collapse
|
28
|
Franke J, Batts WN, Ahne W, Kurath G, Winton JR. Sequence motifs and prokaryotic expression of the reptilian paramyxovirus fusion protein. Arch Virol 2005; 151:449-64. [PMID: 16328138 PMCID: PMC7086783 DOI: 10.1007/s00705-005-0663-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2005] [Accepted: 09/19/2005] [Indexed: 10/25/2022]
Abstract
Fourteen reptilian paramyxovirus isolates were chosen to represent the known extent of genetic diversity among this novel group of viruses. Selected regions of the fusion (F) gene were sequenced, analyzed and compared. The F gene of all isolates contained conserved motifs homologous to those described for other members of the family Paramyxoviridae including: signal peptide, transmembrane domain, furin cleavage site, fusion peptide, N-linked glycosylation sites, and two heptad repeats, the second of which (HRB-LZ) had the characteristics of a leucine zipper. Selected regions of the fusion gene of isolate Gono-GER85 were inserted into a prokaryotic expression system to generate three recombinant protein fragments of various sizes. The longest recombinant protein was cleaved by furin into two fragments of predicted length. Western blot analysis with virus-neutralizing rabbit-antiserum against this isolate demonstrated that only the longest construct reacted with the antiserum. This construct was unique in containing 30 additional C-terminal amino acids that included most of the HRB-LZ. These results indicate that the F genes of reptilian paramyxoviruses contain highly conserved motifs typical of other members of the family and suggest that the HRB-LZ domain of the reptilian paramyxovirus F protein contains a linear antigenic epitope.
Collapse
Affiliation(s)
- J Franke
- U.S. Geological Survey, Western Fisheries Research Center, Seattle, Washington 98115, USA
| | | | | | | | | |
Collapse
|
29
|
Faisca P, Anh DBT, Desmecht DJM. Sendai virus-induced alterations in lung structure/function correlate with viral loads and reveal a wide resistance/susceptibility spectrum among mouse strains. Am J Physiol Lung Cell Mol Physiol 2005; 289:L777-87. [PMID: 16006482 DOI: 10.1152/ajplung.00240.2005] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The Paramyxoviridae family includes some of the most important and ubiquitous disease-causing viruses of infants and children, most of which cause significant infections of the respiratory tract. Evidence is accumulating in humans that genetic factors are involved in the severity of clinical presentation. As a first step toward the identification of the genes involved, this study was undertaken to establish whether laboratory mouse strains differ in susceptibility to Sendai virus, the murine counterpart of human type-1 parainfluenza virus which, historically, has been used extensively in studies that have defined the basic biological properties of paramyxoviruses in general. With this purpose in mind, double-chamber plethysmography data were collected daily for 7 days after inoculation of Sendai virus in six inbred strains of mice. In parallel, histological examinations and lung viral titration were carried out from day 5 to day 7 after inoculation. Pulmonary structure/function values closely reflected the success of viral replication in the lungs and revealed a pattern of continuous variation with resistant, intermediate, and susceptible strains. The results unambiguously suggest that BALB/c (resistant) and 129Sv (susceptible) strains should be used in crossing experiments aimed at identifying the genes involved in resistance to Paramyxoviridae by the positional cloning approach.
Collapse
Affiliation(s)
- Pedro Faisca
- Department of Pathology, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | | | | |
Collapse
|
30
|
Zimmer G, Bossow S, Kolesnikova L, Hinz M, Neubert WJ, Herrler G. A chimeric respiratory syncytial virus fusion protein functionally replaces the F and HN glycoproteins in recombinant Sendai virus. J Virol 2005; 79:10467-77. [PMID: 16051839 PMCID: PMC1182616 DOI: 10.1128/jvi.79.16.10467-10477.2005] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Entry of most paramyxoviruses is accomplished by separate attachment and fusion proteins that function in a cooperative manner. Because of this close interdependence, it was not possible with most paramyxoviruses to replace either of the two protagonists by envelope glycoproteins from related paramyxoviruses. By using reverse genetics of Sendai virus (SeV), we demonstrate that chimeric respiratory syncytial virus (RSV) fusion proteins containing either the cytoplasmic domain of the SeV fusion protein or in addition the transmembrane domain were efficiently incorporated into SeV particles provided the homotypic SeV-F was deleted. In the presence of SeV-F, the chimeric glycoproteins were incorporated with significantly lower efficiency, indicating that determinants in the SeV-F ectodomain exist that contribute to glycoprotein uptake. Recombinant SeV in which the homotypic fusion protein was replaced with chimeric RSV fusion protein replicated in a trypsin-independent manner and was neutralized by antibodies directed to RSV-F. However, replication of this virus also relied on the hemagglutinin-neuraminidase (HN) as pretreatment of cells with neuraminidase significantly reduced the infection rate. Finally, recombinant SeV was generated with chimeric RSV-F as the only envelope glycoprotein. This virus was not neutralized by antibodies to SeV and did not use sialic acids for attachment. It replicated more slowly than hybrid virus containing HN and produced lower virus titers. Thus, on the one hand RSV-F can mediate infection in an autonomous way while on the other hand it accepts support by a heterologous attachment protein.
