1351
|
Zhang H, Wang G, Li J, Nie Y, Shi X, Lian G, Wang W, Yin X, Zhao Y, Qu X, Ding M, Deng H. Identification of an antigenic determinant on the S2 domain of the severe acute respiratory syndrome coronavirus spike glycoprotein capable of inducing neutralizing antibodies. J Virol 2004; 78:6938-45. [PMID: 15194770 PMCID: PMC421668 DOI: 10.1128/jvi.78.13.6938-6945.2004] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Severe acute respiratory syndrome (SARS) is a life-threatening disease caused by a newly identified coronavirus (CoV), SARS-CoV. The spike (S) glycoprotein of CoV is the major structural protein responsible for induction of host immune response and virus neutralization by antibodies. Hence, knowledge of neutralization determinants on the S protein is helpful for designing protective vaccines. To analyze the antigenic structure of the SARS-CoV S2 domain, the carboxyl-terminal half of the S protein, we first used sera from convalescent SARS patients to test the antigenicity of 12 overlapping fragments spanning the entire S2 and identified two antigenic determinants (Leu 803 to Ala 828 and Pro 1061 to Ser 1093). To determine whether neutralizing antibodies can be elicited by these two determinants, we immunized animals and found that both of them could induce the S2-specific antisera. In some animals, however, only one determinant (Leu 803 to Ala 828) was able to induce the antisera with the binding ability to the native S protein and the neutralizing activity to the SARS-CoV pseudovirus. This determinant is highly conserved across different SARS-CoV isolates. Identification of a conserved antigenic determinant on the S2 domain of the SARS-CoV S protein, which has the potential for inducing neutralizing antibodies, has implications in the development of effective vaccines against SARS-CoV.
Collapse
Affiliation(s)
- Hong Zhang
- Department of Cell Biology and Genetics, College of Life Sciences, Peking University, Beijing 100871, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1352
|
Brooks JE, Rainer AC, Parr RL, Woolcock P, Hoerr F, Collisson EW. Comparisons of envelope through 5B sequences of infectious bronchitis coronaviruses indicates recombination occurs in the envelope and membrane genes. Virus Res 2004; 100:191-8. [PMID: 15019237 PMCID: PMC7127682 DOI: 10.1016/j.virusres.2003.11.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2003] [Revised: 11/20/2003] [Accepted: 11/20/2003] [Indexed: 02/04/2023]
Abstract
A 1.78 kb sequence, including the E, M, 5a and 5b genes, and the intergenic region between the M and 5a genes, of six US strains of infectious bronchitis (corona)virus (IBV) were sequenced and compared to the published sequences for two additional strains. The overall identities as determined through pairwise analyses of nucleotide sequences of the entire 1.78 kb region ranged from 90 to 99%, with the 5b open reading frame (ORF) having the greatest identity (94–99%) while the identities of the E, 5a and M ORFs ranged from 87 to 100%. Nucleotide sequencing of recent field isolates from Alabama (Ala1) and California (Cal3) revealed distinct shifts in homology in the M gene, indicating two apparent recombination events between the Holland 52/Mass41-like strain and an Ark-like strain, both origins of commonly used vaccine strains. Putative sites of recombination could also be identified in both the E and M ORFs of laboratory strains of IBV.
Collapse
Affiliation(s)
- Judy Elaine Brooks
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA
| | - Aaron Cameron Rainer
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA
| | - Rebecca Lynn Parr
- Department of Biochemistry and Biophysics, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Peter Woolcock
- California Veterinary Diagnostic Laboratories System, University of California, Davis, CA 95616, USA
| | - Fred Hoerr
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Ellen Whited Collisson
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA
- Corresponding author. Tel.: +1-409-845-4122; fax: +1-409-862-1088.
| |
Collapse
|
1353
|
Zhu J, Xiao G, Xu Y, Yuan F, Zheng C, Liu Y, Yan H, Cole DK, Bell JI, Rao Z, Tien P, Gao GF. Following the rule: formation of the 6-helix bundle of the fusion core from severe acute respiratory syndrome coronavirus spike protein and identification of potent peptide inhibitors. Biochem Biophys Res Commun 2004; 319:283-8. [PMID: 15158473 PMCID: PMC7111185 DOI: 10.1016/j.bbrc.2004.04.141] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2004] [Indexed: 12/22/2022]
Abstract
Severe acute respiratory syndrome (SARS) coronavirus (SARS-CoV) is a newly identified member of Family Coronaviridae. Coronavirus envelope spike protein S is a class I viral fusion protein which is characterized by the existence of two heptad repeat regions (HR1 and HR2) (forming a complex called fusion core). Here we report that by using in vitro bio-engineering techniques, SARS-CoV HR1 and HR2 bind to each other and form a typical 6-helix bundle. The HR2, either as a synthetic peptide or as a GST-fusion polypeptide, is a potent inhibitor of virus entry. The results do show that SARS-CoV follows the general fusion mechanism of class I viruses and this lays the ground for identification of virus fusion/entry inhibitors for this devastating emerging virus.
Collapse
Affiliation(s)
- Jieqing Zhu
- Institute of Microbiology, Chinese Academy of Sciences, Beijing 100080, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1354
|
Wu Q, Xu Z, Wei T, Zeng H, Li J, Gang H, Sun M, Jiang F, Wang X, Dong W, Yang L, Wang J. Development of Taqman RT-nested PCR system for clinical SARS-CoV detection. J Virol Methods 2004; 119:17-23. [PMID: 15109816 PMCID: PMC7119531 DOI: 10.1016/j.jviromet.2004.02.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2003] [Revised: 01/29/2004] [Accepted: 02/12/2004] [Indexed: 11/26/2022]
Abstract
Severe acute respiratory syndrome (SARS) is an acute newly emerged infectious respiratory illness. The etiologic agent of SARS was named ‘SARS-associated coronavirus’ (SARS-CoV) that can be detected with reverse transcription-polymerase chain reaction (RT-PCR) assays. In this study, 12 sets of nested primers covering the SARS-CoV genome have been screened and showed sufficient sensitivity to detect SARS-CoV in RNA isolated from virus cultured in Vero 6 cells. To optimize further the reaction condition of those nested primers sets, seven sets of nested primers have been chosen to compare their reverse transcribed efficiency with specific and random primers, which is useful to combine RT with the first round of PCR into a one-step RT-PCR. Based on the sensitivity and simplicity of results, the no. 73 primer set was chosen as the candidate primer set for clinical diagnoses. To specify the amplicon to minimize false positive results, a Taqman RT-nested PCR system of no. 73 nested primer set was developed. Through investigations on a test panel of whole blood obtained from 30 SARS patients and 9 control persons, the specificity and sensitivity of the Taqman RT-nested PCR system was found to be 100 and 83%, respectively, which suggests that the method is a promising one to diagnose SARS in early stages.
Collapse
Affiliation(s)
- Qingfa Wu
- Beijing Genomics Institute, Chinese Academy of Sciences, Airport Industrial Zone, Bldg 6, Beijing 101300, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1355
|
Wu CY, Jan JT, Ma SH, Kuo CJ, Juan HF, Cheng YSE, Hsu HH, Huang HC, Wu D, Brik A, Liang FS, Liu RS, Fang JM, Chen ST, Liang PH, Wong CH. Small molecules targeting severe acute respiratory syndrome human coronavirus. Proc Natl Acad Sci U S A 2004; 101:10012-7. [PMID: 15226499 PMCID: PMC454157 DOI: 10.1073/pnas.0403596101] [Citation(s) in RCA: 366] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Severe acute respiratory syndrome (SARS) is an infectious disease caused by a novel human coronavirus. Currently, no effective antiviral agents exist against this type of virus. A cell-based assay, with SARS virus and Vero E6 cells, was developed to screen existing drugs, natural products, and synthetic compounds to identify effective anti-SARS agents. Of >10,000 agents tested, approximately 50 compounds were found active at 10 microM; among these compounds, two are existing drugs (Reserpine 13 and Aescin 5) and several are in clinical development. These 50 active compounds were tested again, and compounds 2-6, 10, and 13 showed active at 3 microM. The 50% inhibitory concentrations for the inhibition of viral replication (EC(50)) and host growth (CC(50)) were then measured and the selectivity index (SI = CC(50)/EC(50)) was determined. The EC(50), based on ELISA, and SI for Reserpine, Aescim, and Valinomycin are 3.4 microM (SI = 7.3), 6.0 microM (SI = 2.5), and 0.85 microM (SI = 80), respectively. Additional studies were carried out to further understand the mode of action of some active compounds, including ELISA, Western blot analysis, immunofluorescence and flow cytometry assays, and inhibition against the 3CL protease and viral entry. Of particular interest are the two anti-HIV agents, one as an entry blocker and the other as a 3CL protease inhibitor (K(i) = 0.6 microM).
Collapse
Affiliation(s)
- Chung-Yi Wu
- Genomics Research Center and Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1356
|
Abstract
The Severe Acute Respiratory Syndrome (SARS) is a serious respiratory illness that has recently been reported in parts of Asia and Canada. In this study, we use molecular dynamics (MD) simulations and docking techniques to screen 29 approved and experimental drugs against the theoretical model of the SARS CoV proteinase as well as the experimental structure of the transmissible gastroenteritis virus (TGEV) proteinase. Our predictions indicate that existing HIV-1 protease inhibitors, l-700,417 for instance, have high binding affinities and may provide good starting points for designing SARS CoV proteinase inhibitors.
Collapse
Affiliation(s)
| | - Ram Samudrala
- Corresponding author. Tel.: +1-206-732-6122; fax: +1-206-732-6055
| |
Collapse
|
1357
|
Qinfen Z, Jinming C, Xiaojun H, Huanying Z, Jicheng H, Ling F, Kunpeng L, Jingqiang Z. The life cycle of SARS coronavirus in Vero E6 cells. J Med Virol 2004; 73:332-7. [PMID: 15170625 PMCID: PMC7166737 DOI: 10.1002/jmv.20095] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2004] [Indexed: 12/16/2022]
Abstract
The aim of the study was to establish the life cycle of severe acute respiratory syndrome-associated coronavirus (SARS CoV) in host cells and determine the pathogenesis of SARS. Vero E6 cells (African green monkey kidney cells) were inoculated with SARS coronavirus for 3, 7, 24, 48, and 72 hr, respectively, and were observed under electron microscope. It was found that the SARS coronavirus entered the cells through membrane fusion instead of endocytosis, and then the nucleocapsids assembled in the RER and matured by budding into the smooth vesicles, which were derived from the Golgi apparatus. The smooth vesicles fused with the cell membrane, and the mature particles were released. A special phenomenon was that some virus-like particles appeared in the nucleus. We propose a scheme of the life cycle of SARS coronavirus and discuss the mechanism of its replication in Vero E6 cells.
