101
|
Cui X, Lee LM, Heng X, Zhong W, Sternberg PW, Psaltis D, Yang C. Lensless high-resolution on-chip optofluidic microscopes for Caenorhabditis elegans and cell imaging. Proc Natl Acad Sci U S A 2008; 105:10670-5. [PMID: 18663227 PMCID: PMC2488383 DOI: 10.1073/pnas.0804612105] [Citation(s) in RCA: 178] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2008] [Indexed: 01/09/2023] Open
Abstract
Low-cost and high-resolution on-chip microscopes are vital for reducing cost and improving efficiency for modern biomedicine and bioscience. Despite the needs, the conventional microscope design has proven difficult to miniaturize. Here, we report the implementation and application of two high-resolution (approximately 0.9 microm for the first and approximately 0.8 microm for the second), lensless, and fully on-chip microscopes based on the optofluidic microscopy (OFM) method. These systems abandon the conventional microscope design, which requires expensive lenses and large space to magnify images, and instead utilizes microfluidic flow to deliver specimens across array(s) of micrometer-size apertures defined on a metal-coated CMOS sensor to generate direct projection images. The first system utilizes a gravity-driven microfluidic flow for sample scanning and is suited for imaging elongate objects, such as Caenorhabditis elegans; and the second system employs an electrokinetic drive for flow control and is suited for imaging cells and other spherical/ellipsoidal objects. As a demonstration of the OFM for bioscience research, we show that the prototypes can be used to perform automated phenotype characterization of different Caenorhabditis elegans mutant strains, and to image spores and single cellular entities. The optofluidic microscope design, readily fabricable with existing semiconductor and microfluidic technologies, offers low-cost and highly compact imaging solutions. More functionalities, such as on-chip phase and fluorescence imaging, can also be readily adapted into OFM systems. We anticipate that the OFM can significantly address a range of biomedical and bioscience needs, and engender new microscope applications.
Collapse
Affiliation(s)
- Xiquan Cui
- Departments of *Electrical Engineering and
| | | | - Xin Heng
- Departments of *Electrical Engineering and
| | - Weiwei Zhong
- Division of Biology, California Institute of Technology, Pasadena, CA 91125; and
| | - Paul W. Sternberg
- Division of Biology, California Institute of Technology, Pasadena, CA 91125; and
| | - Demetri Psaltis
- Departments of *Electrical Engineering and
- School of Engineering, Ecole Polytechnique Federale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Changhuei Yang
- Departments of *Electrical Engineering and
- Bioengineering, and
| |
Collapse
|
102
|
Bai B, Hu Q, Hu H, Zhou P, Shi Z, Meng J, Lu B, Huang Y, Mao P, Wang H. Virus-like particles of SARS-like coronavirus formed by membrane proteins from different origins demonstrate stimulating activity in human dendritic cells. PLoS One 2008; 3:e2685. [PMID: 18628832 PMCID: PMC2441860 DOI: 10.1371/journal.pone.0002685] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Accepted: 06/10/2008] [Indexed: 11/19/2022] Open
Abstract
The pathogenesis of SARS coronavirus (CoV) remains poorly understood. In the current study, two recombinant baculovirus were generated to express the spike (S) protein of SARS-like coronavirus (SL-CoV) isolated from bats (vAcBS) and the envelope (E) and membrane (M) proteins of SARS-CoV, respectively. Co-infection of insect cells with these two recombinant baculoviruses led to self-assembly of virus-like particles (BVLPs) as demonstrated by electron microscopy. Incorporation of S protein of vAcBS (BS) into VLPs was confirmed by western blot and immunogold labeling. Such BVLPs up-regulated the level of CD40, CD80, CD86, CD83, and enhanced the secretion of IL-6, IL-10 and TNF-α in immature dendritic cells (DCs). Immune responses were compared in immature DCs inoculated with BVLPs or with VLPs formed by S, E and M proteins of human SARS-CoV. BVLPs showed a stronger ability to stimulate DCs in terms of cytokine induction as evidenced by 2 to 6 fold higher production of IL-6 and TNF-α. Further study indicated that IFN-γ+ and IL-4+ populations in CD4+ T cells increased upon co-cultivation with DCs pre-exposed with BVLPs or SARS-CoV VLPs. The observed difference in DC-stimulating activity between BVLPs and SARS CoV VLPs was very likely due to the S protein. In agreement, SL-CoV S DNA vaccine evoked a more vigorous antibody response and a stronger T cell response than SARS-CoV S DNA in mice. Our data have demonstrated for the first time that SL-CoV VLPs formed by membrane proteins of different origins, one from SL-CoV isolated from bats (BS) and the other two from human SARS-CoV (E and M), activated immature DCs and enhanced the expression of co-stimulatory molecules and the secretion of cytokines. Finding in this study may provide important information for vaccine development as well as for understanding the pathogenesis of SARS-like CoV.
Collapse
Affiliation(s)
- Bingke Bai
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, People's Republic of China
- Department of Virology, Institute of Infectious Disease, Beijing, People's Republic of China
- Chinese People's Liberation Army Postgraduate Medical School, Beijing, People's Republic of China
| | - Qinxue Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Hui Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Peng Zhou
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, People's Republic of China
- Graduate School of the Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Zhengli Shi
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Jin Meng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, People's Republic of China
- Graduate School of the Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Baojing Lu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, People's Republic of China
- Graduate School of the Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Yi Huang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Panyong Mao
- Department of Virology, Institute of Infectious Disease, Beijing, People's Republic of China
| | - Hanzhong Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, People's Republic of China
- * E-mail: .
| |
Collapse
|
103
|
Zhou B, Liu J, Wang Q, Liu X, Li X, Li P, Ma Q, Cao C. The nucleocapsid protein of severe acute respiratory syndrome coronavirus inhibits cell cytokinesis and proliferation by interacting with translation elongation factor 1alpha. J Virol 2008; 82:6962-71. [PMID: 18448518 PMCID: PMC2446950 DOI: 10.1128/jvi.00133-08] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2008] [Accepted: 04/24/2008] [Indexed: 11/20/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV) is the etiological agent of SARS, an emerging disease characterized by atypical pneumonia. Using a yeast two-hybrid screen with the nucleocapsid (N) protein of SARS-CoV as a bait, the C terminus (amino acids 251 to 422) of the N protein was found to interact with human elongation factor 1-alpha (EF1alpha), an essential component of the translational machinery with an important role in cytokinesis, promoting the bundling of filamentous actin (F-actin). In vitro and in vivo interaction was then confirmed by immuno-coprecipitation, far-Western blotting, and surface plasmon resonance. It was demonstrated that the N protein of SARS-CoV induces aggregation of EF1alpha, inhibiting protein translation and cytokinesis by blocking F-actin bundling. Proliferation of human peripheral blood lymphocytes and other human cell lines was significantly inhibited by the infection of recombinant retrovirus expressing SARS-CoV N protein.