Collapse
Affiliation(s)
- Gert Zimmer
- Institut für Virologie, Tierärztliche Hochschule Hannover, Bünteweg 17, D-30559 Hannover, Germany.
| | | | | | | | | | | |
Collapse
|
31
|
Schickli JH, Kaur J, Ulbrandt N, Spaete RR, Tang RS. An S101P substitution in the putative cleavage motif of the human metapneumovirus fusion protein is a major determinant for trypsin-independent growth in vero cells and does not alter tissue tropism in hamsters. J Virol 2005; 79:10678-89. [PMID: 16051860 PMCID: PMC1182652 DOI: 10.1128/jvi.79.16.10678-10689.2005] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human metapneumovirus (hMPV), a recently described paramyxovirus, is a major etiological agent for lower respiratory tract disease in young children that can manifest with severe cough, bronchiolitis, and pneumonia. The hMPV fusion glycoprotein (F) shares conserved functional domains with other paramyxovirus F proteins that are important for virus entry and spread. For other paramyxovirus F proteins, cleavage of a precursor protein (F0) into F1 and F2 exposes a fusion peptide at the N terminus of the F1 fragment, a likely prerequisite for fusion activity. Many hMPV strains have been reported to require trypsin for growth in tissue culture. The majority of these strains contain RQSR at the putative cleavage site. However, strains hMPV/NL/1/00 and hMPV/NL/1/99 expanded in our laboratory contain the sequence RQPR and do not require trypsin for growth in Vero cells. The contribution of this single amino acid change was verified directly by generating recombinant virus (rhMPV/NL/1/00) with either proline or serine at position 101 in F. These results suggested that cleavage of F protein in Vero cells could be achieved by trypsin or S101P amino acid substitution in the putative cleavage site motif. Moreover, trypsin-independent cleavage of hMPV F containing 101P was enhanced by the amino acid substitution E93K. In hamsters, rhMPV/93K/101S and rhMPV/93K/101P grew to equivalent titers in the respiratory tract and replication was restricted to respiratory tissues. The ability of these hMPV strains to replicate efficiently in the absence of trypsin should greatly facilitate the generation, preclinical testing, and manufacturing of attenuated hMPV vaccine candidates.
Collapse
Affiliation(s)
- Jeanne H Schickli
- MedImmune Vaccines, Inc., 297 N. Bernardo Ave., Mountain View, CA 94043, USA.
| | | | | | | | | |
Collapse
|
32
|
Springfeld C, von Messling V, Tidona CA, Darai G, Cattaneo R. Envelope targeting: hemagglutinin attachment specificity rather than fusion protein cleavage-activation restricts Tupaia paramyxovirus tropism. J Virol 2005; 79:10155-63. [PMID: 16051808 PMCID: PMC1182650 DOI: 10.1128/jvi.79.16.10155-10163.2005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To engineer a targeting envelope for gene and oncolytic vector delivery, we characterized and modified the envelope proteins of Tupaia paramyxovirus (TPMV), a relative of the morbilli- and henipaviruses that neither infects humans nor has cross-reactive relatives that infect humans. We completed the TPMV genomic sequence and noted that the predicted fusion (F) protein cleavage-activation site is not preceded by a canonical furin cleavage sequence. Coexpression of the TPMV F and hemagglutinin (H) proteins induced fusion of Tupaia baby fibroblasts but not of human cells, a finding consistent with the restricted TPMV host range. To identify the factors restricting fusion of non-Tupaia cells, we initially analyzed F protein cleavage. Even without an oligo- or monobasic protease cleavage sequence, TPMV F was cleaved in F1 and F2 subunits in human cells. Edman degradation of the F1 subunit yielded the sequence IFWGAIIA, placing the conserved phenylalanine in position 2, a novelty for paramyxoviruses but not the cause of fusion restriction. We then verified whether the lack of a TPMV H receptor limits fusion. Toward this end, we displayed a single-chain antibody (scFv) specific for the designated receptor human carcinoembryonic antigen on the TPMV H ectodomain. The H-scFv hybrid protein coexpressed with TPMV F mediated fusion of cells expressing the designated receptor, proving that the lack of a receptor limits fusion and that TPMV H can be retargeted. Targeting competence and the absence of antibodies in humans define the TPMV envelope as a module to be adapted for ferrying ribonucleocapsids of oncolytic viruses and gene delivery vectors.
Collapse
Affiliation(s)
- Christoph Springfeld
- Mayo Clinic Rochester, Molecular Medicine Program, Guggenheim 1838, 200 First St. SW, Rochester, MN 55902, USA
| | | | | | | | | |
Collapse
|
33
|
Diederich S, Moll M, Klenk HD, Maisner A. The nipah virus fusion protein is cleaved within the endosomal compartment. J Biol Chem 2005; 280:29899-903. [PMID: 15961384 DOI: 10.1074/jbc.m504598200] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Nipah virus (NiV) is a recently emerged and highly pathogenic paramyxovirus that causes a systemic infection in animals and humans and can infect a wide range of cultured cells. Interestingly, the NiV fusion (F) protein has a single arginine at the cleavage site similar to paramyxoviruses that are activated by exogenous trypsin-like enzymes only present in specific cells and tissues and therefore only cause localized infections. We show here that NiV F activation is not mediated by an exogenous serum protease but by an endogenous ubiquitous cellular protease after endocytosis of the protein. In addition to endocytosis, acidification of the endosome is a prerequisite for F cleavage. These results show that activation of the NiV F protein depends on a type of proteolytic cleavage that is clearly different from what is known for other paramyxoviral and orthomyxoviral fusion proteins. To our knowledge, this is the first example of a viral class I fusion protein whose activation depends on clathrin-mediated constitutive endocytosis.