Collapse
Affiliation(s)
- Zhang Qinfen
- State Key Lab for Biocontrol, Zhongshan University, Guangzhou, China
| | - Cui Jinming
- State Key Lab for Biocontrol, Zhongshan University, Guangzhou, China
| | - Huang Xiaojun
- State Key Lab for Biocontrol, Zhongshan University, Guangzhou, China
| | - Zheng Huanying
- Center for Disease Control and Prevention of Guangdong Province, Guangzhou, China
| | - Huang Jicheng
- Center for Disease Control and Prevention of Guangdong Province, Guangzhou, China
| | - Fang Ling
- Center for Disease Control and Prevention of Guangdong Province, Guangzhou, China
| | - Li Kunpeng
- State Key Lab for Biocontrol, Zhongshan University, Guangzhou, China
| | - Zhang Jingqiang
- State Key Lab for Biocontrol, Zhongshan University, Guangzhou, China
| |
Collapse
|
1358
|
Agranovski IE, Safatov AS, Pyankov OV, Sergeev AN, Agafonov AP, Ignatiev GM, Ryabchikova EI, Borodulin AI, Sergeev AA, Doerr HW, Rabenau HF, Agranovski V. Monitoring of viable airborne SARS virus in ambient air. ATMOSPHERIC ENVIRONMENT (OXFORD, ENGLAND : 1994) 2004; 38:3879-3884. [PMID: 32288549 PMCID: PMC7129584 DOI: 10.1016/j.atmosenv.2004.03.044] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2003] [Revised: 02/23/2004] [Accepted: 03/10/2004] [Indexed: 05/20/2023]
Abstract
Due to recent SARS related issues (Science 300 (5624) 1394; Nature 423 (2003) 240; Science 300 (5627) 1966), the development of reliable airborne virus monitoring procedures has become galvanized by an exceptional sense of urgency and is presently in a high demand (In: Cox, C.S., Wathers, C.M. (Eds.), Bioaerosols Handbook, Lewis Publishers, Boca Raton, FL, 1995, pp. 247-267). Based on engineering control method (Aerosol Science and Technology 31 (1999) 249; 35 (2001) 852), which was previously applied to the removal of particles from gas carriers, a new personal bioaerosol sampler has been developed. Contaminated air is bubbled through porous medium submerged into liquid and subsequently split into multitude of very small bubbles. The particulates are scavenged by these bubbles, and, thus, effectively removed. The current study explores its feasibility for monitoring of viable airborne SARS virus. It was found that the natural decay of such virus in the collection fluid was around 0.75 and 1.76 lg during 2 and 4 h of continuous operation, respectively. Theoretical microbial recovery rates of higher than 55 and 19% were calculated for 1 and 2 h of operation, respectively. Thus, the new sampling method of direct non-violent collection of viable airborne SARS virus into the appropriate liquid environment was found suitable for monitoring of such stress sensitive virus.
Collapse
Affiliation(s)
- Igor E Agranovski
- Faculty of Environmental Sciences, Griffith University, Brisbane, 4111 QLD, Australia
| | - Alexander S Safatov
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Novosibirsk Region 630559, Russia
| | - Oleg V Pyankov
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Novosibirsk Region 630559, Russia
| | - Alexander N Sergeev
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Novosibirsk Region 630559, Russia
| | - Alexander P Agafonov
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Novosibirsk Region 630559, Russia
| | - Georgy M Ignatiev
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Novosibirsk Region 630559, Russia
| | - Elena I Ryabchikova
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Novosibirsk Region 630559, Russia
| | - Alexander I Borodulin
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Novosibirsk Region 630559, Russia
| | - Artemii A Sergeev
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Novosibirsk Region 630559, Russia
| | - Hans W Doerr
- Institute for Medical Virology, Johann Wolfgang Goethe University, D-60596 Frankfurt/Main, Germany
| | - Holger F Rabenau
- Institute for Medical Virology, Johann Wolfgang Goethe University, D-60596 Frankfurt/Main, Germany
| | - Victoria Agranovski
- School of Physical and Chemical Sciences, Queensland University of Technology, Brisbane, 4000 QLD, Australia
| |
Collapse
|
1359
|
Tan YJ, Teng E, Shen S, Tan THP, Goh PY, Fielding BC, Ooi EE, Tan HC, Lim SG, Hong W. A novel severe acute respiratory syndrome coronavirus protein, U274, is transported to the cell surface and undergoes endocytosis. J Virol 2004; 78:6723-34. [PMID: 15194747 PMCID: PMC421683 DOI: 10.1128/jvi.78.13.6723-6734.2004] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2003] [Accepted: 02/23/2004] [Indexed: 01/28/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus (SARS-CoV) genome contains open reading frames (ORFs) that encode for several genes that are homologous to proteins found in all known coronaviruses. These are the replicase gene 1a/1b and the four structural proteins, nucleocapsid (N), spike (S), membrane (M), and envelope (E), and these proteins are expected to be essential for the replication of the virus. In addition, this genome also contains nine other potential ORFs varying in length from 39 to 274 amino acids. The largest among these is the first ORF of the second longest subgenomic RNA, and this protein (termed U274 in the present study) consists of 274 amino acids and contains three putative transmembrane domains. Using antibody specific for the C terminus of U274, we show U274 to be expressed in SARS-CoV-infected Vero E6 cells and, in addition to the full-length protein, two other processed forms were also detected. By indirect immunofluorescence, U274 was localized to the perinuclear region, as well as to the plasma membrane, in both transfected and infected cells. Using an N terminus myc-tagged U274, the topology of U274 and its expression on the cell surface were confirmed. Deletion of a cytoplasmic domain of U274, which contains Yxxphi and diacidic motifs, abolished its transport to the cell surface. In addition, U274 expressed on the cell surface can internalize antibodies from the culture medium into the cells. Coimmunoprecipitation experiments also showed that U274 could interact specifically with the M, E, and S structural proteins, as well as with U122, another protein that is unique to SARS-CoV.
Collapse
Affiliation(s)
- Yee-Joo Tan
- Institute of Molecular and Cell Biology, 30 Medical Dr., Singapore 117609, Singapore.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1360
|
Netesova NA, Belavin PA, Seregina EV, Ignat'ev GM, Sandakhchiev LS. Cloning, expression, and purification of the nucleocapsid protein of SARS coronavirus. DOKL BIOCHEM BIOPHYS 2004; 397:239-41. [PMID: 15523835 PMCID: PMC7088110 DOI: 10.1023/b:dobi.0000039473.88598.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- N A Netesova
- State Research Center for Virology and Biotechnology Vektor, pos. Kol'tsovo, Novosibirsk oblast, 630559 Russia
| | | | | | | | | |
Collapse
|
1361
|
Affiliation(s)
- James D Cherry
- David Geffen School of Medicine at UCLA and Mattel's Children's Hospital at UCLA, Department of Pediatrics, Los Angeles, CA 90095-1752, USA.
| | | |
Collapse
|
1362
|
Hofmann H, Hattermann K, Marzi A, Gramberg T, Geier M, Krumbiegel M, Kuate S, Uberla K, Niedrig M, Pöhlmann S. S protein of severe acute respiratory syndrome-associated coronavirus mediates entry into hepatoma cell lines and is targeted by neutralizing antibodies in infected patients. J Virol 2004; 78:6134-42. [PMID: 15163706 PMCID: PMC416513 DOI: 10.1128/jvi.78.12.6134-6142.2004] [Citation(s) in RCA: 161] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The severe acute respiratory syndrome-associated coronavirus (SARS-CoV) causes severe pneumonia with a fatal outcome in approximately 10% of patients. SARS-CoV is not closely related to other coronaviruses but shares a similar genome organization. Entry of coronaviruses into target cells is mediated by the viral S protein. We functionally analyzed SARS-CoV S using pseudotyped lentiviral particles (pseudotypes). The SARS-CoV S protein was found to be expressed at the cell surface upon transient transfection. Coexpression of SARS-CoV S with human immunodeficiency virus-based reporter constructs yielded viruses that were infectious for a range of cell lines. Most notably, viral pseudotypes harboring SARS-CoV S infected hepatoma cell lines but not T- and B-cell lines. Infection of the hepatoma cell line Huh-7 was also observed with replication-competent SARS-CoV, indicating that hepatocytes might be targeted by SARS-CoV in vivo. Inhibition of vacuolar acidification impaired infection by SARS-CoV S-bearing pseudotypes, indicating that S-mediated entry requires low pH. Finally, infection by SARS-CoV S pseudotypes but not by vesicular stomatitis virus G pseudotypes was efficiently inhibited by a rabbit serum raised against SARS-CoV particles and by sera from SARS patients, demonstrating that SARS-CoV S is a target for neutralizing antibodies and that such antibodies are generated in SARS-CoV-infected patients. Our results show that viral pseudotyping can be employed for the analysis of SARS-CoV S function. Moreover, we provide evidence that SARS-CoV infection might not be limited to lung tissue and can be inhibited by the humoral immune response in infected patients.
Collapse
Affiliation(s)
- Heike Hofmann
- Institute for Clinical and Molecular Virology, University Erlangen-Nürnberg, Nikolaus-Fiebiger-Center, Glückstrasse 6, D-91054 Erlangen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1363
|
Buchholz UJ, Bukreyev A, Yang L, Lamirande EW, Murphy BR, Subbarao K, Collins PL. Contributions of the structural proteins of severe acute respiratory syndrome coronavirus to protective immunity. Proc Natl Acad Sci U S A 2004; 101:9804-9. [PMID: 15210961 PMCID: PMC470755 DOI: 10.1073/pnas.0403492101] [Citation(s) in RCA: 321] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2004] [Indexed: 01/12/2023] Open
Abstract
We investigated the contributions of the structural proteins of severe acute respiratory syndrome (SARS) coronavirus (CoV) to protective immunity by expressing them individually and in combinations from a recombinant parainfluenza virus (PIV) type 3 vector called BHPIV3. This vector provided direct immunization of the respiratory tract, the major site of SARS transmission, replication, and disease. The BHPIV3/SARS recombinants were evaluated for immunogenicity and protective efficacy in hamsters, which support a high level of pulmonary SARS-CoV replication. A single intranasal administration of BHPIV3 expressing the SARS-CoV spike protein (S) induced a high titer of SARS-CoV-neutralizing serum antibodies, only 2-fold less than that induced by SARS-CoV infection. The expression of S with the two other putative virion envelope proteins, the matrix M and small envelope E proteins, did not augment the neutralizing antibody response. In absence of S, expression of M and E or the nucleocapsid protein N did not induce a detectable serum SARS-CoV-neutralizing antibody response. Immunization with BHPIV3 expressing S provided complete protection against SARS-CoV challenge in the lower respiratory tract and partial protection in the upper respiratory tract. This was augmented slightly by coexpression with M and E. Expression of M, E, or N in the absence of S did not confer detectable protection. These results identify S among the structural proteins as the only significant SARS-CoV neutralization antigen and protective antigen and show that a single mucosal immunization is highly protective in an experimental animal that supports efficient replication of SARS-CoV.
Collapse
Affiliation(s)
- Ursula J Buchholz
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-8007, USA.
| | | | | | | | | | | | | |
Collapse
|
1364
|
Zhao Z, Li H, Wu X, Zhong Y, Zhang K, Zhang YP, Boerwinkle E, Fu YX. Moderate mutation rate in the SARS coronavirus genome and its implications. BMC Evol Biol 2004; 4:21. [PMID: 15222897 PMCID: PMC446188 DOI: 10.1186/1471-2148-4-21] [Citation(s) in RCA: 162] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2004] [Accepted: 06/28/2004] [Indexed: 11/10/2022] Open
Abstract
Background The outbreak of severe acute respiratory syndrome (SARS) caused a severe global epidemic in 2003 which led to hundreds of deaths and many thousands of hospitalizations. The virus causing SARS was identified as a novel coronavirus (SARS-CoV) and multiple genomic sequences have been revealed since mid-April, 2003. After a quiet summer and fall in 2003, the newly emerged SARS cases in Asia, particularly the latest cases in China, are reinforcing a wide-spread belief that the SARS epidemic would strike back. With the understanding that SARS-CoV might be with humans for years to come, knowledge of the evolutionary mechanism of the SARS-CoV, including its mutation rate and emergence time, is fundamental to battle this deadly pathogen. To date, the speed at which the deadly virus evolved in nature and the elapsed time before it was transmitted to humans remains poorly understood. Results Sixteen complete genomic sequences with available clinical histories during the SARS outbreak were analyzed. After careful examination of multiple-sequence alignment, 114 single nucleotide variations were identified. To minimize the effects of sequencing errors and additional mutations during the cell culture, three strategies were applied to estimate the mutation rate by 1) using the closely related sequences as background controls; 2) adjusting the divergence time for cell culture; or 3) using the common variants only. The mutation rate in the SARS-CoV genome was estimated to be 0.80 – 2.38 × 10-3 nucleotide substitution per site per year which is in the same order of magnitude as other RNA viruses. The non-synonymous and synonymous substitution rates were estimated to be 1.16 – 3.30 × 10-3 and 1.67 – 4.67 × 10-3 per site per year, respectively. The most recent common ancestor of the 16 sequences was inferred to be present as early as the spring of 2002. Conclusions The estimated mutation rates in the SARS-CoV using multiple strategies were not unusual among coronaviruses and moderate compared to those in other RNA viruses. All estimates of mutation rates led to the inference that the SARS-CoV could have been with humans in the spring of 2002 without causing a severe epidemic.