Collapse
Affiliation(s)
- Bing Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
104
|
|
105
|
Abstract
Protein-protein interactions play a central role in biological processes and thus are an appealing target for innovative drug design a nd development. They can be targeted bysmall molecule inhibitors, peptides and peptidomimetics, which represent an alternative to protein therapeutics that carry many disadvantages. In this chapter, I describe specific protein-protein interactions suggested by a novel model of immune signaling, the Signaling Chain HOmoOLigomerization (SCHOOL) model, to be critical for cell activation mediated by multichain immune recognition receptors (MIRRs) expressed on different cells of the hematopoietic system. Unraveling a long-standing mystery of MIRR triggering and transmembrane signaling, the SCHOOL model reveals the intrareceptor transmembrane interactions and interreceptor cytoplasmic homointeractions as universal therapeutic targets for a diverse variety of disorders mediated by immune cells. Further, assuming that the general principles underlying MIRR-mediated transmembrane signaling mechanisms are similar, the SCHOOL model can be applied to any particular receptor of the MIRR family. Thus, an important application of the SCHOOL model is that global therapeutic strategies targeting key protein-protein interactions involved in MIRR triggering and transmembrane signal transduction may be used to treat a diverse set of immune-mediated diseases. This assumes that clinical knowledge and therapeutic strategies can be transferred between seemingly disparate disorders, such as T-cell-mediated skin diseases and platelet disorders, or combined to develop novel pharmacological approaches. Intriguingly, the SCHOOL model unravels the molecular mechanisms underlying ability of different human viruses such as human immunodeficiency virus, cytomegalovirus and severe acute respiratory syndrome coronavirus to modulate and/or escape the host immune response. It also demonstrates how the lessons learned from viral pathogenesis can be used practically for rational drug design. Application of this model to platelet collagen receptor signaling has already led to the development of a novel concept of platelet inhibition and the invention of new platelet inhibitors, thus proving the suggested hypothesis and highlighting the importance and broad perspectives of the SCHOOL model in the development of new targeting strategies.
Collapse
|
106
|
Severe acute respiratory syndrome coronavirus as an agent of emerging and reemerging infection. Clin Microbiol Rev 2007; 20:660-94. [PMID: 17934078 DOI: 10.1128/cmr.00023-07] [Citation(s) in RCA: 681] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Before the emergence of severe acute respiratory syndrome (SARS) coronavirus (SARS-CoV) in 2003, only 12 other animal or human coronaviruses were known. The discovery of this virus was soon followed by the discovery of the civet and bat SARS-CoV and the human coronaviruses NL63 and HKU1. Surveillance of coronaviruses in many animal species has increased the number on the list of coronaviruses to at least 36. The explosive nature of the first SARS epidemic, the high mortality, its transient reemergence a year later, and economic disruptions led to a rush on research of the epidemiological, clinical, pathological, immunological, virological, and other basic scientific aspects of the virus and the disease. This research resulted in over 4,000 publications, only some of the most representative works of which could be reviewed in this article. The marked increase in the understanding of the virus and the disease within such a short time has allowed the development of diagnostic tests, animal models, antivirals, vaccines, and epidemiological and infection control measures, which could prove to be useful in randomized control trials if SARS should return. The findings that horseshoe bats are the natural reservoir for SARS-CoV-like virus and that civets are the amplification host highlight the importance of wildlife and biosecurity in farms and wet markets, which can serve as the source and amplification centers for emerging infections.
Collapse
|
107
|
Guo Y, Korteweg C, McNutt MA, Gu J. Pathogenetic mechanisms of severe acute respiratory syndrome. Virus Res 2007; 133:4-12. [PMID: 17825937 PMCID: PMC7114157 DOI: 10.1016/j.virusres.2007.01.022] [Citation(s) in RCA: 136] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2006] [Revised: 12/23/2006] [Accepted: 01/09/2007] [Indexed: 02/08/2023]
Abstract
Severe acute respiratory syndrome (SARS) is an acute respiratory disease with significant morbidity and mortality. While its clinical manifestations have been extensively studied, its pathogenesis is not yet fully understood. A limited number of autopsy studies have revealed that the lungs and the immune system are the organs that sustain the most severe damage. Other organs affected include the kidneys, brain, digestive tract, heart, liver, thyroid gland and urogenital tract. The primary target cells are pneumocytes and enterocytes, both cell types abundantly expressing angiotensin-converting enzyme 2 which is the main SARS-CoV receptor. Other cell types infected include the epithelial cells of renal tubules, cerebral neurons, and immune cells. The pathology of this disease results from both direct and indirect injury. Direct injury is caused by infection of the target cells by the virus. Indirect injury mainly results from immune responses, circulatory dysfunction, and hypoxia. In this review, we summarize the major pathological findings at the gross, cellular and molecular levels and discuss the various possible mechanisms that may contribute to the pathogenesis of SARS. The implications of the proposed pathogenesis for prevention, diagnosis and therapy of the disease are discussed.
Collapse
Affiliation(s)
| | | | | | - Jiang Gu
- Corresponding author at: Department of Pathology, School of Medical Sciences, Infectious Disease Center, Peking (Beijing) University, 38 Xueyuan Road, 100083 Beijing, China. Tel.: +86 10 8280 1237; fax: +86 10 8280 1380.
| |
Collapse
|
108
|
Chan KYK, Ching JCY, Xu MS, Cheung ANY, Yip SP, Yam LYC, Lai ST, Chu CM, Wong ATY, Song YQ, Huang FP, Liu W, Chung PH, Leung GM, Chow EYD, Chan EYT, Chan JCK, Ngan H, Tam P, Chan LC, Sham P, Chan VSF, Peiris M, Lin SCL, Khoo US. Association of ICAM3 genetic variant with severe acute respiratory syndrome. J Infect Dis 2007; 196:271-80. [PMID: 17570115 PMCID: PMC7202406 DOI: 10.1086/518892] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2005] [Accepted: 02/16/2007] [Indexed: 12/12/2022] Open
Abstract
Genetic polymorphisms have been demonstrated to be associated with vulnerability to human infection. ICAM3, an intercellular adhesion molecule important for T cell activation, and FCER2 (CD23), an immune response gene, both located on chromosome 19p13.3, were investigated for host genetic susceptibility and association with clinical outcome. A case-control study based on 817 patients with confirmed severe acute respiratory syndrome (SARS), 307 health care worker control subjects, 290 outpatient control subjects, and 309 household control subjects unaffected by SARS from Hong Kong was conducted to test for genetic association. No significant association to susceptibility to SARS infection caused by the novel coronavirus (SARS-CoV) was found for the FCER2 and the ICAM3 single nucleotide polymorphisms. However, patients with SARS homozygous for ICAM3 Gly143 showed significant association with higher lactate dehydrogenase levels (P=.0067; odds ratio [OR], 4.31 [95% confidence interval {CI}, 1.37-13.56]) and lower total white blood cell counts (P=.022; OR, 0.30 [95% CI, 0.10-0.89]) on admission. These findings support the role of ICAM3 in the immunopathogenesis of SARS.