Collapse
|
34
|
Hou X, Suquilanda E, Zeledon A, Kacsinta A, Moore A, Seto J, McQueen N. Mutations in Sendai virus variant F1-R that correlate with plaque formation in the absence of trypsin. Med Microbiol Immunol 2005; 194:129-36. [PMID: 15834752 PMCID: PMC7086596 DOI: 10.1007/s00430-004-0224-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2004] [Indexed: 11/11/2022]
Abstract
With the emergence of new viruses, such as the SARS virus and the avian influenza virus, the importance of investigations on the genetic basis of viral infections becomes clear. Sendai virus causes a localized respiratory tract infection in rodents, while a mutant, F1-R, causes a systemic infection. It has been suggested that two determinants are responsible for the systemic infection caused by F1-R [Okada et al (1998) Arch Virol 143:2343-2352]. The primary determinant of the pantropism is the enhanced proteolytic cleavability of the fusion (F) protein of F1-R, which allows the virus to undergo multiple rounds of replication in many different organs, whereas wild-type virus can only undergo multiple rounds of replication in the lungs. The enhanced cleavability of F1-R F was previously attributed to an amino acid change at F115 that is adjacent to the cleavage site at amino acid 116. Secondly, wild-type virus buds only from the apical domain of bronchial epithelium, releasing virus into the lumen of the respiratory tract, whereas F1-R buds from both apical and basolateral domains. Thus, virus is released into the basement membrane where it can easily gain access to the bloodstream for dissemination. The microtubule disruption is attributed to two amino acid differences in M protein. To confirm that the F and M gene mutations described above are solely responsible for the phenotypic differences seen in wild-type versus F1-R infections, reverse genetics was used to construct recombinant Sendai viruses with various combinations of the mutations found in the M and F genes of F1-R. Plaque assays were performed with or without trypsin addition. A recombinant virus containing all F1-R M and F mutations formed plaques in LLC-MK2 cells and underwent multiple cycles of replication without trypsin addition. To clarify which mutation(s) are necessary for plaque formation, plaque assays were done using other recombinant viruses. A virus with only the F115 change, which was previously thought to be the only change important for plaque formation of F 1-R F, did not confer upon the virus the ability to form plaques without the addition of trypsin. Another virus with the F115 and both M changes gave the same result. Therefore, more than one mutation in the F gene contributes to the ability of F1-R to form plaques without trypsin addition.
Collapse
Affiliation(s)
- Xiaogang Hou
- Department of Biological Sciences, California State University, Los Angeles, 5151 State University Drive, Los Angeles, CA 90032 USA
| | - Edgar Suquilanda
- Department of Biological Sciences, California State University, Los Angeles, 5151 State University Drive, Los Angeles, CA 90032 USA
| | - Ana Zeledon
- Department of Biological Sciences, California State University, Los Angeles, 5151 State University Drive, Los Angeles, CA 90032 USA
| | - Apollo Kacsinta
- Department of Biological Sciences, California State University, Los Angeles, 5151 State University Drive, Los Angeles, CA 90032 USA
| | - Akila Moore
- Department of Biological Sciences, California State University, Los Angeles, 5151 State University Drive, Los Angeles, CA 90032 USA
| | - Joseph Seto
- Department of Biological Sciences, California State University, Los Angeles, 5151 State University Drive, Los Angeles, CA 90032 USA
| | - Nancy McQueen
- Department of Biological Sciences, California State University, Los Angeles, 5151 State University Drive, Los Angeles, CA 90032 USA
| |
Collapse
|
35
|
Miyamae T. Differential Invasion by Sendai Virus of Abdominal Parenchymal Organs and Brain Tissues in Cortisone- and Cyclophosphamide-Based Immunosuppressed Mice. J Vet Med Sci 2005; 67:369-77. [PMID: 15876786 DOI: 10.1292/jvms.67.369] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Pneumotropic virus invasion of parenchymal organs in mice with immunosuppressants is worth studying from an etiopathogenetic viewpoint as an animal model of a compromised host. This study analyzed the invasion by Sendai virus (SeV) of mouse organs in immunosuppressive states induced by cortisone (CO) and cyclophosphamide (CY). After subcutaneous inoculation of CO or CY, or both, in mice infected intra-nasally with SeV, the SeV invasion was investigated by detecting viremia and viral antigen in organs. SeV Nagoya strain and one treatment of CO or CY caused viremia at 18 hr or 3 d, respectively, with infection in tracheal lymph nodes, but not in untreated mice. SeV invaded hepatocytes and splenolymphocytes on days 10 and 5-10, respectively, after infection. CO or CY treatment, three times, induced viral invasion of brain tissues or serious respiratory infection, respectively, but progressive invasion of abdominal parenchymal organs was not different between treated groups. One CO and two CY treatments intensified viral invasion into many organs, maintaining seronegativity. Cerebral blood vessels had the highest incidence of viral antigen in the brain. Astroglial sheets, choroid plexuses, pia maters, and ventricular epithelia tested positive; test-positive neurons were few. SeV MN strain caused progressive invasion of the brain with gliosis and neuronophagy. Blood-brain barrier disruption was caused by virulence of the MN strain. Half the infected mice in two groups treated with CO once and CY twice succumbed to delayed hypersensitivity, suggested by cerebro-microvascular nodulation.