Collapse
Affiliation(s)
- Zhongming Zhao
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA 23219, USA
- Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Haipeng Li
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Xiaozhuang Wu
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yixi Zhong
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Keqin Zhang
- Laboratory for Conservation and Utilization of Bio-resource, Yunnan University, Kunming, China
| | - Ya-Ping Zhang
- Laboratory for Conservation and Utilization of Bio-resource, Yunnan University, Kunming, China
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yun-Xin Fu
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Laboratory for Conservation and Utilization of Bio-resource, Yunnan University, Kunming, China
| |
Collapse
|
1365
|
Cavanagh D. Severe acute respiratory syndrome vaccine development: experiences of vaccination against avian infectious bronchitis coronavirus. Avian Pathol 2004; 32:567-82. [PMID: 14676007 PMCID: PMC7154303 DOI: 10.1080/03079450310001621198] [Citation(s) in RCA: 225] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Vaccines against infectious bronchitis of chickens (Gallus gallus domesticus) have arguably been the most successful, and certainly the most widely used, of vaccines for diseases caused by coronaviruses, the others being against bovine, canine, feline and porcine coronaviruses. Infectious bronchitis virus (IBV), together with the genetically related coronaviruses of turkey (Meleagris gallopovo) and ring-necked pheasant (Phasianus colchicus), is a group 3 coronavirus, severe acute respiratory syndrome (SARS) coronavirus being tentatively in group 4, the other known mammalian coronaviruses being in groups 1 and 2. IBV replicates not only in respiratory tissues (including the nose, trachea, lungs and airsacs, causing respiratory disease), but also in the kidney (associated with minor or major nephritis), oviduct, and in many parts of the alimentary tract--the oesophagus, proventriculus, duodenum, jejunum, bursa of Fabricius, caecal tonsils (near the distal end of the tract), rectum and cloaca (the common opening for release of eggs and faeces), usually without clinical effects. The virus can persist, being re-excreted at the onset of egg laying (4 to 5 months of age), believed to be a consequence of the stress of coming into lay. Genetic lines of chickens differ in the extent to which IBV causes mortality in chicks, and in respect of clearance of the virus after the acute phase. Live attenuated (by passage in chicken embryonated eggs) IBV strains were introduced as vaccines in the 1950s, followed a couple of decades later by inactivated vaccines for boosting protection in egg-laying birds. Live vaccines are usually applied to meat-type chickens at 1 day of age. In experimental situations this can result in sterile immunity when challenged by virulent homologous virus. Although 100% of chickens may be protected (against clinical signs and loss of ciliary activity in trachea), sometimes 10% of vaccinated chicks do not respond with a protective immune response. Protection is short lived, the start of the decline being apparent 9 weeks after vaccination with vaccines based on highly attenuated strains. IBV exists as scores of serotypes (defined by the neutralization test), cross-protection often being poor. Consequently, chickens may be re-vaccinated, with the same or another serotype, two or three weeks later. Single applications of inactivated virus has generally led to protection of <50% of chickens. Two applications have led to 90 to 100% protection in some reports, but remaining below 50% in others. In practice in the field, inactivated vaccines are used in laying birds that have previously been primed with two or three live attenuated virus vaccinations. This increases protection of the laying birds against egg production losses and induces a sustained level of serum antibody, which is passed to progeny. The large spike glycoprotein (S) comprises a carboxy-terminal S2 subunit (approximately 625 amino acid residues), which anchors S in the virus envelope, and an amino-terminal S1 subunit (approximately 520 residues), believed to largely form the distal bulbous part of S. The S1 subunit (purified from IBV virus, expressed using baculovirus or expressed in birds from a fowlpoxvirus vector) induced virus neutralizing antibody. Although protective immune responses were induced, multiple inoculations were required and the percentage of protected chickens was too low (<50%) for commercial application. Remarkably, expression of S1 in birds using a non-pathogenic fowl adenovirus vector induced protection in 90% and 100% of chickens in two experiments. Differences of as little as 5% between the S1 sequences can result in poor cross-protection. Differences in S1 of 2 to 3% (10 to 15 amino acids) can change serotype, suggesting that a small number of epitopes are immunodominant with respect to neutralizing antibody. Initial studies of the role of the IBV nucleocapsid protein (N) in immunity suggested that immunization with bacterially expressed N, while not inducing protection directly, improved the induction of protection by a subsequent inoculation with inactivated IBV. In another study, two intramuscular immunizations of a plasmid expressing N induced protective immunity. The basis of immunity to IBV is not well understood. Serum antibody levels do not correlate with protection, although local antibody is believed to play a role. Adoptive transfer of IBV-infection-induced alphabeta T cells bearing CD8 antigen protected chicks from challenge infection. In conclusion, live attenuated IBV vaccines induce good, although short-lived, protection against homologous challenge, although a minority of individuals may respond poorly. Inactivated IBV vaccines are insufficiently efficacious when applied only once and in the absence of priming by live vaccine. Two applications of inactivated IBV are much more efficacious, although this is not a commercially viable proposition in the poultry industry. However, the cost and logistics of multiple application of a SARS inactivated vaccine would be more acceptable for the protection of human populations, especially if limited to targeted groups (e.g. health care workers and high-risk contacts). Application of a SARS vaccine is perhaps best limited to a minimal number of targeted individuals who can be monitored, as some vaccinated persons might, if infected by SARS coronavirus, become asymptomatic excretors of virus, thereby posing a risk to non-vaccinated people. Looking further into the future, the high efficacy of the fowl adenovirus vector expressing the IBV S1 subunit provides optimism for a live SARS vaccine, if that were deemed to be necessary, with the possibility of including the N protein gene.
Collapse
Affiliation(s)
- Dave Cavanagh
- Institute for Animal Health, Division of Molecular Biology, Compton Laboratory, Newbury, Berkshire, UK.
| |
Collapse
|
1366
|
Hertzig T, Scandella E, Schelle B, Ziebuhr J, Siddell SG, Ludewig B, Thiel V. Rapid identification of coronavirus replicase inhibitors using a selectable replicon RNA. J Gen Virol 2004; 85:1717-1725. [PMID: 15166457 DOI: 10.1099/vir.0.80044-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
A previously unknown coronavirus (CoV) is the aetiological agent causing severe acute respiratory syndrome (SARS), for which an effective antiviral treatment is urgently needed. To enable the rapid and biosafe identification of coronavirus replicase inhibitors, we have generated a non-cytopathic, selectable replicon RNA (based on human CoV 229E) that can be stably maintained in eukaryotic cells. Most importantly, the replicon RNA mediates reporter gene expression as a marker for coronavirus replication. We have used a replicon RNA-containing cell line to test the inhibitory effect of several compounds that are currently being assessed for SARS treatment. Amongst those, interferon-alpha displayed the strongest inhibitory activity. Our results demonstrate that coronavirus replicon cell lines provide a versatile and safe assay for the identification of coronavirus replicase inhibitors. Once this technology is adapted to SARS-CoV replicon RNAs, it will allow high throughput screening for SARS-CoV replicase inhibitors without the need to grow infectious SARS-CoV.
Collapse
Affiliation(s)
- Tobias Hertzig
- Institute of Virology and Immunology, University of Würzburg, Würzburg, Germany
| | - Elke Scandella
- Research Department, Cantonal Hospital St Gallen, CH-9007 St Gallen, Switzerland
| | - Barbara Schelle
- Institute of Virology and Immunology, University of Würzburg, Würzburg, Germany
| | - John Ziebuhr
- Institute of Virology and Immunology, University of Würzburg, Würzburg, Germany
| | - Stuart G Siddell
- Department of Pathology and Microbiology, School of Medical Sciences, University of Bristol, Bristol, UK
| | - Burkhard Ludewig
- Research Department, Cantonal Hospital St Gallen, CH-9007 St Gallen, Switzerland
| | - Volker Thiel
- Research Department, Cantonal Hospital St Gallen, CH-9007 St Gallen, Switzerland
| |
Collapse
|
1367
|
Abstract
PURPOSE OF REVIEW Severe acute respiratory syndrome (SARS) is a new disease that caused large outbreaks in several countries in the first half of 2003, resulting in infection in more than 8000 people and more than 900 deaths. During that time, a large body of literature rapidly emerged describing the clinical disease, the etiologic viral agent, and management options. This paper reviews the current status of this knowledge base, with particular reference to the critically ill patient. RECENT FINDINGS Case series of patients from Hong Kong, China, Singapore, and Toronto have characterized the disease with remarkable uniformity. A similarly consistent picture of the SARS patient with respiratory failure has emerged from studies from Toronto and Singapore. Worldwide collaboration led to the rapid identification and gene sequencing of the etiologic virus, SARS-CoV. SUMMARY SARS is a predominantly respiratory illness, spread through droplets from respiratory secretions and possibly via a fecal-oral route. A small number of "super-spreaders" appear to have contributed to the rapid proliferation of the disease. Infection control precautions are an essential component of management. Approximately 20% of patients develop progressive pulmonary infiltrates and respiratory failure, and the mortality rate is as high as 10%. Treatment strategies with antiviral agents such as ribavirin have not clearly demonstrated a benefit, but high-dose corticosteroids appear to be beneficial in patients with progressive disease. The recent outbreaks highlighted the potential for this disease to overwhelm critical care resources, by the volume of patients and loss of healthcare workers to illness and quarantine.
Collapse
Affiliation(s)
- Stephen E Lapinsky
- Mount Sinai Hospital, University Health Network, and the Interdepartmental Division of Critical Care, University of Toronto, Toronto, Ontario, Canada.
| | | |
Collapse
|
1368
|
Yu CJ, Chen YC, Hsiao CH, Kuo TC, Chang SC, Lu CY, Wei WC, Lee CH, Huang LM, Chang MF, Ho HN, Lee FJS. Identification of a novel protein 3a from severe acute respiratory syndrome coronavirus. FEBS Lett 2004; 565:111-6. [PMID: 15135062 PMCID: PMC7126674 DOI: 10.1016/j.febslet.2004.03.086] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2004] [Revised: 03/22/2004] [Accepted: 03/30/2004] [Indexed: 12/03/2022]
Abstract
The open reading frame 3 of the severe acute respiratory syndrome coronavirus (SARS‐CoV) genome encodes a predicted protein 3a, consisting of 274 amino acids, that lacks any significant similarities to any known protein. We generated specific antibodies against SARS protein 3a by using a synthetic peptide (P2) corresponding to amino acids 261–274 of the putative protein. Anti‐P2 antibodies and the sera from SARS patients could specifically detect the recombinant SARS protein 3a expressed in Escherichia coli and in Vero E6 cells. Expression of SARS protein 3a was detected at 8–12 h after infection and reached a higher level after ∼24 h in SARS‐CoV‐infected Vero E6 cells. Protein 3a was also detected in the alveolar lining pneumocytes and some intra‐alveolar cells of a SARS‐CoV‐infected patient's lung specimen. Recombinant protein 3a expressed in Vero E6 cells and protein 3a in the SARS‐CoV‐infected cells was distributed over the cytoplasm in a fine punctate pattern with partly concentrated staining in the Golgi apparatus. Our study demonstrates that SARS‐CoV indeed expresses a novel protein 3a, which is present only in SARS‐CoV and not in other known CoVs.