Collapse
Affiliation(s)
- Kelvin Y. K. Chan
- Department of Pathology, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - Johannes C. Y. Ching
- Department of Pathology, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - M. S. Xu
- Department of Pathology, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - Annie N. Y. Cheung
- Department of Pathology, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - Shea-Ping Yip
- Department of Health Technology and Informatics, Hong Kong Polytechnic UniversityHong Kong
| | | | | | | | | | - You-Qiang Song
- Department of Biochemistry, Hong Kong Jockey Club Clinical Research CentreHong Kong
- Genome Research Center, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - Fang-Ping Huang
- Department of Pathology, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - Wei Liu
- Department of Pathology, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | | | - G. M. Leung
- Department of Community Medicine, Li Ka Shing Faculty of Medicine, University of Hong KongHong Kong
| | | | - Eric Y. T. Chan
- Department of Pathology, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - Jane C. K. Chan
- Hospital Authority Severe Acute Respiratory Syndrome Collaborative Group, Hong Kong Hospital Authority Head OfficeHong Kong
| | - Hextan Ngan
- Department of Obstetrics and Gynecology, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - Paul Tam
- Department of Surgery, Hong Kong Jockey Club Clinical Research CentreHong Kong
- Genome Research Center, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - Li-Chong Chan
- Department of Pathology, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - Pak Sham
- Department of Psychiatry, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - Vera S. F. Chan
- Department of Biosurgery and Surgical Technology, Imperial College LondonLondon, United Kingdom
| | - Malik Peiris
- Department of Microbiology, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - Steve C. L. Lin
- Department of Biosurgery and Surgical Technology, Imperial College LondonLondon, United Kingdom
| | - Ui-Soon Khoo
- Department of Pathology, Hong Kong Jockey Club Clinical Research CentreHong Kong
| |
Collapse
|
109
|
Enjuanes L, Almazán F, Sola I, Zuñiga S. Biochemical aspects of coronavirus replication and virus-host interaction. Annu Rev Microbiol 2006; 60:211-30. [PMID: 16712436 DOI: 10.1146/annurev.micro.60.080805.142157] [Citation(s) in RCA: 160] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Infection by different coronaviruses (CoVs) causes alterations in the transcriptional and translational patterns, cell cycle, cytoskeleton, and apoptosis pathways of the host cells. In addition, CoV infection may cause inflammation, alter immune and stress responses, and modify the coagulation pathways. The balance between the up- and downregulated genes could explain the pathogenesis caused by these viruses. We review specific aspects of CoV-host interactions. CoV genome replication takes place in the cytoplasm in a membrane-protected microenvironment and may control the cell machinery by locating some of their proteins in the host cell nucleus. CoVs initiate translation by cap-dependent and cap-independent mechanisms. CoV transcription involves a discontinuous RNA synthesis (template switching) during the extension of a negative copy of the subgenomic mRNAs. The requirement for base-pairing during transcription has been formally demonstrated in arteriviruses and CoVs. CoV N proteins have RNA chaperone activity that may help initiate template switching. Both viral and cellular proteins are required for replication and transcription, and the role of selected proteins is addressed.
Collapse
Affiliation(s)
- Luis Enjuanes
- Department of Molecular and Cell Biology, CNB, CSIC, 28049 Madrid, Spain.
| | | | | | | |
Collapse
|
110
|
Lawler JV, Endy TP, Hensley LE, Garrison A, Fritz EA, Lesar M, Baric RS, Kulesh DA, Norwood DA, Wasieloski LP, Ulrich MP, Slezak TR, Vitalis E, Huggins JW, Jahrling PB, Paragas J. Cynomolgus macaque as an animal model for severe acute respiratory syndrome. PLoS Med 2006; 3:e149. [PMID: 16605302 PMCID: PMC1435788 DOI: 10.1371/journal.pmed.0030149] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2005] [Accepted: 01/10/2006] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The emergence of severe acute respiratory syndrome (SARS) in 2002 and 2003 affected global health and caused major economic disruption. Adequate animal models are required to study the underlying pathogenesis of SARS-associated coronavirus (SARS-CoV) infection and to develop effective vaccines and therapeutics. We report the first findings of measurable clinical disease in nonhuman primates (NHPs) infected with SARS-CoV. METHODS AND FINDINGS In order to characterize clinically relevant parameters of SARS-CoV infection in NHPs, we infected cynomolgus macaques with SARS-CoV in three groups: Group I was infected in the nares and bronchus, group II in the nares and conjunctiva, and group III intravenously. Nonhuman primates in groups I and II developed mild to moderate symptomatic illness. All NHPs demonstrated evidence of viral replication and developed neutralizing antibodies. Chest radiographs from several animals in groups I and II revealed unifocal or multifocal pneumonia that peaked between days 8 and 10 postinfection. Clinical laboratory tests were not significantly changed. Overall, inoculation by a mucosal route produced more prominent disease than did intravenous inoculation. Half of the group I animals were infected with a recombinant infectious clone SARS-CoV derived from the SARS-CoV Urbani strain. This infectious clone produced disease indistinguishable from wild-type Urbani strain. CONCLUSIONS SARS-CoV infection of cynomolgus macaques did not reproduce the severe illness seen in the majority of adult human cases of SARS; however, our results suggest similarities to the milder syndrome of SARS-CoV infection characteristically seen in young children.
Collapse
Affiliation(s)
- James V Lawler
- 1Infectious Diseases Department, National Naval Medical Center (NNMC), Bethesda, Maryland, United States of America
| | - Timothy P Endy
- 2Virology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Maryland, United States of America
| | - Lisa E Hensley
- 2Virology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Maryland, United States of America
| | - Aura Garrison
- 2Virology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Maryland, United States of America
| | - Elizabeth A Fritz
- 2Virology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Maryland, United States of America
| | - May Lesar
- 3Radiology Division, National Naval Medical Center (NNMC); Bethesda, Maryland, United States of America
| | - Ralph S Baric
- 4Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - David A Kulesh
- 5Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Maryland, United States of America
| | - David A Norwood
- 5Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Maryland, United States of America
| | - Leonard P Wasieloski
- 5Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Maryland, United States of America
| | - Melanie P Ulrich
- 5Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Maryland, United States of America
| | - Tom R Slezak
- 6Lawrence Livermore National Laboratory, Livermore, California, United States of America
| | - Elizabeth Vitalis
- 6Lawrence Livermore National Laboratory, Livermore, California, United States of America
| | - John W Huggins
- 2Virology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Maryland, United States of America
| | - Peter B Jahrling
- 7Headquarters Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Maryland, United States of America
| | - Jason Paragas
- 2Virology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Maryland, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
111
|
Perlman S, Dandekar AA. Glucocorticoid contribution to lymphopaenia and immunpathology in patients with SARS. Nat Rev Immunol 2006. [PMCID: PMC7096955 DOI: 10.1038/nri1835-c2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
112
|
Poccia F, Agrati C, Castilletti C, Bordi L, Gioia C, Horejsh D, Ippolito G, Chan PKS, Hui DSC, Sung JJY, Capobianchi MR, Malkovsky M. Anti-severe acute respiratory syndrome coronavirus immune responses: the role played by V gamma 9V delta 2 T cells. J Infect Dis 2006; 193:1244-9. [PMID: 16586361 PMCID: PMC7110256 DOI: 10.1086/502975] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2005] [Accepted: 11/29/2005] [Indexed: 01/14/2023] Open
Abstract
Severe acute respiratory syndrome (SARS) is caused by a novel coronavirus (SARS-CoV) strain. Analyses of T cell repertoires in health care workers who survived SARS-CoV infection during the 2003 outbreak revealed that their effector memory Vγ9Vδ2 T cell populations were selectively expanded ∼3 months after the onset of disease. No such expansion of their αβ T cell pools was detected. The expansion of the Vγ9Vδ2 T cell population was associated with higher anti–SARS-CoV immunoglobulin G titers. In addition, in vitro experiments demonstrated that stimulated Vγ9Vδ2 T cells display an interferon-γ–dependent anti–SARS-CoV activity and are able to directly kill SARS-CoV–infected target cells. These findings are compatible with the possibility that Vγ9Vδ2 T cells play a protective role during SARS
Collapse
Affiliation(s)
- Fabrizio Poccia
- Unit of Cellular Immunology, National Institute for Infectious Diseases Lazzaro Spallanzani, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
113
|
Taylor DR. Obstacles and advances in SARS vaccine development. Vaccine 2006; 24:863-71. [PMID: 16191455 PMCID: PMC7115537 DOI: 10.1016/j.vaccine.2005.08.102] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2005] [Revised: 07/29/2005] [Accepted: 08/25/2005] [Indexed: 02/08/2023]
Abstract
The emergence of the severe acute respiratory syndrome (SARS) that resulted in a pandemic in 2003 spurred a flurry of interest in the development of vaccines to prevent and treat the potentially deadly viral infection. Researchers around the world pooled their scientific resources and shared early data in an unprecedented manner in light of the impending public health crisis. There are still large gaps in knowledge about the pathogenesis of this virus. While significant advances have been made in the development of animal models, the practicality of their use may be hampered by a lack of pathological similarity with human disease. Described here are issues related to progress in vaccine development and the obstacles that lie ahead for both researchers and regulatory agencies.