Collapse
Affiliation(s)
- Takeo Miyamae
- School of Health Sciences, Fujita Health University, Toyoake, Aichi, Japan
| |
Collapse
|
36
|
Moll M, Diederich S, Klenk HD, Czub M, Maisner A. Ubiquitous activation of the Nipah virus fusion protein does not require a basic amino acid at the cleavage site. J Virol 2004; 78:9705-12. [PMID: 15331703 PMCID: PMC514977 DOI: 10.1128/jvi.78.18.9705-9712.2004] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Nipah virus (NiV), a highly pathogenic paramyxovirus, causes a systemic infection in vivo and is able to replicate in cultured cells of many species and organs. Such pantropic paramyxoviruses generally encode fusion (F) proteins with multibasic cleavage sites activated by furin or other ubiquitous intracellular host cell proteases. In contrast, NiV has an F protein with a single arginine (R109) at the cleavage site, as is the case with paramyxoviruses that are activated by trypsin-like proteases only present in specific cells or tissues and therefore only cause localized infections. Unlike these viruses, cleavage of the NiV F protein is ubiquitous and does not require the addition of exogenous proteases in cell culture. To determine the importance of the amino acid sequence at the NiV F protein cleavage site for ubiquitous activation, we generated NiV F proteins with mutations around R109. Surprisingly, neither the exchange of amino acids upstream of R109 nor replacement of the basic residue itself interfered with F cleavage. Thus, R109 is not essential for F cleavage and activation. Our data demonstrate that NiV F-protein activation depends on a novel type of proteolytic cleavage that has not yet been described for any other paramyxovirus F protein. NiV F activation is mediated by a ubiquitous protease that requires neither a monobasic nor a multibasic cleavage site and therefore differs from the furin- or trypsin-like proteases known to activate other ortho- and paramyxovirus fusion proteins.
Collapse
Affiliation(s)
- Markus Moll
- Institut für Virologie, Robert-Koch-Str. 17, 35037 Marburg, Germany
| | | | | | | | | |
Collapse
|
37
|
Sato M, Yoshida S, Iida K, Tomozawa T, Kido H, Yamashita M. A novel influenza A virus activating enzyme from porcine lung: purification and characterization. Biol Chem 2003; 384:219-27. [PMID: 12675514 DOI: 10.1515/bc.2003.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Proteolytic activation of hemagglutinin, an envelope glycoprotein of the influenza virus, by host proteases is essential for infection and proliferation of the virus. However, there is no well-defined, inherent source of host proteases in man or swine, both of which are natural hosts for human influenza viruses. We have recently isolated a 32 kDa protein in a high salt extract from porcine lungs, which possess the hemagglutinin processing activity. In this study, we attempted to purify another hemagglutinin processing enzyme from porcine lung. The purified enzyme, named tryptase TC30, exhibited a molecular mass of about 30 kDa by SDS-PAGE and 28.5 kDa by gel filtration chromatography, suggesting that it is a monomer. Tryptase TC30 cleaved peptide substrates with Arg at the P1 position, and preferentially substrates with the Ser-Ile-Gin-Ser-Arg sequence corresponding to the HA cleavage site sequence of the A/PR/8/34 influenza virus. Among various inhibitors tested, trypsin-type serine protease inhibitors, such as aprotinin, antipain, benzamidine and leupeptin, efficiently inhibited the proteolytic activity of the enzyme. The N-terminal 40 amino acid sequence of tryptase TC30 exhibits more than 60% homology to mast cell tryptases from mice MCP-6 and human tryptase-alpha and -beta. These data indicate that tryptase TC30, the 30 kDa enzyme from porcine lung, is a novel hemagglutinin-cleaving enzyme.
Collapse
Affiliation(s)
- Masayuki Sato
- Biological Research Laboratories, Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | | | | | | | | | | |
Collapse
|
38
|
Towatari T, Ide M, Ohba K, Chiba Y, Murakami M, Shiota M, Kawachi M, Yamada H, Kido H. Identification of ectopic anionic trypsin I in rat lungs potentiating pneumotropic virus infectivity and increased enzyme level after virus infection. EUROPEAN JOURNAL OF BIOCHEMISTRY 2002; 269:2613-21. [PMID: 12027901 DOI: 10.1046/j.1432-1033.2002.02937.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Extracellular cleavage of virus envelope fusion glycoproteins by host cellular proteases is a prerequisite for the infectivity of mammalian and nonpathogenic avian influenza viruses, and Sendai virus. In search of such target processing proteases in the airway, we recently found a new candidate trypsin-like processing protease in rat lungs, which was induced by Sendai virus infection, and identified as ectopic rat anionic trypsin I. On SDS/PAGE under reducing and nonreducing conditions, the purified enzyme gave protein bands corresponding to 29 and 22 kDa, respectively, i.e. at the same positions as rat pancreatic anionic trypsin I. It exhibited an apparent molecular mass of 31 kDa on molecular sieve chromatography and its isoelectric point was pH 4.7. The amino-acid sequences of the N-terminus and proteolytic digest peptides of the purified enzyme were consistent with those of rat pancreatic anionic trypsin I. Its substrate specificities and inhibitor sensitivities were the same as those of the pancreatic enzyme. The purified enzyme efficiently processed the fusion glycoprotein precursor of Sendai virus and hemagglutinin of human influenza A virus, and potentiated the infectivity of Sendai virus in the same dose-dependent manner as the pancreatic one. Immunohistochemical studies revealed that this protease is located in the stromal cells in peri-bronchiolar regions. These results suggest that ectopic anionic trypsin I in rat lungs induced by virus infection may trigger virus spread in rat lungs.