Collapse
Affiliation(s)
- Chia-Jung Yu
- Department of Medical Research, National Taiwan University Hospital, 7 Chung Shan South Road, Taipei 100, Taiwan
| | - Yee-Chun Chen
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Cheng-Hsiang Hsiao
- Department of Pathology, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Tse-Chun Kuo
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan
- Institute of Molecular Medicine, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Shin C. Chang
- Institute of Microbiology, National Taiwan University, College of Medicine, Taipei 100, Taiwan
| | - Chun-Yi Lu
- Department of Pediatrics, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Wen-Chin Wei
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chia-Huei Lee
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Li-Min Huang
- Department of Pediatrics, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Ming-Fu Chang
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Hong-Nerng Ho
- Department of Medical Research, National Taiwan University Hospital, 7 Chung Shan South Road, Taipei 100, Taiwan
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Fang-Jen S. Lee
- Department of Medical Research, National Taiwan University Hospital, 7 Chung Shan South Road, Taipei 100, Taiwan
- Institute of Molecular Medicine, National Taiwan University College of Medicine, Taipei 100, Taiwan
| |
Collapse
|
1369
|
Zhang J, Meng B, Liao D, Zhou L, Zhang X, Chen L, Guo Z, Peng C, Zhu B, Lee PP, Xu X, Zhou T, Deng Z, Hu Y, Li K. De novo synthesis of PCR templates for the development of SARS diagnostic assay. Mol Biotechnol 2004; 25:107-12. [PMID: 14526121 PMCID: PMC7090507 DOI: 10.1385/mb:25:2:107] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
A novel coronavirus was identified as the cause for severe acute respiratory syndrome (SARS). The complete sequence of SARS genome has provided an opportunity for the development of molecular diagnostic assays. To restrain further outbreak of SARS, the World Health Organization has posted several pairs of polymerase chain reaction (PCR) primers for early diagnosis and urged more research to be done on PCR protocols. Here we report a strategy for the de novo synthesis of PCR templates complimentary to the SARS virus genome, which has the advantage of working on PCR templates without concern about viral infection and also has the advantage that it can be used by those who do not have access to the SARS virus. This highly efficient and safe strategy for obtaining SARS gene fragments is useful for the development of PCR assays, as well as for the preparation of reliable positive controls for PCR testing kits.
Collapse
Affiliation(s)
| | - Bo Meng
- Shanghai Institute of Brain Functional Genomics, East China Normal University, Shanghai
| | - Duanfang Liao
- Institute of SNP, Institute of Pharmacy and Pharmacology, National University, Hengyang
| | - Lvyi Zhou
- Shanghai Institute of Brain Functional Genomics, East China Normal University, Shanghai
| | | | | | | | | | - Bingyang Zhu
- Institute of SNP, Institute of Pharmacy and Pharmacology, National University, Hengyang
| | - Peggy P. Lee
- University of California in San Diego, San Diego, USA
| | - Xiangmin Xu
- Department of Medical Genetics, The First Military Medical University, Guangzhou
| | - Tianhong Zhou
- Institute of Life Science and Biotechnology, Jinana University, Guangzhou
| | - Zemin Deng
- Genomapping Inc., Tianjin
- Genomics Group, Loma Linda, University, CA
| | - Yinghe Hu
- Shanghai Institute of Brain Functional Genomics, East China Normal University, Shanghai
| | - Kai Li
- Genomapping Inc., Tianjin
- Institute of SNP, Institute of Pharmacy and Pharmacology, National University, Hengyang
- Institute of Life Science and Biotechnology, Jinana University, Guangzhou
| |
Collapse
|
1370
|
Abstract
PURPOSE OF REVIEW In November 2003, a new, life-threatening, respiratory illness named severe acute respiratory syndrome (SARS) arose from Guangdong Province in China. The illness spread across the globe, caused many major outbreaks, and had an overall mortality rate of 11%. The purpose of this review is primarily to review the clinical features, diagnosis, and management of SARS, but also to comment briefly on the epidemiology and pathogen. RECENT FINDINGS SARS is caused by a novel coronavirus that primarily affects the lower respiratory tract. It starts with an influenza-like illness characterized by nonspecific, systemic symptoms. This is followed by the rapid development of a non-specific bronchopneumonia associated with lower tract respiratory symptoms, or gastrointestinal symptoms. Most patients recover after a week or 2, but some go on to develop acute respiratory distress syndrome. There is no proven treatment, although cocktails of broad-spectrum antibiotics, antiviral, and immunomodulatory therapy have been tried. Secondary spread can be prevented and outbreaks brought under control provided that staff wear personal protective equipment and pay close attention to good personal hygiene, and patients are isolated. The most urgent needs at present are to develop a vaccine, to develop rapid, inexpensive, accurate diagnostic tests that can give results early in the illness and within a few hours of sampling. Other needs are to investigate which therapies have the lowest adverse event/efficacy ratios. SUMMARY Up-to-date knowledge of SARS should help in early detection, isolation of high-risk patients, to reduce mortality and morbidity, and to prevent a new global epidemic arising.
Collapse
Affiliation(s)
- Timothy H Rainer
- Accident and Emergency Medicine Academic Unit, The Chinese University of Hong Kong, Shatin, NT, Hong Kong.
| |
Collapse
|
1371
|
Leung DTM, Chi Hang TF, Chun Hung M, Sheung Chan PK, Cheung JLK, Niu H, Tam JSL, Lim PL. Antibody response of patients with severe acute respiratory syndrome (SARS) targets the viral nucleocapsid. J Infect Dis 2004; 190:379-86. [PMID: 15216476 PMCID: PMC7110057 DOI: 10.1086/422040] [Citation(s) in RCA: 170] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2003] [Accepted: 02/02/2004] [Indexed: 12/18/2022] Open
Abstract
The recent outbreak of severe acute respiratory syndrome (SARS) provided an opportunity to study the antibody response of infected individuals to the causative virus, SARS coronavirus. We examined serum samples obtained from 46 patients with SARS, 40 patients with non-SARS pneumonia, and 38 healthy individuals, by use of Western blotting (WB), enzyme-linked immunoassay (ELISA), and immunofluorescence assay, using both native and bacterially produced antigens of the virus. We found a highly restricted, immunoglobulin G-dominated antibody response in patients with SARS, directed most frequently (89% by ELISA) and predominantly at the nucleocapsid. Almost all of the subjects without SARS had no antinucleocapsid antibodies. The spike protein was the next most frequently targeted, but only 63% of the patients (by ELISA) responded. Other targets of the response identified by use of WB included antigens of 80 and 60 kDa. Several nonstructural proteins cloned were not antigenic, and the culture-derived nucleocapsid appeared to be specifically degraded.
Collapse
Affiliation(s)
| | | | - Ma Chun Hung
- Clinical Immunology Unit, Shatin, New Territories, Hong Kong, HKSAR, China
| | - Paul Kay Sheung Chan
- Department of Microbiology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong, HKSAR, China
| | - Jo Lai Ken Cheung
- Department of Microbiology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong, HKSAR, China
| | - Haitao Niu
- Clinical Immunology Unit, Shatin, New Territories, Hong Kong, HKSAR, China
| | - John Siu Lun Tam
- Department of Microbiology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong, HKSAR, China
| | - Pak Leong Lim
- Clinical Immunology Unit, Shatin, New Territories, Hong Kong, HKSAR, China
- Reprints or correspondence: Prof. Pak Leong Lim, Clinical Immunology Unit, The Chinese University of Hong Kong, Prince ofWales Hospital, Shatin, New Territories, Hong Kong, HKSAR, China ()
| |
Collapse
|
1372
|
Wang YD, Sin WYF, Xu GB, Yang HH, Wong TY, Pang XW, He XY, Zhang HG, Ng JNL, Cheng CSS, Yu J, Meng L, Yang RF, Lai ST, Guo ZH, Xie Y, Chen WF, Yang HH. T-cell epitopes in severe acute respiratory syndrome (SARS) coronavirus spike protein elicit a specific T-cell immune response in patients who recover from SARS. J Virol 2004; 78:5612-8. [PMID: 15140958 PMCID: PMC415819 DOI: 10.1128/jvi.78.11.5612-5618.2004] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The immunogenicity of HLA-A2-restricted T-cell epitopes in the S protein of the Severe acute respiratory syndrome coronavirus (SARS-CoV) and of human coronavirus strain 229e (HCoV-229e) was analyzed for the elicitation of a T-cell immune response in donors who had fully recovered from SARS-CoV infection. We employed online database analysis to compare the differences in the amino acid sequences of the homologous T epitopes of HCoV-229e and SARS-CoV. The identified T-cell epitope peptides were synthesized, and their binding affinities for HLA-A2 were validated and compared in the T2 cell system. The immunogenicity of all these peptides was assessed by using T cells obtained from donors who had fully recovered from SARS-CoV infection and from healthy donors with no history of SARS-CoV infection. HLA-A2 typing by indirect immunofluorescent antibody staining showed that 51.6% of SARS-CoV-infected patients were HLA-A2 positive. Online database analysis and the T2 cell binding test disclosed that the number of HLA-A2-restricted immunogenic epitopes of the S protein of SARS-CoV was decreased or even lost in comparison with the homologous sequences of the S protein of HCoV-229e. Among the peptides used in the study, the affinity of peptides from HCoV-229e (H77 and H881) and peptides from SARS-CoV (S978 and S1203) for binding to HLA-A2 was higher than that of other sequences. The gamma interferon (IFN-gamma) release Elispot assay revealed that only SARS-CoV-specific peptides S1203 and S978 induced a high frequency of IFN-gamma-secreting T-cell response in HLA-A2(+) donors who had fully recovered from SARS-CoV infection; such a T-cell epitope-specific response was not observed in HLA-A2(+) healthy donors or in HLA-A2(-) donors who had been infected with SARS-CoV after full recovery. Thus, T-cell epitopes S1203 and S978 are immunogenic and elicit an overt specific T-cell response in HLA-A2(+) SARS-CoV-infected patients.