Collapse
Affiliation(s)
- Deborah R Taylor
- Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research (CBER), US Food and Drug Administration, Bethesda, MD 20892, USA.
| |
Collapse
|
114
|
Abstract
The world was shocked in early 2003 when a pandemic of severe acute respiratory syndrome (SARS) was imminent. The outbreak of this novel disease, caused by a novel coronavirus (the SARS-coronavirus), hit hardest in the Asian Pacific region, though eventually it spread to five continents. The speed of the spread of the SARS epidemic was unprecedented due to the highly efficient intercontinental transportation. An international collaborative effort through the World Health Organization (WHO) has helped to identify the aetiological agent about 1 month after the onset of the epidemic. The power of molecular biology and bioinformatics has enabled the complete decoding of the viral genome within weeks. Over 1000 publications on the phylogeny, epidemiology, genomics, laboratory diagnostics, antiviral, immunization, pathogenesis, clinical disease, and management accumulated within just 1 year. Although the exact animal reservoir of virus and how it evolved into a human pathogen are still obscure, accurate diagnosis and epidemiological control of the disease are now possible. This article reviews what is currently known about the virus and the disease.
Collapse
Affiliation(s)
- Samson S. Y. Wong
- Department of Microbiology, The University of Hong Kong, University Pathology Building, Queen Mary Hospital, Pokfulam Road, Hong Kong
| | - K. Y. Yuen
- Department of Microbiology, The University of Hong Kong, University Pathology Building, Queen Mary Hospital, Pokfulam Road, Hong Kong
| |
Collapse
|
115
|
Hänel K, Stangler T, Stoldt M, Willbold D. Solution structure of the X4 protein coded by the SARS related coronavirus reveals an immunoglobulin like fold and suggests a binding activity to integrin I domains. J Biomed Sci 2005; 13:281-93. [PMID: 16328780 PMCID: PMC7089389 DOI: 10.1007/s11373-005-9043-9] [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: 07/09/2005] [Accepted: 10/11/2005] [Indexed: 02/08/2023] Open
Abstract
The SARS related Coronavirus genome contains a variety of novel accessory genes. One of these, called ORF7a or ORF8, code for a protein, known as 7a, U122 or X4. We set out to determine the three-dimensional structure of the soluble ectodomain of this type-I transmembrane protein by nuclear magnetic resonance spectroscopy. The fold of the protein is the first member of a further variation of the immunoglobulin like beta-sandwich fold. Because X4 does not reveal significant sequence homologies to proteins in the data bases, we carried out a structure based similarity search for proteins with known function. High structural similarity to Dl domains of ICAM-1 and ICAM-2, and common features in amino acid sequence between X4 and ICAM-1, suggest X4 to possess binding activity for the alpha(L) integrin I domain of LFA-1. Further, based on this structure based prediction, potential functions of X4 in virus replication and pathogenesis are discussed.
Collapse
Affiliation(s)
- Karen Hänel
- Forschungszentrum Jülich, Institut für Biologische Informationsverarbeitung (IBI-2), 52425 Jülich, Germany
- Heinrich-Heine-Universität, Institut für Physikalische Biologie and BMFZ, 40225 Düsseldorf, Germany
| | - Thomas Stangler
- Forschungszentrum Jülich, Institut für Biologische Informationsverarbeitung (IBI-2), 52425 Jülich, Germany
- Heinrich-Heine-Universität, Institut für Physikalische Biologie and BMFZ, 40225 Düsseldorf, Germany
| | - Matthias Stoldt
- Forschungszentrum Jülich, Institut für Biologische Informationsverarbeitung (IBI-2), 52425 Jülich, Germany
- Heinrich-Heine-Universität, Institut für Physikalische Biologie and BMFZ, 40225 Düsseldorf, Germany
| | - Dieter Willbold
- Forschungszentrum Jülich, Institut für Biologische Informationsverarbeitung (IBI-2), 52425 Jülich, Germany
- Heinrich-Heine-Universität, Institut für Physikalische Biologie and BMFZ, 40225 Düsseldorf, Germany
| |
Collapse
|
116
|
Zhang Z, Xu D, Li Y, Jin L, Shi M, Wang M, Zhou X, Wu H, Gao GF, Wang FS. Longitudinal alteration of circulating dendritic cell subsets and its correlation with steroid treatment in patients with severe acute respiratory syndrome. Clin Immunol 2005; 116:225-35. [PMID: 15964242 PMCID: PMC7106242 DOI: 10.1016/j.clim.2005.04.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2004] [Revised: 04/15/2005] [Accepted: 04/17/2005] [Indexed: 02/04/2023]
Abstract
In this study, we found that 74 patients with severe acute respiratory syndrome (SARS) exhibited a rapid, dramatic decrease in numbers of circulating myeloid and plasmacytoid dendritic cells (mDCs and pDCs) during the first 2 weeks of illness (5.3- and 28.4-fold reductions for mDCs and pDCs compared with 25 healthy individuals, respectively), with slow return to normal cell numbers during convalescence (weeks 5–7 of illness on average). In addition, numbers of circulating CD4 and CD8 T cells exhibited milder reductions (2.1- and 1.8-fold at week 1) and earlier return to normal at a mean of weeks 3 and 4, respectively. A significant inverse correlation was found between numbers of DC and T-cell subsets and high-dose steroid treatment. Our novel findings thus suggest that the acute SARS-coronavirus infection probably contributes to the initial reduction of DC and T-cell subsets in blood, and that high-dose steroid administration may subsequently exacerbate and prolong low expression of the cell subsets. These findings will aid the framing of further studies of the immunopathogenesis of SARS.
Collapse
Affiliation(s)
- Zheng Zhang
- Research Centre for Biological Therapy, Beijing Institute of Infectious Diseases, Beijing 302 Hospital, 100 Xi Si Huan Middle Road, Beijing 100039, China
| | - Dongping Xu
- Research Centre for Biological Therapy, Beijing Institute of Infectious Diseases, Beijing 302 Hospital, 100 Xi Si Huan Middle Road, Beijing 100039, China
| | - Yonggang Li
- Research Centre for Biological Therapy, Beijing Institute of Infectious Diseases, Beijing 302 Hospital, 100 Xi Si Huan Middle Road, Beijing 100039, China
| | - Lei Jin
- Research Centre for Biological Therapy, Beijing Institute of Infectious Diseases, Beijing 302 Hospital, 100 Xi Si Huan Middle Road, Beijing 100039, China
| | - Ming Shi
- Research Centre for Biological Therapy, Beijing Institute of Infectious Diseases, Beijing 302 Hospital, 100 Xi Si Huan Middle Road, Beijing 100039, China
| | - Min Wang
- Research Centre for Biological Therapy, Beijing Institute of Infectious Diseases, Beijing 302 Hospital, 100 Xi Si Huan Middle Road, Beijing 100039, China
| | - Xianzhi Zhou
- Research Centre for Biological Therapy, Beijing Institute of Infectious Diseases, Beijing 302 Hospital, 100 Xi Si Huan Middle Road, Beijing 100039, China
| | - Hao Wu
- Beijing You'an Hospital, Capital Medical University, Beijing 100054, China
| | - George F. Gao
- Institute of Microbiology, Chinese Academy of Sciences, Beijing 100080, China
| | - Fu-Sheng Wang
- Research Centre for Biological Therapy, Beijing Institute of Infectious Diseases, Beijing 302 Hospital, 100 Xi Si Huan Middle Road, Beijing 100039, China
- Corresponding author. Fax: +86 10 6383 1870.