Collapse
Affiliation(s)
- Takae Towatari
- Division of Enzyme Chemistry, Institute for Enzyme Research, University of Tokushima, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Murakami M, Towatari T, Ohuchi M, Shiota M, Akao M, Okumura Y, Parry MA, Kido H. Mini-plasmin found in the epithelial cells of bronchioles triggers infection by broad-spectrum influenza A viruses and Sendai virus. ACTA ACUST UNITED AC 2001; 268:2847-55. [PMID: 11358500 DOI: 10.1046/j.1432-1327.2001.02166.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Extracellular cleavage of virus envelope fusion glycoproteins by host cellular proteases is a prerequisite for the infectivity of mammalian and nonpathogenic avian influenza viruses, and Sendai virus. Here we report a protease present in the airway that, like tryptase Clara, can process influenza A virus haemagglutinin and Sendai virus envelope fusion glycoprotein. This protease was extracted from the membrane fraction of rat lungs, purified and then identified as a mini-plasmin. Mini-plasmin was distributed predominantly in the epithelial cells of the upward divisions of bronchioles and potentiated the replication of broad-spectrum influenza A viruses and Sendai virus, even that of the plasmin-insensitive influenza A virus strain. In comparison with plasmin, its increased hydrophobicity, leading to its higher local concentrations on membranes, and decreased molecular mass may enable mini-plasmin to gain ready access to the cleavage sites of various haemagglutinins and fusion glycoproteins after expression of these viral proteins on the cell surface. These findings suggest that mini-plasmin in the airway may play a pivotal role in the spread of viruses and their pathogenicity.
Collapse
Affiliation(s)
- M Murakami
- Division of Enzyme Chemistry, Institute for Enzyme Research, University of Tokushima, Japan
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Michalski WP, Crameri G, Wang L, Shiell BJ, Eaton B. The cleavage activation and sites of glycosylation in the fusion protein of Hendra virus. Virus Res 2000; 69:83-93. [PMID: 11018278 DOI: 10.1016/s0168-1702(00)00169-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Hendra virus (HeV) is an unclassified member of the Paramyxoviridae family that causes systemic infections in humans, horses, cats, guinea pigs and flying foxes. The fusion protein (F(0)) of members of the Paramyxoviridae family that cause systemic infections in vivo contains a basic amino acid-rich region at which the protein is activated by cleavage into two subunits (F(1) and F(2)). HeV F(0) lacks such a domain. We have determined the cleavage site in HeV F(0) by sequencing the amino terminus of the F(1) subunit and in view of the potential effect of glycosylation on the cleavage process have ascertained the sites at which F(0) is glycosylated. The results indicate that unlike other members of the family that replicate in cultured cells and cause systemic infections in vivo, cleavage of HeV F(0) occurs at a single lysine (reside 109) in the sequence Asp-Val-Lys- downward arrow-Leu. Although HeV genotypically resembles members of the Respirovirus and Rubulavirus genera in having potential N-linked glycosylation sites in both the F(1) and F(2) subunits, we show that phenotypically HeV may more closely resemble members of the Morbillivirus genus that contain N-linked glycans only in the F(2) subunit.
Collapse
Affiliation(s)
- W P Michalski
- CSIRO Animal Health, Australian Animal Health Laboratory, Private Bag 24, Vic. 3220, Geelong, Australia.
| | | | | | | | | |
Collapse
|
41
|
Chen Y, Shiota M, Ohuchi M, Towatari T, Tashiro J, Murakami M, Yano M, Yang B, Kido H. Mast cell tryptase from pig lungs triggers infection by pneumotropic Sendai and influenza A viruses. Purification and characterization. EUROPEAN JOURNAL OF BIOCHEMISTRY 2000; 267:3189-97. [PMID: 10824103 DOI: 10.1046/j.1432-1327.2000.01346.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A novel trypsin-type serine proteinase, which processes the precursors of the envelope fusion glycoproteins of pneumotropic Sendai and human influenza A viruses, was purified to homogeneity from pig lungs. On SDS/PAGE, the purified enzyme gave a protein band corresponding to about 32 kDa, and has an apparent molecular mass of 120 kDa, as determined by gel permeation chromatography. Immunohistochemical staining with antibodies against this enzyme revealed that the enzyme is located in pig lung mast cells. The N-terminal 44-amino-acid sequence of the enzyme exhibits about 80% identity with those of mast cell tryptases from other species. Of the inhibitors tested, di-isopropyl fluorophosphate, antipain, leupeptin, benzamidine and a few proteinaceous inhibitors, such as mucus protease inhibitor and aprotinin, inhibited this enzyme activity. Heparin stabilized the enzyme, but high-ionic-strength conditions did not, unlike for human mast cell tryptase. The purified enzyme efficiently processed the fusion glycoprotein precursor of Sendai virus and slowly processed hemagglutinin of human influenza A virus, and triggered the infectivity of Sendai virus in a dose-dependent manner, although human mast cell tryptase beta and rat mast cell tryptase (rat MCP-7) from lungs did not process these fusion glycoproteins at all. These results suggest that mast cell tryptase in pig lungs is the possible trigger of the pneumotropic virus infections.