Collapse
Affiliation(s)
- Yue-Dan Wang
- Department of Immunology, Peking University Health Science Centre, 38, Xueyuanlu, Beijing, 100083, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1373
|
Ivanov KA, Thiel V, Dobbe JC, van der Meer Y, Snijder EJ, Ziebuhr J. Multiple enzymatic activities associated with severe acute respiratory syndrome coronavirus helicase. J Virol 2004; 78:5619-32. [PMID: 15140959 PMCID: PMC415832 DOI: 10.1128/jvi.78.11.5619-5632.2004] [Citation(s) in RCA: 333] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV), a newly identified group 2 coronavirus, is the causative agent of severe acute respiratory syndrome, a life-threatening form of pneumonia in humans. Coronavirus replication and transcription are highly specialized processes of cytoplasmic RNA synthesis that localize to virus-induced membrane structures and were recently proposed to involve a complex enzymatic machinery that, besides RNA-dependent RNA polymerase, helicase, and protease activities, also involves a series of RNA-processing enzymes that are not found in most other RNA virus families. Here, we characterized the enzymatic activities of a recombinant form of the SARS-CoV helicase (nonstructural protein [nsp] 13), a superfamily 1 helicase with an N-terminal zinc-binding domain. We report that nsp13 has both RNA and DNA duplex-unwinding activities. SARS-CoV nsp13 unwinds its substrates in a 5'-to-3' direction and features a remarkable processivity, allowing efficient strand separation of extended regions of double-stranded RNA and DNA. Characterization of the nsp13-associated (deoxy)nucleoside triphosphatase ([dNTPase) activities revealed that all natural nucleotides and deoxynucleotides are substrates of nsp13, with ATP, dATP, and GTP being hydrolyzed slightly more efficiently than other nucleotides. Furthermore, we established an RNA 5'-triphosphatase activity for the SARS-CoV nsp13 helicase which may be involved in the formation of the 5' cap structure of viral RNAs. The data suggest that the (d)NTPase and RNA 5'-triphosphatase activities of nsp13 have a common active site. Finally, we established that, in SARS-CoV-infected Vero E6 cells, nsp13 localizes to membranes that appear to be derived from the endoplasmic reticulum and are the likely site of SARS-CoV RNA synthesis.
Collapse
Affiliation(s)
- Konstantin A Ivanov
- Institute of Virology and Immunology, University of Würzburg, Würzburg, Germany
| | | | | | | | | | | |
Collapse
|
1374
|
Neuman BW, Stein DA, Kroeker AD, Paulino AD, Moulton HM, Iversen PL, Buchmeier MJ. Antisense morpholino-oligomers directed against the 5' end of the genome inhibit coronavirus proliferation and growth. J Virol 2004; 78:5891-9. [PMID: 15140987 PMCID: PMC415795 DOI: 10.1128/jvi.78.11.5891-5899.2004] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Conjugation of a peptide related to the human immunodeficiency virus type 1 Tat represents a novel method for delivery of antisense morpholino-oligomers. Conjugated and unconjugated oligomers were tested to determine sequence-specific antiviral efficacy against a member of the Coronaviridae, Mouse hepatitis virus (MHV). Specific antisense activity designed to block translation of the viral replicase polyprotein was first confirmed by reduction of luciferase expression from a target sequence-containing reporter construct in both cell-free and transfected cell culture assays. Peptide-conjugated morpholino-oligomers exhibited low toxicity in DBT astrocytoma cells used for culturing MHV. Oligomer administered at micromolar concentrations was delivered to >80% of cells and inhibited virus titers 10- to 100-fold in a sequence-specific and dose-responsive manner. In addition, targeted viral protein synthesis, plaque diameter, and cytopathic effect were significantly reduced. Inhibition of virus infectivity by peptide-conjugated morpholino was comparable to the antiviral activity of the aminoglycoside hygromycin B used at a concentration fivefold higher than the oligomer. These results suggest that this composition of antisense compound has therapeutic potential for control of coronavirus infection.
Collapse
Affiliation(s)
- Benjamin W Neuman
- The Scripps Research Institute, Department of Neuropharmacology, Division of Virology, 10550 North Torrey Pines Rd., La Jolla, CA 92037, USA.
| | | | | | | | | | | | | |
Collapse
|
1375
|
Yang ZY, Huang Y, Ganesh L, Leung K, Kong WP, Schwartz O, Subbarao K, Nabel GJ. pH-dependent entry of severe acute respiratory syndrome coronavirus is mediated by the spike glycoprotein and enhanced by dendritic cell transfer through DC-SIGN. J Virol 2004; 78:5642-50. [PMID: 15140961 PMCID: PMC415834 DOI: 10.1128/jvi.78.11.5642-5650.2004] [Citation(s) in RCA: 388] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus (SARS-CoV) synthesizes several putative viral envelope proteins, including the spike (S), membrane (M), and small envelope (E) glycoproteins. Although these proteins likely are essential for viral replication, their specific roles in SARS-CoV entry have not been defined. In this report, we show that the SARS-CoV S glycoprotein mediates viral entry through pH-dependent endocytosis. Further, we define its cellular tropism and demonstrate that virus transmission occurs through cell-mediated transfer by dendritic cells. The S glycoprotein was used successfully to pseudotype replication-defective retroviral and lentiviral vectors that readily infected Vero cells as well as primary pulmonary and renal epithelial cells from human, nonhuman primate, and, to a lesser extent, feline species. The tropism of this reporter virus was similar to that of wild-type, replication-competent SARS-CoV, and binding of purified S to susceptible target cells was demonstrated by flow cytometry. Although myeloid dendritic cells were able to interact with S and to bind virus, these cells could not be infected by SARS-CoV. However, these cells were able to transfer the virus to susceptible target cells through a synapse-like structure. Both cell-mediated infection and direct infection were inhibited by anti-S antisera, indicating that strategies directed toward this gene product are likely to confer a therapeutic benefit for antiviral drugs or the development of a SARS vaccine.
Collapse
Affiliation(s)
- Zhi-Yong Yang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bldg. 40, Room 4502, MSC-3005, 40 Convent Dr., Bethesda, MD 20892-3005, USA
| | | | | | | | | | | | | | | |
Collapse
|
1376
|
Abstract
The severe acute respiratory syndrome (SARS) is a highly contagious infection caused by a newly discovered strain of coronavirus (SARS-CoV). Infants born to pregnant women with SARS did not appear to acquire the infection through vertical transmission. Some newborn infants, however, developed severe intrauterine growth retardation and life-threatening gastrointestinal complications. It is now known that the clinical course and prognosis are different between paediatric and adult SARS patients. Young children (< 12 years), in general, run a less aggressive clinical course than do teenage and adult patients. Thus far, no fatalities have been reported in the paediatric age group (< or =18 years). This review describes the current understanding of the clinical manifestations, diagnostic tests, immunological profiles, patient management and outcomes of SARS-CoV infection in the paediatric population.
Collapse
Affiliation(s)
- Pak C Ng
- Department of Paediatrics, Prince of Wales Hospital, Chinese University of Hong Kong, Shatin, New Territories, Hong Kong. pakcheungngcuhk.edu.hk
| | | | | | | | | |
Collapse
|
1377
|
Ingallinella P, Bianchi E, Finotto M, Cantoni G, Eckert DM, Supekar VM, Bruckmann C, Carfi A, Pessi A. Structural characterization of the fusion-active complex of severe acute respiratory syndrome (SARS) coronavirus. Proc Natl Acad Sci U S A 2004; 101:8709-14. [PMID: 15161975 PMCID: PMC423260 DOI: 10.1073/pnas.0402753101] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2003] [Indexed: 11/18/2022] Open
Abstract
The causative agent of a recent outbreak of an atypical pneumonia, known as severe acute respiratory syndrome (SARS), has been identified as a coronavirus (CoV) not belonging to any of the previously identified groups. Fusion of coronaviruses with the host cell is mediated by the envelope spike protein. Two regions within the spike protein of SARS-CoV have been identified, showing a high degree of sequence conservation with the other CoV, which are characterized by the presence of heptad repeats (HR1 and HR2). By using synthetic and recombinant peptides corresponding to the HR1 and HR2 regions, we were able to characterize the fusion-active complex formed by this novel CoV by CD, native PAGE, proteolysis protection analysis, and size-exclusion chromatography. HR1 and HR2 of SARS-CoV associate into an antiparallel six-helix bundle, with structural features typical of the other known class I fusion proteins. We have also mapped the specific boundaries of the region, within the longer HR1 domain, making contact with the shorter HR2 domain. Notably, the inner HR1 coiled coil is a stable alpha-helical domain even in the absence of interaction with the HR2 region. Inhibitors binding to HR regions of fusion proteins have been shown to be efficacious against many viruses, notably HIV. Our results may help in the design of anti-SARS therapeutics.
Collapse
Affiliation(s)
- Paolo Ingallinella
- Istituto di Ricerche di Biologia Molecolare P. Angeletti, Via Pontina Km 30.600, 00040 Pomezia, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
1378
|
Wong CK, Lam CWK, Wu AKL, Ip WK, Lee NLS, Chan IHS, Lit LCW, Hui DSC, Chan MHM, Chung SSC, Sung JJY. Plasma inflammatory cytokines and chemokines in severe acute respiratory syndrome. Clin Exp Immunol 2004; 136:95-103. [PMID: 15030519 PMCID: PMC1808997 DOI: 10.1111/j.1365-2249.2004.02415.x] [Citation(s) in RCA: 937] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Severe acute respiratory syndrome (SARS) is a recently emerged infectious disease caused by a novel coronavirus, but its immunopathological mechanisms have not yet been fully elucidated. We investigated changes in plasma T helper (Th) cell cytokines, inflammatory cytokines and chemokines in 20 patients diagnosed with SARS. Cytokine profile of SARS patients showed marked elevation of Th1 cytokine interferon (IFN)-gamma, inflammatory cytokines interleukin (IL)-1, IL-6 and IL-12 for at least 2 weeks after disease onset, but there was no significant elevation of inflammatory cytokine tumour necrosis factor (TNF)-alpha, anti-inflammatory cytokine IL-10, Th1 cytokine IL-2 and Th2 cytokine IL-4. The chemokine profile demonstrated significant elevation of neutrophil chemokine IL-8, monocyte chemoattractant protein-1 (MCP-1), and Th1 chemokine IFN-gamma-inducible protein-10 (IP-10). Corticosteroid reduced significantly IL-8, MCP-1 and IP-10 concentrations from 5 to 8 days after treatment (all P < 0.001). Together, the elevation of Th1 cytokine IFN-gamma, inflammatory cytokines IL-1, IL-6 and IL-12 and chemokines IL-8, MCP-1 and IP-10 confirmed the activation of Th1 cell-mediated immunity and hyperinnate inflammatory response in SARS through the accumulation of monocytes/macrophages and neutrophils.
Collapse
Affiliation(s)
- C K Wong
- Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1379
|
Kiemer L, Lund O, Brunak S, Blom N. Coronavirus 3CLpro proteinase cleavage sites: possible relevance to SARS virus pathology. BMC Bioinformatics 2004; 5:72. [PMID: 15180906 PMCID: PMC442122 DOI: 10.1186/1471-2105-5-72] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2004] [Accepted: 06/06/2004] [Indexed: 11/30/2022] Open
Abstract
Background Despite the passing of more than a year since the first outbreak of Severe Acute Respiratory Syndrome (SARS), efficient counter-measures are still few and many believe that reappearance of SARS, or a similar disease caused by a coronavirus, is not unlikely. For other virus families like the picornaviruses it is known that pathology is related to proteolytic cleavage of host proteins by viral proteinases. Furthermore, several studies indicate that virus proliferation can be arrested using specific proteinase inhibitors supporting the belief that proteinases are indeed important during infection. Prompted by this, we set out to analyse and predict cleavage by the coronavirus main proteinase using computational methods. Results We retrieved sequence data on seven fully sequenced coronaviruses and identified the main 3CL proteinase cleavage sites in polyproteins using alignments. A neural network was trained to recognise the cleavage sites in the genomes obtaining a sensitivity of 87.0% and a specificity of 99.0%. Several proteins known to be cleaved by other viruses were submitted to prediction as well as proteins suspected relevant in coronavirus pathology. Cleavage sites were predicted in proteins such as the cystic fibrosis transmembrane conductance regulator (CFTR), transcription factors CREB-RP and OCT-1, and components of the ubiquitin pathway. Conclusions Our prediction method NetCorona predicts coronavirus cleavage sites with high specificity and several potential cleavage candidates were identified which might be important to elucidate coronavirus pathology. Furthermore, the method might assist in design of proteinase inhibitors for treatment of SARS and possible future diseases caused by coronaviruses. It is made available for public use at our website: .