| |
Collapse
|
117
|
Xu J, Zhong S, Liu J, Li L, Li Y, Wu X, Li Z, Deng P, Zhang J, Zhong N, Ding Y, Jiang Y. Detection of severe acute respiratory syndrome coronavirus in the brain: potential role of the chemokine mig in pathogenesis. Clin Infect Dis 2005; 41:1089-96. [PMID: 16163626 PMCID: PMC7107994 DOI: 10.1086/444461] [Citation(s) in RCA: 384] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2005] [Accepted: 06/14/2005] [Indexed: 01/08/2023] Open
Abstract
Background. Previous studies have shown that common human coronavirus might be neurotropic, although it was first isolated as a pathogen of the respiratory tract. We noticed that a few patients with severe acute respiratory syndrome (SARS) experienced central nervous symptoms during the course of illness. In the present study, we isolated a SARS coronavirus strain from a brain tissue specimen obtained from a patient with SARS with significant central nervous symptoms. Methods. Using transmission electronic microscopy and nested reverse transcription–polymerase chain reaction, the causative pathogen was identified in cultures of a brain tissue specimen obtained from the patient with SARS. Histopathologic examination of the brain tissue was performed using the methods of immunohistochemistry analysis and double immunofluorescence staining. Fifteen cytokines and chemokines were detected in the blood of the patient with SARS by means of a bead-based multiassay system. Results. A fragment specific for SARS human coronavirus was amplified from cultures of the brain suspension, and transmission electronic microscopy revealed the presence of an enveloped virus morphologically compatible with a coronavirus isolated in the cultures. Pathologic examination of the brain tissue revealed necrosis of neuron cells and broad hyperplasia of gliocytes. Immunostaining demonstrated that monokine induced by interferon-Γ (Mig) was expressed in gliocytes with the infiltration of CD68+ monocytes/macrophages and CD3+ T lymphocytes in the brain mesenchyme. Cytokine/chemokine assay revealed that levels of interferon-Γ–inducible protein 10 and Mig in the blood were highly elevated, although the levels of other cytokines and chemokines were close to normal. Conclusions. This study provides direct evidence that SARS human coronavirus is capable of infecting the central nervous system, and that Mig might be involved in the brain immunopathology of SARS.
Collapse
Affiliation(s)
- Jun Xu
- Guangzhou Institute of Respiratory Diseases, Guangzhou, People's Republic of China
| | - Shuqing Zhong
- Guangzhou Institute of Respiratory Diseases, Guangzhou, People's Republic of China
| | - Jinghua Liu
- Key Laboratory of Functional Proteomics of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| | - Li Li
- Guangzhou Institute of Respiratory Diseases, Guangzhou, People's Republic of China
| | - Yong Li
- Guangzhou Institute of Respiratory Diseases, Guangzhou, People's Republic of China
| | - Xinwei Wu
- Guangzhou Center for Diseases Control and Prevention, Guangzhou, People's Republic of China
| | - Zhijie Li
- Key Laboratory of Functional Proteomics of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| | - Peng Deng
- Key Laboratory of Functional Proteomics of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| | - Jingqiang Zhang
- Electronic Microscope Center, Sun Yatsen University, Guangzhou, People's Republic of China
| | - Nanshan Zhong
- Guangzhou Institute of Respiratory Diseases, Guangzhou, People's Republic of China
| | - Yanqing Ding
- Key Laboratory of Functional Proteomics of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| | - Yong Jiang
- Key Laboratory of Functional Proteomics of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
- Reprints or correspondence: Dr. Yong Jiang, Key Laboratory of Functional, Proteomics of Guangdong Province, Dept. of Pathophysiology, Southern Medical University, Tonghe, Guangzhou 510515, People's Republic of China ()
| |
Collapse
|
118
|
Gu J, Gong E, Zhang B, Zheng J, Gao Z, Zhong Y, Zou W, Zhan J, Wang S, Xie Z, Zhuang H, Wu B, Zhong H, Shao H, Fang W, Gao D, Pei F, Li X, He Z, Xu D, Shi X, Anderson VM, Leong ASY. Multiple organ infection and the pathogenesis of SARS. J Exp Med 2005; 202:415-24. [PMID: 16043521 PMCID: PMC2213088 DOI: 10.1084/jem.20050828] [Citation(s) in RCA: 1101] [Impact Index Per Article: 55.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2005] [Accepted: 06/16/2005] [Indexed: 11/04/2022] Open
Abstract
After >8,000 infections and >700 deaths worldwide, the pathogenesis of the new infectious disease, severe acute respiratory syndrome (SARS), remains poorly understood. We investigated 18 autopsies of patients who had suspected SARS; 8 cases were confirmed as SARS. We evaluated white blood cells from 22 confirmed SARS patients at various stages of the disease. T lymphocyte counts in 65 confirmed and 35 misdiagnosed SARS cases also were analyzed retrospectively. SARS viral particles and genomic sequence were detected in a large number of circulating lymphocytes, monocytes, and lymphoid tissues, as well as in the epithelial cells of the respiratory tract, the mucosa of the intestine, the epithelium of the renal distal tubules, the neurons of the brain, and macrophages in different organs. SARS virus seemed to be capable of infecting multiple cell types in several organs; immune cells and pulmonary epithelium were identified as the main sites of injury. A comprehensive theory of pathogenesis is proposed for SARS with immune and lung damage as key features.
Collapse
Affiliation(s)
- Jiang Gu
- Department of Pathology, Peking University, Beijing, China 100083.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
119
|
Skowronski DM, Astell C, Brunham RC, Low DE, Petric M, Roper RL, Talbot PJ, Tam T, Babiuk L. Severe acute respiratory syndrome (SARS): a year in review. Annu Rev Med 2005; 56:357-81. [PMID: 15660517 DOI: 10.1146/annurev.med.56.091103.134135] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Severe acute respiratory syndrome (SARS) emerged from China as an untreatable and rapidly spreading respiratory illness of unknown etiology. Following point source exposure in February 2003, more than a dozen guests infected at a Hong Kong hotel seeded multi-country outbreaks that persisted through the spring of 2003. The World Health Organization responded by invoking traditional public health measures and advanced technologies to control the illness and contain the cause. A novel coronavirus was implicated and its entire genome was sequenced by mid-April 2003. The urgency of responding to this threat focused scientific endeavor and stimulated global collaboration. Through real-time application of accumulating knowledge, the world proved capable of arresting the first pandemic threat of the twenty-first century, despite early respiratory-borne spread and global susceptibility. This review synthesizes lessons learned from this remarkable achievement. These lessons can be applied to re-emergence of SARS or to the next pandemic threat to arise.