Collapse
Affiliation(s)
- Y Chen
- Division of Enzyme Chemistry, Institute for Enzyme Research, University of Tokushima, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Maisner A, Mrkic B, Herrler G, Moll M, Billeter MA, Cattaneo R, Klenk HD. Recombinant measles virus requiring an exogenous protease for activation of infectivity. J Gen Virol 2000; 81:441-9. [PMID: 10644843 DOI: 10.1099/0022-1317-81-2-441] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Proteolytic cleavage of the fusion protein (F) is an important control mechanism of the biological activity of paramyxoviruses. The sequence R-R-H-K-R(112) at the cleavage site of the F protein of measles virus (MV) was altered by site-directed mutagenesis to R-N-H-N-R(112), which is not recognized by the ubiquitous cellular protease furin. When transiently expressed in cell cultures standard F protein was cleaved, whereas the mutant remained in the uncleaved form. Syncytium formation by the mutant that was analysed after coexpression with haemagglutinin protein depended on the presence of trypsin. Recombinant MV containing the mutation required trypsin activation for fusion and infectivity in cell culture. Intranasal infection of transgenic mice susceptible to MV infection (Ifnar(tm)-CD46Ge) resulted in a moderately productive infection and inflammation of the lung. In contrast to parental virus, intracerebral inoculation did not induce neural disease. The possible effects of the change in cleavage activation on tissue tropism and pathogenicity are discussed.
Collapse
Affiliation(s)
- A Maisner
- Institut für Virologie, Philipps-Universität Marburg, Robert-Koch-Str. 17, 35037 Marburg, Germany.
| | | | | | | | | | | | | |
Collapse
|
43
|
Ludwig S, Pleschka S, Wolff T. A fatal relationship--influenza virus interactions with the host cell. Viral Immunol 1999; 12:175-96. [PMID: 10532647 DOI: 10.1089/vim.1999.12.175] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Influenza A viruses are important worldwide pathogens for humans and different animal species. The infectious agent is the prototype of the orthomyxoviridae which are characterized by a segmented negative strand RNA genome that is replicated in the nucleus of the infected cell. The genome has a combined coding capacity of about 13 kb and contains the genetic information for ten viral proteins. Despite this relatively small coding capacity--large DNA viruses like herpes or poxviruses express about 150-200 gene products--influenza A viruses are able to successfully infect and multiply in a wide range of mammalian and avian species. It is therefore not surprising that influenza A viruses extensively use and manipulate host cell functions. This includes multiple interactions of viral proteins with cellular proteins. In recent years an increasing amount of information about the identity of the cellular factors that are involved in viral transcription and replication, intracellular trafficking of viral components and assembly of the virus particle has accumulated. This article aims to review recent developments in this field with a focus on cellular factors and processes which are activated by the virus to either support viral replication or to counteract host-cell defense mechanisms.
Collapse
Affiliation(s)
- S Ludwig
- Institut für Medizinische Strahlenkunde und Zellforschung, Julius-Maximilians Universität, Würzburg, Germany.
| | | | | |
Collapse
|
44
|
Yamaoka K, Masuda K, Ogawa H, Takagi K, Umemoto N, Yasuoka S. Cloning and characterization of the cDNA for human airway trypsin-like protease. J Biol Chem 1998; 273:11895-901. [PMID: 9565616 DOI: 10.1074/jbc.273.19.11895] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Previously we isolated a trypsin-like enzyme designated human airway trypsin-like protease from the sputum of patients with chronic airway diseases. This paper describes the cDNA cloning, characterization of the primary protein structure deduced from the cDNA, and gene expression of this enzyme in various human tissues. We obtained an entire 1517-base pair sequence of cDNA with an open reading frame encoding a polypeptide with 418-amino acid residues. The polypeptide consisted of a 232-residue catalytic region and a 186-residue noncatalytic region with a hydrophobic putative transmembrane domain near the NH2 terminus. The polypeptide was suggested to be a type II integral membrane protein in which the COOH-terminal catalytic region is extracellular. Therefore, this protein is thought to be synthesized as a membrane-bound precursor and to mature to a soluble and active protease by limited proteolysis. It showed 29-38% identity in the sequence of the catalytic region with human hepsin, enteropeptidase, acrosin, and mast cell tryptase. The noncatalytic region had little similarity to other known proteins. In Northern blot analysis a transcript of 1.9 kilobases was detectable most prominently in the trachea among 17 human tissues examined.