Collapse
Affiliation(s)
- Lars Kiemer
- Center for Biological Sequence Analysis BioCentrum-DTU, Building 208 Technical University of Denmark DK-2800 Lyngby, Denmark
| | - Ole Lund
- Center for Biological Sequence Analysis BioCentrum-DTU, Building 208 Technical University of Denmark DK-2800 Lyngby, Denmark
| | - Søren Brunak
- Center for Biological Sequence Analysis BioCentrum-DTU, Building 208 Technical University of Denmark DK-2800 Lyngby, Denmark
| | - Nikolaj Blom
- Center for Biological Sequence Analysis BioCentrum-DTU, Building 208 Technical University of Denmark DK-2800 Lyngby, Denmark
| |
Collapse
|
1380
|
Svoboda T, Henry B, Shulman L, Kennedy E, Rea E, Ng W, Wallington T, Yaffe B, Gournis E, Vicencio E, Basrur S, Glazier RH. Public health measures to control the spread of the severe acute respiratory syndrome during the outbreak in Toronto. N Engl J Med 2004; 350:2352-61. [PMID: 15175437 DOI: 10.1056/nejmoa032111] [Citation(s) in RCA: 157] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Toronto was the site of North America's largest outbreak of the severe acute respiratory syndrome (SARS). An understanding of the patterns of transmission and the effects on public health in relation to control measures that were taken will help health officials prepare for any future outbreaks. METHODS We analyzed SARS case, quarantine, and hotline records in relation to control measures. The two phases of the outbreak were compared. RESULTS Toronto Public Health investigated 2132 potential cases of SARS, identified 23,103 contacts of SARS patients as requiring quarantine, and logged 316,615 calls on its SARS hotline. In Toronto, 225 residents met the case definition of SARS, and all but 3 travel-related cases were linked to the index patient, from Hong Kong. SARS spread to 11 (58 percent) of Toronto's acute care hospitals. Unrecognized SARS among in-patients with underlying illness caused a resurgence, or a second phase, of the outbreak, which was finally controlled through active surveillance of hospitalized patients. In response to the control measures of Toronto Public Health, the number of persons who were exposed to SARS in nonhospital and nonhousehold settings dropped from 20 (13 percent) before the control measures were instituted (phase 1) to 0 afterward (phase 2). The number of patients who were exposed while in a hospital ward rose from 25 (17 percent) in phase 1 to 68 (88 percent) in phase 2, and the number exposed while in the intensive care unit dropped from 13 (9 percent) in phase 1 to 0 in phase 2. Community spread (the length of the chains of transmission outside of hospital settings) was significantly reduced in phase 2 of the outbreak (P<0.001). CONCLUSIONS The transmission of SARS in Toronto was limited primarily to hospitals and to households that had had contact with patients. For every case of SARS, health authorities should expect to quarantine up to 100 contacts of the patients and to investigate 8 possible cases. During an outbreak, active in-hospital surveillance for SARS-like illnesses and heightened infection-control measures are essential.
Collapse
Affiliation(s)
- Tomislav Svoboda
- Inner City Health Research Unit, Toronto Public Health, Toronto, Canada.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1381
|
Yao YX, Ren J, Heinen P, Zambon M, Jones IM. Cleavage and serum reactivity of the severe acute respiratory syndrome coronavirus spike protein. J Infect Dis 2004; 190:91-8. [PMID: 15195247 PMCID: PMC7110131 DOI: 10.1086/421280] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2003] [Accepted: 12/11/2003] [Indexed: 01/13/2023] Open
Abstract
Severe acute respiratory syndrome (SARS) coronavirus (SCoV) spike (S) protein is the major surface antigen of the virus and is responsible for receptor binding and the generation of neutralizing antibody. To investigate SCoV S protein, full-length and individual domains of S protein were expressed on the surface of insect cells and were characterized for cleavability and reactivity with serum samples obtained from patients during the convalescent phase of SARS. S protein could be cleaved by exogenous trypsin but not by coexpressed furin, suggesting that the protein is not normally processed during infection. Reactivity was evident by both flow cytometry and Western blot assays, but the pattern of reactivity varied according to assay and sequence of the antigen. The antibody response to SCoV S protein involves antibodies to both linear and conformational epitopes, with linear epitopes associated with the carboxyl domain and conformational epitopes associated with the amino terminal domain. Recombinant SCoV S protein appears to be a suitable antigen for the development of an efficient and sensitive diagnostic test for SARS, but our data suggest that assay format and choice of S antigen are important considerations.
Collapse
Affiliation(s)
- Yong Xiu Yao
- School of Animal and Microbial Sciences, University of Reading, Reading,London, United Kingdom
| | - Junyuan Ren
- School of Animal and Microbial Sciences, University of Reading, Reading,London, United Kingdom
| | - Paul Heinen
- Public Health Laboratory Service, Enteric and Respiratory Virus Laboratory, London, United Kingdom
| | - Maria Zambon
- Public Health Laboratory Service, Enteric and Respiratory Virus Laboratory, London, United Kingdom
| | - Ian M. Jones
- School of Animal and Microbial Sciences, University of Reading, Reading,London, United Kingdom
- Reprints or correspondence: Dr. Ian M. Jones, School of Animal and Microbial Sciences, University of Reading, Reading RG6 6AJ, UK()
| |
Collapse
|
1382
|
Lai KN, Tsang KW, Seto WH, Ooi CGC. Clinical, Laboratory, and Radiologic Manifestation of SARS. Curr Infect Dis Rep 2004; 6:213-219. [PMID: 15142485 PMCID: PMC7088556 DOI: 10.1007/s11908-004-0011-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Severe acute respiratory syndrome (SARS) is a highly contagious and predominantly pneumonic illness caused by a novel coronavirus now commonly known as SARS-CoV. This article describes the key diagnostic clinical features, radiologic features, and investigation profiles of affected patients. We summarize our understanding from anecdotal experience and limited published data on the use of antiviral and corticosteroid therapy in the management of this highly contagious disease.
Collapse
Affiliation(s)
- Kar Neng Lai
- University Department of Medicine, The University of Hong Kong, Room 409, Professorial Block, Queen Mary Hospital, Pokfulam, Hong Kong.
| | | | | | | |
Collapse
|
1383
|
Surjit M, Liu B, Kumar P, Chow VTK, Lal SK. The nucleocapsid protein of the SARS coronavirus is capable of self-association through a C-terminal 209 amino acid interaction domain. Biochem Biophys Res Commun 2004; 317:1030-6. [PMID: 15094372 PMCID: PMC7111157 DOI: 10.1016/j.bbrc.2004.03.154] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2004] [Indexed: 12/12/2022]
Abstract
Severe acute respiratory syndrome (SARS) coronavirus (SARS-CoV) caused a severe outbreak in several regions of the world in 2003. The virus is a novel coronavirus isolated from patients exhibiting atypical pneumonia and may have originated from wild animals such as civet cats in southern China. The genome of SARS-CoV is a positive-sense, single-stranded RNA whose sequence is distantly related to all known coronaviruses that infect humans and animals. Like other known coronaviruses, SARS-CoV is an enveloped virus containing three outer structural proteins, namely the membrane (M), envelope (E), and spike (S) proteins. The nucleocapsid (N) protein together with the viral RNA genome presumably form a helical core located within the viral envelope. The SARS-CoV nucleocapsid (N) protein is a 423 amino-acid, predicted phospho-protein of 46 kDa that shares little homology with other members of the coronavirus family. A short serine-rich stretch, and a putative bipartite nuclear localization signal are unique to it, thus suggesting its involvement in many important functions during the viral life cycle. In this report we have cloned the N gene of the SARS coronavirus, and studied its property of self-association to form dimers. We expressed the N protein as a fusion protein in the yeast two-hybrid system to demonstrate self-association and confirmed dimerization of the N protein from mammalian cell lysates by coimmunoprecipitation. Furthermore, via deletion analysis, we have shown that the C-terminal 209 amino-acid region constitutes the interaction domain responsible for self-association of the N protein to form dimers.
Collapse
Affiliation(s)
- Milan Surjit
- Virology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Road, New Delhi 110067, India
| | | | | | | | | |
Collapse
|
1384
|
Kim TW, Lee JH, Hung CF, Peng S, Roden R, Wang MC, Viscidi R, Tsai YC, He L, Chen PJ, Boyd DAK, Wu TC. Generation and characterization of DNA vaccines targeting the nucleocapsid protein of severe acute respiratory syndrome coronavirus. J Virol 2004; 78:4638-45. [PMID: 15078946 PMCID: PMC387705 DOI: 10.1128/jvi.78.9.4638-4645.2004] [Citation(s) in RCA: 145] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Severe acute respiratory syndrome (SARS) is a serious threat to public health and the economy on a global scale. The SARS coronavirus (SARS-CoV) has been identified as the etiological agent for SARS. Thus, vaccination against SARS-CoV may represent an effective approach to controlling SARS. DNA vaccines are an attractive approach for SARS vaccine development, as they offer many advantages over conventional vaccines, including stability, simplicity, and safety. Our investigators have previously shown that DNA vaccination with antigen linked to calreticulin (CRT) dramatically enhances major histocompatibility complex class I presentation of linked antigen to CD8(+) T cells. In this study, we have employed this CRT-based enhancement strategy to create effective DNA vaccines using SARS-CoV nucleocapsid (N) protein as a target antigen. Vaccination with naked CRT/N DNA generated the most potent N-specific humoral and T-cell-mediated immune responses in vaccinated C57BL/6 mice among all of the DNA constructs tested. Furthermore, mice vaccinated with CRT/N DNA were capable of significantly reducing the titer of challenging vaccinia virus expressing the N protein of the SARS virus. These results show that a DNA vaccine encoding CRT linked to a SARS-CoV antigen is capable of generating strong N-specific humoral and cellular immunity and may potentially be useful for control of infection with SARS-CoV.
Collapse
Affiliation(s)
- Tae Woo Kim
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1385
|
de Paiva TM, Kisielius JJ, Benega MA, Ueda M, Sugahara TKN, Simoes Santos CL, Timenetsky MDCST, Cruz AS, Carvalhanas TRMP, Barbosa HA, Paradella RM. Severe acute respiratory syndrome-a global concern-influenza virus isolated from suspected cases in Brazil from April to June 2003. INTERNATIONAL CONGRESS SERIES 2004; 1263:422-425. [PMID: 32288143 PMCID: PMC7135486 DOI: 10.1016/j.ics.2004.02.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Severe acute respiratory syndrome (SARS) is a condition associated with substantial morbidity and mortality. Coronavirus has been associated with this severe emerging disease, with a pattern suggesting droplet or contact transmission. From April to June 2003, Institute Adolfo Lutz received 16 respiratory secretions from hospitalized patients with recent history of travel to an area with local transmission of SARS. Rapid antigen detection for influenza A and B, parainfluenza types 1, 2 and 3, respiratory syncytial virus and adenovirus; electron microscopy, polymerase chain reaction (PCR), reverse transcription PCR (RT-PCR) and serologic assay were performed. Virus isolation attempts were performed in Hep-2, Vero, MDCK, NCI-H292, MRC-5, LLC-MK2 and FRhK-4. Influenza virus of type A/Panama/2007/99 (H3N2) and A/New Caledonia/20/99 (H1N2) were identified.