Collapse
Affiliation(s)
- Danuta M Skowronski
- University of British Columbia Center for Disease Control, Vancouver, British Columbia, Canada V5Z 4R4.
| | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Chen H, Hou J, Jiang X, Ma S, Meng M, Wang B, Zhang M, Zhang M, Tang X, Zhang F, Wan T, Li N, Yu Y, Hu H, Yang R, He W, Wang X, Cao X. Response of memory CD8+ T cells to severe acute respiratory syndrome (SARS) coronavirus in recovered SARS patients and healthy individuals. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2005; 175:591-598. [PMID: 15972696 DOI: 10.4049/jimmunol.175.1.591] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
To date, the pathogenesis of severe acute respiratory syndrome (SARS) in humans is still not well understood. SARS coronavirus (SARS-CoV)-specific CTL responses, in particular their magnitude and duration of postinfection immunity, have not been extensively studied. In this study, we found that heat-inactivated SARS-CoV elicited recall CTL responses to newly identified spike protein-derived epitopes (SSp-1, S978, and S1202) in peripheral blood of all HLA-A*0201(+) recovered SARS patients over 1 year postinfection. Intriguingly, heat-inactivated SARS-CoV elicited recall-like CTL responses to SSp-1 but not to S978, S1202, or dominant epitopes from several other human viruses in 5 of 36 (13.8%) HLA-A*0201(+) healthy donors without any contact history with SARS-CoV. SSp-1-specific CTLs expanded from memory T cells of both recovered SARS patients, and the five exceptional healthy donors shared a differentiated effector CTL phenotype, CD45RA(+)CCR7(-)CD62L(-), and expressed CCR5 and CD44. However, compared with the high avidity of SSp-1-specific CTLs derived from memory T cells of recovered SARS patients, SSp-1-specific CTLs from the five exceptional healthy donors were of low avidity, as determined by their rapid tetramer dissociation kinetics and reduced cytotoxic reactivity, IFN-gamma secretion, and intracellular production of IFN-gamma, TNF-alpha, perforin, and granzyme A. These results indicate that SARS-CoV infection induces strong and long-lasting CTL-mediated immunity in surviving SARS patients, and that cross-reactive memory T cells to SARS-CoV may exist in the T cell repertoire of a small subset of healthy individuals and can be reactivated by SARS-CoV infection.
Collapse
Affiliation(s)
- Huabiao Chen
- Institute of Immunology, Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
121
|
Liang Y, Wan Y, Qiu LW, Zhou J, Ni B, Guo B, Zou Q, Zou L, Zhou W, Jia Z, Che XY, Wu Y. Comprehensive antibody epitope mapping of the nucleocapsid protein of severe acute respiratory syndrome (SARS) coronavirus: insight into the humoral immunity of SARS. Clin Chem 2005; 51:1382-96. [PMID: 15976093 PMCID: PMC7108199 DOI: 10.1373/clinchem.2005.051045] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background: The epidemic outbreak of severe acute respiratory syndrome (SARS) posed a worldwide threat to public health and economic stability. Although the pandemic has been contained, concerns over its recurrence remain. It is essential to identify specific diagnostic agents and antiviral vaccine candidates to fight this highly contagious disease. Methods: We generated 14 monoclonal antibodies (mAbs) specific to the SARS coronavirus (SARS-CoV) nucleocapsid (N) protein and used these to thoroughly map the N protein antigenic determinants. We identified the immunodominant antigenic sites responsible for the antibodies in sera from SARS patients and antisera from small animals and differentiated the linear from the conformational antibody-combining sites comprising the natural epitopes by use of yeast surface display. Results: We identified 5 conformational and 3 linear epitopes within the entire N protein; 3 conformational and 3 linear epitopes were immunodominant. The antibody responses to the N protein fragments in mammalian sera revealed that 3 regions of the N protein are strong antigenic domains. We expanded the specificity of the N protein epitope and identified 4 novel conformational epitopes (amino acids 1–69, 68–213, 212–341, and 337–422). Conclusion: The antigenic structures identified for the SARS-CoV N protein, the epitope-specific mAbs, and the serum antibody profile in SARS patients have potential use in the clinical diagnosis and understanding of the protective immunity to SARS-CoV.
Collapse
Affiliation(s)
- Yunfei Liang
- The Institute of Immunology, PLA, The Third Military Medical University, Shapingba District, Chongqing, China
| | - Ying Wan
- The Institute of Immunology, PLA, The Third Military Medical University, Shapingba District, Chongqing, China
| | - Li-wen Qiu
- Central Laboratory, Zhujiang Hospital, The Southern Medical University, Guangzhou, China
| | - Jingran Zhou
- The Institute of Immunology, PLA, The Third Military Medical University, Shapingba District, Chongqing, China
| | - Bing Ni
- The Institute of Immunology, PLA, The Third Military Medical University, Shapingba District, Chongqing, China
| | - Bo Guo
- The Institute of Immunology, PLA, The Third Military Medical University, Shapingba District, Chongqing, China
| | - Qiang Zou
- The Institute of Immunology, PLA, The Third Military Medical University, Shapingba District, Chongqing, China
| | - Liyun Zou
- The Institute of Immunology, PLA, The Third Military Medical University, Shapingba District, Chongqing, China
| | - Wei Zhou
- The Institute of Immunology, PLA, The Third Military Medical University, Shapingba District, Chongqing, China
| | - Zhengcai Jia
- The Institute of Immunology, PLA, The Third Military Medical University, Shapingba District, Chongqing, China
| | - Xiao-yan Che
- Central Laboratory, Zhujiang Hospital, The Southern Medical University, Guangzhou, China
- Address correspondence to Yuzhang Wu at: The Institute of Immunology, PLA, The Third Military Medical University, Shapingba District, Chongqing 400038, People’s Republic of China; fax 086-023-68752789; e-mail ; or Xiao-yan Che at: Central Laboratory, Zhujiang Hospital, The Southern Medical University, Guangzhou 510282, People’s Republic of China; fax 086-020-61643592; e-mail
| | - Yuzhang Wu
- The Institute of Immunology, PLA, The Third Military Medical University, Shapingba District, Chongqing, China
- Address correspondence to Yuzhang Wu at: The Institute of Immunology, PLA, The Third Military Medical University, Shapingba District, Chongqing 400038, People’s Republic of China; fax 086-023-68752789; e-mail ; or Xiao-yan Che at: Central Laboratory, Zhujiang Hospital, The Southern Medical University, Guangzhou 510282, People’s Republic of China; fax 086-020-61643592; e-mail
| |
Collapse
|
122
|
Muller MP, Tomlinson G, Marrie TJ, Tang P, McGeer A, Low DE, Detsky AS, Gold WL. Can routine laboratory tests discriminate between severe acute respiratory syndrome and other causes of community-acquired pneumonia? Clin Infect Dis 2005; 40:1079-86. [PMID: 15791504 PMCID: PMC7107805 DOI: 10.1086/428577] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2004] [Accepted: 11/10/2004] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND The clinical presentation of severe acute respiratory syndrome (SARS) resembles that of other etiologies of community-acquired pneumonia, making diagnosis difficult. Hematological and biochemical abnormalities, particularly lymphopenia, are common in patients with SARS. METHODS With the use of 2 databases, we compared the ability of the absolute lymphocyte count, absolute neutrophil count, lactate dehydrogenase level, creatine kinase level, alanine aminotransferase level, and serum calcium level at hospital admission to discriminate between cases of SARS and cases of community-acquired pneumonia. The SARS database contained data for 144 patients with SARS from the 2003 Toronto SARS outbreak. The community-acquired pneumonia database contained data for 8044 patients with community-acquired pneumonia from Edmonton, Canada. Patients from the SARS database were matched to patients from the community-acquired pneumonia database according to age, and receiver operating characteristic curves were constructed for each laboratory variable. RESULTS The areas under the receiver operating characteristic curves (AUCs) demonstrated fair to poor discriminatory ability for all laboratory variables tested except absolute neutrophil count, which had an AUC of 0.80, indicating good discriminatory ability (although there was no cutoff value of the absolute neutrophil count at which reasonable sensitivity or specificity could be obtained). Combinations of any 2 tests did not perform significantly better than did the absolute neutrophil count alone. CONCLUSIONS Routine laboratory tests, including determination of absolute lymphocyte count, should not be used in the diagnosis of SARS or incorporated into current case definitions of SARS. The role of the absolute neutrophil count in SARS diagnosis is likely limited, but it should be assessed further.