Collapse
Affiliation(s)
- K Yamaoka
- Teijin Institute for Biomedical Research, 4-3-2 Asahigaoka, Hino, Tokyo 191, Japan
| | | | | | | | | | | |
Collapse
|
45
|
Kido H, Beppu Y, Sakai K, Towatari T. Molecular basis of proteolytic activation of Sendai virus infection and the defensive compounds for infection. Biol Chem 1997; 378:255-63. [PMID: 9165079 DOI: 10.1515/bchm.1997.378.3-4.255] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
It has been proposed that the pathogenicity of Sendai virus is primarily determined by a host cellular protease(s) that activates viral infectivity by proteolytic cleavage of envelope fusion glycoproteins. We isolated a trypsin-like serine protease, tryptase Clara, localized in and secreted from Clara cells of the bronchial epithelium of rats. The enzyme specifically cleaved the precursor of fusion glycoprotein F0 of Sendai virus at residue Arg116 in the consensus cleavage motif, Gln(Glu)-X-Arg, resulting in the presentation of the membrane fusion domain in the amino-terminus of the F1 subunit. Administration of an antibody against tryptase Clara in the airway significantly inhibited the activation of progeny virus and multiple cycles of viral replication, thus reducing the mortality rate. These findings indicate that tryptase Clara in the airway is a primary determinant of Sendai virus infection and that proteolytic activation occurs extracellularly. We identified two cellular inhibitory compounds against tryptase Clara in bronchial lavage. One was a mucus protease inhibitor, a major serine protease inhibitor of granulocyte elastase in the lining fluids of the human respiratory tract, and the other was a pulmonary surfactant which may adsorb the enzyme, resulting in its inactivation. These compounds inhibited virus activation by tryptase Clara in vitro and in vivo, but did not themselves affect the hemagglutination and the infectivity of the virus. The functional domain of the mucus protease inhibitor against the enzyme, which is organized in two homologous N- and C-terminal domains, is located in the C-terminal. Administration of these compounds in the airway may be useful for preventing infection with Sendai virus.
Collapse
Affiliation(s)
- H Kido
- Division of Enzyme Chemistry, Institute for Enzyme Research, The University of Tokushima, kuramoto-cho, Japan
| | | | | | | |
Collapse
|
46
|
Tashiro M, McQueen NL, Seto JT, Klenk HD, Rott R. Involvement of the mutated M protein in altered budding polarity of a pantropic mutant, F1-R, of Sendai virus. J Virol 1996; 70:5990-7. [PMID: 8709221 PMCID: PMC190619 DOI: 10.1128/jvi.70.9.5990-5997.1996] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Wild-type Sendai virus buds at the apical plasma membrane domain of polarized epithelial MDCK cells, whereas a pantropic mutant, F1-R, buds at both the apical and basolateral domains. In F1-R-infected cells, polarized protein transport and the microtubule network are impaired. It has been suggested that the mutated F and/or M proteins in F1-R are responsible for these changes (M. Tashiro, J. T. Seto, H.-D. Klenk, and R. Rott, J. Virol. 67:5902-5910, 1993). To clarify which gene or mutation(s) was responsible for the microtubule disruption which leads to altered budding of F1-R, MDCK cell lines containing the M gene of either the wild type or F1-R were established. When wild-type M protein was expressed at a level corresponding to that synthesized in virus-infected cells, cellular polarity and the integrity of the microtubules were affected to some extent. On the other hand, expression of the mutated F1-R M protein resulted in the formation of giant cells about 40 times larger than normal MDCK cells. Under these conditions, the effects on the microtubule network were enhanced. The microtubules were disrupted and polarized protein transport was impaired as indicated by the nonpolarized secretion of gp80, a host cell glycoprotein normally secreted from the apical domain, and bipolar budding of wild-type and F1-R Sendai viruses. The mutated F glycoprotein of F1-R was transported bipolarly in cells expressing the F1-R M protein, whereas it was transported predominantly to the apical domain when expressed alone or in cells coexpressing the wild-type M protein. These findings indicate that the M protein of F1-R is involved in the disruption of the microtubular network, leading to impairment of cellular polarity, bipolar transport of the F glycoprotein, and bipolar budding of the virus.
Collapse
Affiliation(s)
- M Tashiro
- Department of Virology 1, National Institute of Health, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
47
|
Tashiro M, Beppu Y, Sakai K, Kido H. Inhibitory effect of pulmonary surfactant on Sendai virus infection in rat lungs. Arch Virol 1996; 141:1571-7. [PMID: 8856034 DOI: 10.1007/bf01718255] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Intranasal infection of rats with active (infectious) Sendai virus enhances secretion of tryptase Clara, a Sendai virus-activating protease, into the bronchial lumen by Clara cells of the bronchial epitheliums, and inversely suppresses secretion of pulmonary surfactant, an inhibitor of the protease, into the lumen [Kido H et al. (1993) FEBS Lett 322: 115-119]. A trypsin-resistant mutant, TR-2, showed similar effects, although its replication was restricted to a single cycle in the lungs. In contrast, neither nonactive (noninfectious) wild-type virus possessing receptor-binding activity and lacking envelope fusion activity nor UV-inactivated virus retaining receptor binding and envelope fusion activities altered the mode of secretions. These results indicate that viral replication is required for producing a condition in the bronchial lumen for proteolytic activation of progeny virus, thereby infection is extended to a fatal pneumonia. On the other hand, intranasal administration of infected rats with pulmonary surfactant suppressed activation of progeny virus and pathological changes in the lungs, suggesting a therapeutic use of pulmonary surfactant for influenza pneumonia.