Collapse
Affiliation(s)
| | | | | | - Marli Ueda
- Serviço de Virologia, Instituto Adolfo Lutz, São Paulo, SP, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
1386
|
Abstract
PURPOSE OF REVIEW Severe acute respiratory syndrome (SARS) is an infectious disease first recognized in November 2002 in Guangdong Province, China. It spread to many countries all over the world during February to June 2003, with 8098 cases reported. Twenty-one percent of the affected people were health care workers. Because SARS is a new emerging disease, this review describes the current understanding about the etiology, clinical pictures, laboratory and radiological findings of SARS. RECENT FINDINGS Severe acute respiratory syndrome-associated coronavirus (SARS-CoV) was quickly found to be the etiological agent of SARS in April 2003. The transmission of SARS-CoV between human beings is mainly due to close contact. Using barrier precautions, the transmission of SARS-CoV can be prevented. The most common clinical presentations of patients with SARS include fever, cough, and dyspnea. The common laboratory findings include lymphopenia, thrombocytopenia, elevated serum alanine and aspartate aminotransferase, lactate dehydrogenase, creatine phosphokinase, and C-reactive protein. The most common radiological finding is pneumonic lesion(s) in the chest radiogram. Many patients experience exacerbation of clinical symptoms in the second week of disease course and some may progress to respiratory failure and need mechanical ventilatory support. The overall case fatality rate is 9.6%. The current method of treatment of SARS is still controversial. SUMMARY SARS is an infectious disease with high contagiousness and a high mortality rate. Early case identification and infection control are two important factors to limit its spread.
Collapse
Affiliation(s)
- Jann-Tay Wang
- Division of Infectious Diseases, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | | |
Collapse
|
1387
|
Turner BC, Hemmila EM, Beauchemin N, Holmes KV. Receptor-dependent coronavirus infection of dendritic cells. J Virol 2004; 78:5486-90. [PMID: 15113927 PMCID: PMC400329 DOI: 10.1128/jvi.78.10.5486-5490.2004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In several mammalian species, including humans, coronavirus infection can modulate the host immune response. We show a potential role of dendritic cells (DC) in murine coronavirus-induced immune modulation and pathogenesis by demonstrating that the JAW SII DC line and primary DC from BALB/c mice and p/p mice with reduced expression of the murine coronavirus receptor, murine CEACAM1a, are susceptible to murine coronavirus infection by a receptor-dependent pathway.
Collapse
Affiliation(s)
- Brian C Turner
- Department of Microbiology, University of Colorado Health Sciences Center, 4200 East 9th Avenue, Denver, CO 80262, USA
| | | | | | | |
Collapse
|
1388
|
Vicenzi E, Canducci F, Pinna D, Mancini N, Carletti S, Lazzarin A, Bordignon C, Poli G, Clementi M. Coronaviridae and SARS-associated coronavirus strain HSR1. Emerg Infect Dis 2004; 10:413-8. [PMID: 15109406 PMCID: PMC3322807 DOI: 10.3201/eid1003.030683] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
During the recent severe acute respiratory (SARS) outbreak, the etiologic agent was identified as a new coronavirus (CoV). We have isolated a SARS-associated CoV (SARS-CoV) strain by injecting Vero cells with a sputum specimen from an Italian patient affected by a severe pneumonia; the patient traveled from Vietnam to Italy in March 2003. Ultrastructural analysis of infected Vero cells showed the virions within cell vesicles and around the cell membrane. The full-length viral genome sequence was similar to those derived from the Hong-Kong Hotel M isolate. By using both real-time reverse transcription–polymerase chain reaction TaqMan assay and an infectivity plaque assay, we determined that approximately 360 viral genomes were required to generate a PFU. In addition, heparin (100 μg/mL) inhibited infection of Vero cells by 50%. Overall, the molecular and biologic characteristics of the strain HSR1 provide evidence that SARS-CoV forms a fourth genetic coronavirus group with distinct genomic and biologic features.
Collapse
Affiliation(s)
- Elisa Vicenzi
- P2-P3 Laboratories, DIBIT, San Raffaele Scientific Institute, Via Olgettina n. 58, 20132 Milan, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
1389
|
Pavlović-Lažetić GM, Mitić NS, Beljanski MV. Bioinformatics analysis of SARS coronavirus genome polymorphism. BMC Bioinformatics 2004; 5:65. [PMID: 15161495 PMCID: PMC434493 DOI: 10.1186/1471-2105-5-65] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2003] [Accepted: 05/25/2004] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We have compared 38 isolates of the SARS-CoV complete genome. The main goal was twofold: first, to analyze and compare nucleotide sequences and to identify positions of single nucleotide polymorphism (SNP), insertions and deletions, and second, to group them according to sequence similarity, eventually pointing to phylogeny of SARS-CoV isolates. The comparison is based on genome polymorphism such as insertions or deletions and the number and positions of SNPs. RESULTS The nucleotide structure of all 38 isolates is presented. Based on insertions and deletions and dissimilarity due to SNPs, the dataset of all the isolates has been qualitatively classified into three groups each having their own subgroups. These are the A-group with "regular" isolates (no insertions / deletions except for 5' and 3' ends), the B-group of isolates with "long insertions", and the C-group of isolates with "many individual" insertions and deletions. The isolate with the smallest average number of SNPs, compared to other isolates, has been identified (TWH). The density distribution of SNPs, insertions and deletions for each group or subgroup, as well as cumulatively for all the isolates is also presented, along with the gene map for TWH. Since individual SNPs may have occurred at random, positions corresponding to multiple SNPs (occurring in two or more isolates) are identified and presented. This result revises some previous results of a similar type. Amino acid changes caused by multiple SNPs are also identified (for the annotated sequences, as well as presupposed amino acid changes for non-annotated ones). Exact SNP positions for the isolates in each group or subgroup are presented. Finally, a phylogenetic tree for the SARS-CoV isolates has been produced using the CLUSTALW program, showing high compatibility with former qualitative classification. CONCLUSIONS The comparative study of SARS-CoV isolates provides essential information for genome polymorphism, indication of strain differences and variants evolution. It may help with the development of effective treatment.
Collapse
Affiliation(s)
- Gordana M Pavlović-Lažetić
- Faculty of Mathematics, University of Belgrade, P.O.B. 550, Studentski trg 16, 11001 Belgrade, Serbia and Montenegro
| | - Nenad S Mitić
- Faculty of Mathematics, University of Belgrade, P.O.B. 550, Studentski trg 16, 11001 Belgrade, Serbia and Montenegro
| | - Miloš V Beljanski
- Institute of General and Physical Chemistry, P.O.B. 551, Studentski trg 16, 11001 Belgrade, Serbia and Montenegro
| |
Collapse
|
1390
|
Sung JJY, Wu A, Joynt GM, Yuen KY, Lee N, Chan PKS, Cockram CS, Ahuja AT, Yu LM, Wong VW, Hui DSC. Severe acute respiratory syndrome: report of treatment and outcome after a major outbreak. Thorax 2004; 59:414-20. [PMID: 15115870 PMCID: PMC1746995 DOI: 10.1136/thx.2003.014076] [Citation(s) in RCA: 136] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND The outcome is reported of a prospective uncontrolled study based on a stepwise treatment protocol during an outbreak of severe acute respiratory syndrome (SARS) in Hong Kong. METHOD One hundred and thirty eight patients were treated with broad spectrum antibiotics, a combination of ribavirin and low dose corticosteroid, and then intravenous high dose methylprednisolone according to responses. Sustained response to treatment was defined as (1) defervescence for > or =4 consecutive days, (2) resolution of lung consolidation by >25%, and (3) oxygen independence by the fourth day without fever. Patients with defervescence who achieved either criterion 2 or 3 were classified as partial responders. Patients who fell short of criteria 2 and 3 were non-responders. RESULTS Laboratory confirmation of SARS coronavirus infection was established in 132 (95.7%). None responded to antibiotics but 25 (18.1%) responded to ribavirin + low dose corticosteroid. Methylprednisolone was used in 107 patients, of whom 95 (88.8%) responded favourably. Evidence of haemolytic anaemia was observed in 49 (36%). A high level of C-reactive protein at presentation was the only independent predictor for use of methylprednisolone (odds ratio 2.18 per 10 mg/dl increase, 95% confidence interval 1.12 to 4.25, p = 0.02). Thirty seven patients (26.8%) required admission to the intensive care unit and 21 (15.2%) required invasive mechanical ventilation. There were 15 deaths (mortality rate 10.9%), most with significant co-morbidities, whereas 122 (88.4%) had been discharged home 4 months after the outbreak onset. CONCLUSION The use of high dose pulse methylprednisolone during the clinical course of a SARS outbreak was associated with clinical improvement, but randomised controlled trials are needed to ascertain its efficacy in this condition.
Collapse
Affiliation(s)
- J J Y Sung
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1391
|
Hoffmaster AR, Ravel J, Rasko DA, Chapman GD, Chute MD, Marston CK, De BK, Sacchi CT, Fitzgerald C, Mayer LW, Maiden MCJ, Priest FG, Barker M, Jiang L, Cer RZ, Rilstone J, Peterson SN, Weyant RS, Galloway DR, Read TD, Popovic T, Fraser CM. Identification of anthrax toxin genes in a Bacillus cereus associated with an illness resembling inhalation anthrax. Proc Natl Acad Sci U S A 2004; 101:8449-54. [PMID: 15155910 PMCID: PMC420414 DOI: 10.1073/pnas.0402414101] [Citation(s) in RCA: 347] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Bacillus anthracis is the etiologic agent of anthrax, an acute fatal disease among mammals. It was thought to differ from Bacillus cereus, an opportunistic pathogen and cause of food poisoning, by the presence of plasmids pXO1 and pXO2, which encode the lethal toxin complex and the poly-gamma-d-glutamic acid capsule, respectively. This work describes a non-B. anthracis isolate that possesses the anthrax toxin genes and is capable of causing a severe inhalation anthrax-like illness. Although initial phenotypic and 16S rRNA analysis identified this isolate as B. cereus, the rapid generation and analysis of a high-coverage draft genome sequence revealed the presence of a circular plasmid, named pBCXO1, with 99.6% similarity with the B. anthracis toxin-encoding plasmid, pXO1. Although homologues of the pXO2 encoded capsule genes were not found, a polysaccharide capsule cluster is encoded on a second, previously unidentified plasmid, pBC218. A/J mice challenged with B. cereus G9241 confirmed the virulence of this strain. These findings represent an example of how genomics could rapidly assist public health experts responding not only to clearly identified select agents but also to novel agents with similar pathogenic potentials. In this study, we combined a public health approach with genome analysis to provide insight into the correlation of phenotypic characteristics and their genetic basis.