Collapse
Affiliation(s)
- Matthew P. Muller
- Departments of Medicine Health Policy, Toronto, Ontario
- Departments of Medicine Health Policy, Management, and Evaluation, Toronto, Ontario
- Mount Sinai Hospital, Toronto, Ontario
- Reprints or correspondence: Dr. Wayne L. Gold, University Health Network, 200 Elizabeth St., 9ES 407, Toronto, Ontario, Canada, M5G 2C4 ()
| | - George Tomlinson
- Departments of Medicine Health Policy, Toronto, Ontario
- Departments of Medicine Health Policy, Management, and Evaluation, Toronto, Ontario
- Mount Sinai Hospital, Toronto, Ontario
- University Health Network, Toronto, Ontario
| | | | - Patrick Tang
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario
| | - Allison McGeer
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario
- Mount Sinai Hospital, Toronto, Ontario
| | - Donald E. Low
- Departments of Medicine Health Policy, Toronto, Ontario
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario
- Toronto Medical Laboratories and Mount Sinai Hospital Department of Microbiology, Toronto, Ontario
| | - Allan S. Detsky
- Departments of Medicine Health Policy, Toronto, Ontario
- Departments of Medicine Health Policy, Management, and Evaluation, Toronto, Ontario
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario
- Mount Sinai Hospital, Toronto, Ontario
| | - Wayne L. Gold
- Departments of Medicine Health Policy, Toronto, Ontario
- University Health Network, Toronto, Ontario
| |
Collapse
|
123
|
Huang JL, Huang J, Duan ZH, Wei J, Min J, Luo XH, Li JG, Tan WP, Wu LZ, Liu RY, Li Y, Shao J, Huang BJ, Zeng YX, Huang W. Th2 predominance and CD8+ memory T cell depletion in patients with severe acute respiratory syndrome. Microbes Infect 2005; 7:427-436. [PMID: 15784184 PMCID: PMC7110803 DOI: 10.1016/j.micinf.2004.11.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2004] [Revised: 11/24/2004] [Accepted: 11/25/2004] [Indexed: 02/07/2023]
Abstract
UNLABELLED The immune spectrum of severe acute respiratory syndrome (SARS) is poorly understood. To define the dynamics of the immune spectrum in SARS, serum levels of cytokines, chemokines, immunoglobulins, complement and specific antibodies against SARS-associated coronavirus (SARS-CoV) were assayed by enzyme-linked immunosorbent assay (ELISA), and phenotypes of peripheral lymphocytes were analyzed by flow cytometry in 95 SARS-infected patients. Results showed that interleukin (IL)-10 and transforming growth factor beta (TGF-beta) were continuously up-regulated during the entirety of SARS. Regulated on activation normally T cell-expressed and secreted (RANTES) levels were decreased, while monocyte chemoattractant protein-1 (MCP-1) was elevated in acute patients. Immunoglobulins and complement were elevated during the first month of SARS. Both serum-positive rates and titers of specific IgM and IgG antibodies responding to SARS-CoV peaked at days 41-60 from the onset of SARS. CD4+ and CD8+ T lymphocytes decreased significantly in acute-phase. CD3+CD8+CD45RO+ T lymphocytes were decreased by 36.78% in the convalescent patients. CONCLUSION SARS-CoV seemed to elicit effective humoral immunity but inhibited cellular immunity, especially CD8+ memory T lymphocytes over time. Prolonged overproduction of IL-10 and TGF-beta may play an important role in the disease.
Collapse
Key Words
- severe acute respiratory syndrome
- immune monitoring
- immune system
- alt, aminotransferase
- anova, analysis of variance
- ast, aspartate aminotransferase
- c, complement
- cov, coronavirus
- ctr, control
- elisa, enzyme-linked immunosorbent assay
- fbs, fetal bovine serum
- ifn-γ, interferon γ
- ig, immunoglobulin
- il, interleukin
- mcp-1, monocyte chemoattractant protein-1
- nf-κb, nuclear factor κb
- pbmc, peripheral blood mononuclear cell
- rantes, regulated on activation normally t cell-expressed and secreted
- sars, severe acute respiratory syndrome
- sars-cov, sars-associated coronavirus
- s.d., standard deviation
- tgf, transforming growth factor
- tnf, tumor necrosis factor
Collapse
Affiliation(s)
- Jia-Ling Huang
- Cancer Institute, Cancer Center and World Health Organization (WHO) Cooperative Cancer Research Center, Cancer Center of Sun Yat-sen University, No. 651 Dongfeng Road East, Guangzhou 510060, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
124
|
Shi X, Gong E, Gao D, Zhang B, Zheng J, Gao Z, Zhong Y, Zou W, Wu B, Fang W, Liao S, Wang S, Xie Z, Lu M, Hou L, Zhong H, Shao H, Li N, Liu C, Pei F, Yang J, Wang Y, Han Z, Shi X, Zhang Q, You J, Zhu X, Gu J. Severe acute respiratory syndrome associated coronavirus is detected in intestinal tissues of fatal cases. Am J Gastroenterol 2005; 100:169-76. [PMID: 15654797 DOI: 10.1111/j.1572-0241.2005.40377.x] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES A significant percentage of confirmed severe acute respiratory syndrome (SARS) patients experienced gastrointestinal symptoms, and the viral sequence was detectable in the stool of most patients. At present, the knowledge of the pathology of the digestive system in SARS patients is limited. Because a resurgence of the SARS epidemic is constantly possible, there is an urgent need to understand the involvement of the digestive system in this new disease. METHODS We performed seven SARS autopsies in which samples of alimentary tract and digestive glands were examined with routine pathology, electron microscopy (EM), in situ hybridization (ISH), immunohistochemistry, and real-time polymerase chain reaction (PCR). RESULTS The main histopathological finding was atrophy of the mucosal lymphoid tissue. A few mucosal epithelial cells and lymphocytes in the intestine were positively stained for coronavirus with ISH. SARS-coronavirus (CoV)-like particles were found in the mucosal epithelial cells under EM and mild focal inflammation was detected in the alimentary tract. One patient who experienced severe diarrhea had pseudomembranous enteritis of the ileum. Fatty degeneration and central lobular necrosis were observed in the liver. No evidence of direct viral infection was found in the esophagus, the stomach, the salivary gland, the liver, or the pancreas. CONCLUSIONS In addition to the lungs, the gastrointestinal tract is another target of SARS-CoV infection, as the intestinal epithelial cells and mucosal lymphoid tissue are infected. The findings provide possible explanations for the gastrointestinal symptoms and the presence of virus in the stool of SARS patients.
Collapse
Affiliation(s)
- Xueying Shi
- Department of Pathology, Peking University Health Science Center, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
125
|
Yip TTC, Chan JWM, Cho WCS, Yip TT, Wang Z, Kwan TL, Law SCK, Tsang DNC, Chan JKC, Lee KC, Cheng WW, Ma VWS, Yip C, Lim CKP, Ngan RKC, Au JSK, Chan A, Lim WWL. Protein chip array profiling analysis in patients with severe acute respiratory syndrome identified serum amyloid a protein as a biomarker potentially useful in monitoring the extent of pneumonia. Clin Chem 2005; 51:47-55. [PMID: 15364884 PMCID: PMC7108187 DOI: 10.1373/clinchem.2004.031229] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND A new strain of coronavirus (CoV) has caused an outbreak of severe acute respiratory syndrome (SARS), with 8098 individuals being infected and 774 deaths worldwide. We carried out protein chip array profiling analysis in an attempt to identify biomarkers that might be useful in monitoring the clinical course of SARS patients. METHODS We performed surface-enhanced laser desorption ionization time-of-flight mass spectrometry on 89 sera collected from 28 SARS patients, 72 sera from 51 control patients with various viral or bacterial infections, and 10 sera from apparently healthy individuals. RESULTS Nine significantly increased and three significantly decreased serum biomarkers were discovered in the SARS patients compared with the controls. Among these biomarkers, one (11,695 Da) was identified to be serum amyloid A (SAA) protein by peptide mapping and tandem mass spectrometric analysis. When we monitored the SAA concentrations longitudinally in 45 sera from four SARS patients, we found a good correlation of SAA concentration with the extent of pneumonia as assessed by a serial chest x-ray opacity score. Increased SAA occurred in three of four patients at the time of extensive pneumonia as indicated by high x-ray scores. Over the course of gradual recovery in two patients, as assessed clinically and radiologically, SAA concentrations gradually decreased. In the third patient, the concentrations were initially increased, but were further increased with superimposed multiple bacterial infections. SAA was not markedly increased in the fourth patient, who had low x-ray scores and whose clinical course was relatively mild. CONCLUSIONS Protein chip array profiling analysis could be potentially useful in monitoring the severity of disease in SARS patients.