Collapse
Affiliation(s)
- M Tashiro
- Department of Virology 1, National Institute of Health, Tokyo, Japan
| | | | | | | |
Collapse
|
48
|
Kido H, Towatari T, Niwa Y, Okumura Y, Beppu Y. Cellular proteases involved in the pathogenicity of human immunodeficiency and influenza viruses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1996; 389:233-40. [PMID: 8861016 DOI: 10.1007/978-1-4613-0335-0_29] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- H Kido
- Division of Enzyme Chemistry, Institute of Enzyme Research, University of Tokushima, Japan
| | | | | | | | | |
Collapse
|
49
|
Kido H, Niwa Y, Beppu Y, Towatari T. Cellular proteases involved in the pathogenicity of enveloped animal viruses, human immunodeficiency virus, influenza virus A and Sendai virus. ADVANCES IN ENZYME REGULATION 1996; 36:325-47. [PMID: 8869754 DOI: 10.1016/0065-2571(95)00016-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In enveloped viruses, post-translational proteolytic activation is a critical step for the fusion activity and thus for the infectivity of the virus. In addition to the membrane receptors for the viruses, proteolytic activation is indispensable for effective virus spread in the infected host and it is a prime determinant for pathogenicity. Here we described the host cellular processing proteases, tryptase Clara and tryptase TL2, which proteolytically activate the infectivity of influenza A and Sendai viruses in the respiratory tract and HIV-1 in human CD4+ T cells, respectively. A novel trypsin-like protease, designated tryptase Clara, was purified from rat lung. The enzyme is localized in Clara cells of the bronchial epithelium and is secreted into the airway lumen. The enzyme specifically recognizes the consensus cleavage motif Gln(Glu)-X-Arg of influenza A and Sendai viruses and proteolytically activates the envelope fusion glycoproteins of the progeny viruses extracellularly in the airway lumen. Human mucus protease inhibitor and pulmonary surfactant in airway fluid inhibited the proteolytic activation of these viruses and also suppressed multiple cycles of viral replication in vitro. These results suggest that an imbalance between the amount of tryptase Clara and that of endogenous inhibitors in airway fluid is a prime determinant for pneumopathogenicity of the viruses. Therefore supplementing an endogenous inhibitor at therapeutic doses may protect against virus infection. In HIV-1 infection, binding of the gp120 envelope glycoprotein to the CD4 receptor is not sufficient in itself to allow virus entry, and an additional component(s) in the membrane is required for cell infection as a cofactor. We isolated a serine protease named tryptase TL2, in the membrane of CD4+ lymphocytes, which specifically binds to the V3 loop of HIV-1 gp120 as a cofactor. After binding, tryptase TL2 proteolytically processed gp120 into two protein species of 70 and 50 kDa and the cleavage was suppressed by a neutralizing antibody against the V3 loop. The amino acids that constitute the cleavage sites in the V3 loop of almost all HIV isolates are variable, but they are restricted to those which are susceptible to chymotryptic and/or tryptic enzyme. The multi-substrate specificity of tryptase TL2, which has tryptic and chymotryptic specificities, may correspond tot he variability of the V3 loop. The selective cleavage of the V3 loop by tryptase TL2 may lead to a conformational change of gp120, resulting in the dissociation of gp120 from gp41, exposing the fusogenic domain of the transmembrane protein gp41 following virus-host cell fusion.
Collapse
Affiliation(s)
- H Kido
- Division of Enzyme Chemistry, University of Tokushima, Japan
| | | | | | | |
Collapse
|
50
|
Rott R, Klenk HD, Nagai Y, Tashiro M. Influenza viruses, cell enzymes, and pathogenicity. Am J Respir Crit Care Med 1995; 152:S16-9. [PMID: 7551406 DOI: 10.1164/ajrccm/152.4_pt_2.s16] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Proteolytic cleavage of the influenza virus hemagglutinin glycoprotein (HA) by cellular proteases is a prerequisite for virus infectivity, spread of the virus in the infected organism, tissue tropism, and viral pathogenicity. Production of infectious virus depends upon the structure at the HA cleavage site as well as the substrate specificity and the distribution of appropriate enzymes. Differences exist in the specificities of the endoproteases that recognize the different sequence motifs at the cleavage site. With avian influenza viruses that cause lethal systemic infections, the cleavage site consists of multibasic amino acids. Furin, which activates this type of HA, is a member of the subtilisin family and represents the prototype of ubiquitously occurring membrane-bound proteases. On the other hand, serine proteases secreted from a restricted number of cell types and some bacterial enzymes recognize a monobasic cleavage signal at HA of the mammalian and the apathogenic avian influenza viruses. The limited occurrence of these proteases results in only localized infection. Implementation of these defined conditions for virus activation may represent a novel type of disease control.
Collapse
Affiliation(s)
- R Rott
- Institut für Virologie, Justus-Liebig-Universität Giessen, Germany
| | | | | | | |
Collapse
|