Collapse
Affiliation(s)
- Alex R Hoffmaster
- Epidemiologic Investigations Laboratory, Meningitis and Special Pathogens Branch, Centers for Disease Control and Prevention, 1600 Clifton Road, MS G34, Atlanta, GA 30333, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1392
|
Davidson A, Siddell S. Potential for antiviral treatment of severe acute respiratory syndrome. Curr Opin Infect Dis 2004; 16:565-71. [PMID: 14624107 DOI: 10.1097/00001432-200312000-00009] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Severe acute respiratory syndrome is a new, sometimes lethal disease of humans that is caused by a novel coronavirus. To date there have been over 750 related deaths and there is clearly an urgent need to develop specific antiviral drugs to combat this disease. In this review, the authors shall focus on the molecular biology of the coronavirus and suggest how this information can be used to identify possible targets for antiviral drugs. RECENT FINDINGS Within a remarkably short period of time, the severe acute respiratory syndrome coronavirus has been isolated, its genome has been sequenced and the structure of at least one key viral enzyme has been deduced. In addition, bioinformatic analysis has predicted a number of enzymatic activities associated with proteins of the viral replicase-transcriptase complex. In some cases, these functions have been confirmed by biochemical analysis. Thus, there has been significant progress in the rational approach to anti-severe acute respiratory syndrome coronavirus drug design. This approach, combined with the random screening of licensed compounds or existing compound libraries, should result in the identification of novel lead compounds and the expeditious development of antiviral drugs. SUMMARY Although the initial severe acute respiratory syndrome epidemic has been controlled by conventional measures, the animal reservoir for the coronavirus progenitor has not been identified. It is therefore likely that the virus will be reintroduced into the human population in the future. When this happens, the most economical and effective way to contain the virus will be the therapeutic use of antiviral drugs.
Collapse
Affiliation(s)
- Andrew Davidson
- Department of Pathology and Microbiology, Medical and Veterinary Sciences, University of Bristol, Bristol, UK
| | | |
Collapse
|
1393
|
Abstract
Two camps continue to evolve in the field of structural biology—a 'systems-oriented' camp, which studies proteins or complexes carefully one system at a time, and a 'discovery-oriented' one, which studies proteins of entire families, pathways or genomes. The end goals of both camps are the same: to decipher the atomic-resolution structures and mechanisms of biological macromolecules and understand them in the context of the living cell.
Collapse
Affiliation(s)
- Raymond C Stevens
- Department of Molecular Biology and Chemistry at the Scripps Research Institute, La Jolla, California 92037, USA.
| |
Collapse
|
1394
|
Hensley LE, Fritz EA, Jahrling PB, Karp C, Huggins JW, Geisbert TW. Interferon-beta 1a and SARS coronavirus replication. Emerg Infect Dis 2004; 10:317-9. [PMID: 15030704 PMCID: PMC3322919 DOI: 10.3201/eid1002.030482] [Citation(s) in RCA: 148] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
A global outbreak of severe acute respiratory syndrome (SARS) caused by a novel coronavirus began in March 2003. The rapid emergence of SARS and the substantial illness and death it caused have made it a critical public health issue. Because no effective treatments are available, an intensive effort is under way to identify and test promising antiviral drugs. Here, we report that recombinant human interferon (IFN)-β 1a potently inhibits SARS coronavirus replication in vitro.
Collapse
Affiliation(s)
- Lisa E. Hensley
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA
| | - Elizabeth A. Fritz
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA
| | - Peter B. Jahrling
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA
| | - Christopher Karp
- Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - John W. Huggins
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA
| | - Thomas W. Geisbert
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA
| |
Collapse
|
1395
|
Weingartl HM, Copps J, Drebot MA, Marszal P, Smith G, Gren J, Andova M, Pasick J, Kitching P, Czub M. Susceptibility of pigs and chickens to SARS coronavirus. Emerg Infect Dis 2004; 10:179-84. [PMID: 15030680 PMCID: PMC3322906 DOI: 10.3201/eid1002.030677] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
An outbreak of severe acute respiratory syndrome (SARS) in humans, associated with a new coronavirus, was reported in Southeast Asia, Europe, and North America in early 2003. To address speculations that the virus originated in domesticated animals, or that domestic species were susceptible to the virus, we inoculated 6-week-old pigs and chickens intravenously, intranasally, ocularly, and orally with 106 PFU of SARS-associated coronavirus (SARS-CoV). Clinical signs did not develop in any animal, nor were gross pathologic changes evident on postmortem examinations. Attempts at virus isolation were unsuccessful; however, viral RNA was detected by reverse transcriptase-polymerase chain reaction in blood of both species during the first week after inoculation, and in chicken organs at 2 weeks after inoculation. Virus-neutralizing antibodies developed in the pigs. Our results indicate that these animals do not play a role as amplifying hosts for SARS-CoV.
Collapse
Affiliation(s)
- Hana M Weingartl
- Canadian Science Centre for Human and Animal Health, Winnipeg, Manitoba, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1396
|
Schmidt M, Brixner V, Ruster B, Hourfar MK, Drosten C, Preiser W, Seifried E, Roth WK. NAT screening of blood donors for severe acute respiratory syndrome coronavirus can potentially prevent transfusion associated transmissions. Transfusion 2004; 44:470-5. [PMID: 15043560 PMCID: PMC7201871 DOI: 10.1111/j.1537-2995.2004.03269.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND: The severe acute respiratory syndrome (SARS) was first described in February 2003. Close contact with symptomatic patients appears to be the main route of transmission, whereas blood transfusion trans‐ mission could not be ruled out. STUDY DESIGN AND METHODS: A SARS coronavirus (SARS‐CoV) detection kit developed by C. Drosten (Bernhard Nocht Institute, BNI) was used to amplify SARS‐CoV sequences from blood donor samples. We tested 31,151 blood donor samples in minipools of up to 96 samples. To validate the sensitivity of the assay, routine donor minipools (88 ± 8 samples per pool) were spiked with plasma of an imported case of SARS or of a subsequently infected contact person, respectively. Gamma‐irradiated cell culture supernatants of Vero E6 cells, infected with SARS‐CoV, were used as positive controls. RESULTS: None of 31,151 blood donors were positive for the presence of SARS. Two 96‐member plasma pools that were each spiked with 100 µL of plasma of the German index patient or his wife, respectively, were positive. Overall, 0.85 percent of test results had to be considered invalid owing to negative internal controls. CONCLUSION: A real‐time CoV PCR test is able to detect SARS‐CoV in viremic blood donor samples even in the beginning of the disease when patients present minor clinical symptoms. Thus the assay could potentially help to prevent transfusion‐associated SARS‐CoV transmissions.
Collapse
Affiliation(s)
- Michael Schmidt
- Institute of Transfusion Medicine and Immunohematology, German Red Cross, Johan Wolfgang Goethe University, Frankfurt, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
1397
|
He R, Dobie F, Ballantine M, Leeson A, Li Y, Bastien N, Cutts T, Andonov A, Cao J, Booth TF, Plummer FA, Tyler S, Baker L, Li X. Analysis of multimerization of the SARS coronavirus nucleocapsid protein. Biochem Biophys Res Commun 2004; 316:476-83. [PMID: 15020242 PMCID: PMC7111152 DOI: 10.1016/j.bbrc.2004.02.074] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2004] [Indexed: 12/20/2022]
Abstract
Severe Acute Respiratory Syndrome (SARS), an emerging disease characterized by atypical pneumonia, has recently been attributed to a novel coronavirus. The genome of SARS Coronavirus (SARS-CoV) has recently been sequenced, and a number of genes identified, including that of the nucleocapsid protein (N). It is noted, however, that the N protein of SARS-CoV (SARS-CoV N) shares little homology with nucleocapsid proteins of other members of the coronavirus family [Science 300 (2003) 1399; Science 300 (2003) 1394]. N proteins of other coronavirus have been reported to be involved in forming the viral core and also in the packaging and transcription of the viral RNA. As data generated from some viral systems other than coronaviruses suggested that viral N–N self-interactions may be necessary for subsequent formation of the nucleocapsid and assembly of the viral particles, we decided to investigate SARS-CoV N–N interaction. By using mammalian two-hybrid system and sucrose gradient fractionations, a homotypic interaction of N, but not M, was detected by the two-hybrid analysis. The mammalian two-hybrid assay revealed an approximately 50-fold increase in SEAP activity (measurement of protein–protein interaction) in N–N interaction compared to that observed in either M–M or mock transfection. Furthermore, mutational analyses characterized that a serine/arginine-rich motif (SSRSSSRSRGNSR) between amino acids 184 and 196 is crucial for N protein oligomerization, since deletion of this region completely abolished the N protein self-multimerization. Finally, the full-length nucleocapsid protein expressed and purified from baculovirus system was found to form different levels of higher order structures as detected by Western blot analysis of the fractionated proteins. Collectively, these results may aid us in elucidating the mechanism pertaining to formation of viral nucleocapsid core, and designing molecular approaches to intervene SARS-CoV replication.
Collapse
Affiliation(s)
- Runtao He
- National Microbiology Laboratory, Health Canada, 1015 Arlington St., Winnipeg, MB, Canada R3E 3R2.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1398
|
Bressler AM, Nolte FS. Preclinical evaluation of two real-time, reverse transcription-PCR assays for detection of the severe acute respiratory syndrome coronavirus. J Clin Microbiol 2004; 42:987-91. [PMID: 15004042 PMCID: PMC356893 DOI: 10.1128/jcm.42.3.987-991.2004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
We verified the analytical performance characteristics of a previously described real-time reverse transcription-PCR (RT-PCR) assay targeting the open reading frame (ORF) 1b region of the severe acute respiratory syndrome coronavirus (SARS-CoV) with RNA transcripts. We then compared it to a novel nucleocapsid gene real-time RT-PCR assay with genomic RNA. The assays differed only in the primer and probe sequences and final concentrations. A commercially available armored RNA (Ambion, Austin, Tex.) was evaluated as positive control for the ORF 1b assay. The analytical sensitivity, reproducibility, amplification efficiency, and dynamic range of the assays were similar. Both were specific for SARS-CoV as determined by testing against human CoV 229E and OC43, specimens from patients without SARS, and by BLAST searches of GenBank for primer and probe sequence homology. The armored RNA was found to be a suitable positive control for the ORF 1b assay that could be reliably recovered and amplified from a variety of clinical specimens.
Collapse
Affiliation(s)
- Adam M Bressler
- Department of Medicine (Infectious Diseases), and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | | |
Collapse
|
1399
|
Abstract
Focused efforts by several international laboratories have resulted in the sequencing of the genome of the causative agent of severe acute respiratory syndrome (SARS), novel coronavirus SARS-CoV, in record time. Using cumulative skew diagrams, I found that mutational patterns in the SARS-CoV genome were strikingly different from other coronaviruses in terms of mutation rates, although they were in general agreement with the model of the coronavirus lifecycle. These findings might be relevant for the development of sequence-based diagnostics and the design of agents to treat SARS.
Collapse
|
1400
|
Zhu G, Chen HW. Monophyletic relationship between severe acute respiratory syndrome coronavirus and group 2 coronaviruses. J Infect Dis 2004; 189:1676-8. [PMID: 15116304 PMCID: PMC7109970 DOI: 10.1086/382892] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2003] [Accepted: 10/22/2003] [Indexed: 11/15/2022] Open
Abstract
Although primary genomic analysis has revealed that severe acute respiratory syndrome coronavirus (SARS CoV) is a new type of coronavirus, the different protein trees published in previous reports have provided no conclusive evidence indicating the phylogenetic position of SARS CoV. To clarify the phylogenetic relationship between SARS CoV and other coronaviruses, we compiled a large data set composed of 7 concatenated protein sequences and performed comprehensive analyses, using the maximum-likelihood, Bayesian-inference, and maximum-parsimony methods. All resulting phylogenetic trees displayed an identical topology and supported the hypothesis that the relationship between SARS CoV and group 2 CoVs is monophyletic. Relationships among all major groups were well resolved and were supported by all statistical analyses.
Collapse
Affiliation(s)
- Guan Zhu
- Department of Veterinary Pathobiology, Texas A and M University, College Station, Texas 77843, USA.
| | | |
Collapse
|