Collapse
Affiliation(s)
| | | | | | | | - Zheng Wang
- Ciphergen Biosystems Incorporation, Fremont, CA
| | | | | | - Dominic NC Tsang
- Pathology, Queen Elizabeth Hospital, Hong Kong Special Administrative Region, The People’s Republic of China
| | - John KC Chan
- Pathology, Queen Elizabeth Hospital, Hong Kong Special Administrative Region, The People’s Republic of China
| | - King-Chung Lee
- Pathology, Queen Elizabeth Hospital, Hong Kong Special Administrative Region, The People’s Republic of China
| | | | | | | | | | | | | | - Angel Chan
- Pathology, Queen Elizabeth Hospital, Hong Kong Special Administrative Region, The People’s Republic of China
| | - Wilina WL Lim
- Hong Kong Government Virus Unit, Department of Health, Hong Kong Special Administrative Region, The People’s Republic of China
| |
Collapse
|
126
|
de Groot-Mijnes JDF, van Dun JM, van der Most RG, de Groot RJ. Natural history of a recurrent feline coronavirus infection and the role of cellular immunity in survival and disease. J Virol 2005; 79:1036-44. [PMID: 15613332 PMCID: PMC538555 DOI: 10.1128/jvi.79.2.1036-1044.2005] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2004] [Accepted: 08/16/2004] [Indexed: 12/16/2022] Open
Abstract
We describe the natural history, viral dynamics, and immunobiology of feline infectious peritonitis (FIP), a highly lethal coronavirus infection. A severe recurrent infection developed, typified by viral persistence and acute lymphopenia, with waves of enhanced viral replication coinciding with fever, weight loss, and depletion of CD4+ and CD8+ T cells. Our combined observations suggest a model for FIP pathogenesis in which virus-induced T-cell depletion and the antiviral T-cell response are opposing forces and in which the efficacy of early T-cell responses critically determines the outcome of the infection. Rising amounts of viral RNA in the blood, consistently seen in animals with end-stage FIP, indicate that progression to fatal disease is the direct consequence of a loss of immune control, resulting in unchecked viral replication. The pathogenic phenomena described here likely bear relevance to other severe coronavirus infections, in particular severe acute respiratory syndrome, for which multiphasic disease progression and acute T-cell lymphopenia have also been reported. Experimental FIP presents a relevant, safe, and well-defined model to study coronavirus-mediated immunosuppression and should provide an attractive and convenient system for in vivo testing of anticoronaviral drugs.
Collapse
Affiliation(s)
- Jolanda D F de Groot-Mijnes
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| | | | | | | |
Collapse
|
127
|
Hung I, Cheng V, Wu A, Tang B, Chan K, Chu C, Wong M, Hui W, Poon L, Tse D, Chan K, Woo P, Lau S, Peiris J, Yuen K. Viral loads in clinical specimens and SARS manifestations. Emerg Infect Dis 2004; 10:1550-7. [PMID: 15498155 PMCID: PMC3320271 DOI: 10.3201/eid1009.040058] [Citation(s) in RCA: 202] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
A retrospective viral load study was performed on clinical specimens from 154 patients with laboratory-confirmed severe acute respiratory syndrome (SARS); the specimens were prospectively collected during patients' illness. Viral load in nasopharyngeal aspirates (n = 142) from day 10 to day 15 after onset of symptoms was associated with oxygen desaturation, mechanical ventilation, diarrhea, hepatic dysfunction, and death. Serum viral load (n = 53) was associated with oxygen desaturation, mechanical ventilation, and death. Stool viral load (n = 94) was associated with diarrhea, and urine viral load (n = 111) was associated with abnormal urinalysis results. Viral replications at different sites are important in the pathogenesis of clinical and laboratory abnormalities of SARS.
Collapse
Affiliation(s)
- I.F.N. Hung
- Queen Mary Hospital at the University of Hong Kong, Hong Kong Special Administrative Region (SAR), People's Republic of China
| | - V.C.C. Cheng
- Queen Mary Hospital at the University of Hong Kong, Hong Kong Special Administrative Region (SAR), People's Republic of China
| | - A.K.L. Wu
- Prince of Wales Hospital, Hong Kong SAR, People's Republic of China
| | - B.S.F. Tang
- Queen Mary Hospital at the University of Hong Kong, Hong Kong Special Administrative Region (SAR), People's Republic of China
| | - K.H. Chan
- Queen Mary Hospital at the University of Hong Kong, Hong Kong Special Administrative Region (SAR), People's Republic of China
| | - C.M. Chu
- United Christian Hospital, Hong Kong SAR, People's Republic of China
| | - M.M.L. Wong
- Caritas Medical Centre, Hong Kong SAR, People's Republic of China
| | - W.T. Hui
- Queen Mary Hospital at the University of Hong Kong, Hong Kong Special Administrative Region (SAR), People's Republic of China
| | - L.L.M. Poon
- Queen Mary Hospital at the University of Hong Kong, Hong Kong Special Administrative Region (SAR), People's Republic of China
| | - D.M.W. Tse
- Caritas Medical Centre, Hong Kong SAR, People's Republic of China
| | - K.S. Chan
- United Christian Hospital, Hong Kong SAR, People's Republic of China
| | - P.C.Y. Woo
- Queen Mary Hospital at the University of Hong Kong, Hong Kong Special Administrative Region (SAR), People's Republic of China
| | - S.K.P. Lau
- Queen Mary Hospital at the University of Hong Kong, Hong Kong Special Administrative Region (SAR), People's Republic of China
| | - J.S.M. Peiris
- Queen Mary Hospital at the University of Hong Kong, Hong Kong Special Administrative Region (SAR), People's Republic of China
| | - K.Y. Yuen
- Queen Mary Hospital at the University of Hong Kong, Hong Kong Special Administrative Region (SAR), People's Republic of China
| |
Collapse
|
128
|
Parashar UD, Anderson LJ. Severe acute respiratory syndrome: review and lessons of the 2003 outbreak. Int J Epidemiol 2004; 33:628-34. [PMID: 15155694 PMCID: PMC7108628 DOI: 10.1093/ije/dyh198] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Umesh D Parashar
- Respiratory and Enteric Viruses Branch, Division of Viral and Rickettsial Diseases, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta GA 30333
| | - Larry J Anderson
- Respiratory and Enteric Viruses Branch, Division of Viral and Rickettsial Diseases, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta GA 30333
| |
Collapse
|
129
|
Finlay BB, See RH, Brunham RC. Rapid response research to emerging infectious diseases: lessons from SARS. Nat Rev Microbiol 2004; 2:602-7. [PMID: 15197395 PMCID: PMC7097457 DOI: 10.1038/nrmicro930] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
New and emerging infectious diseases continue to plague the world, and there is significant concern that recombinant infectious agents can be used as bioterrorism threats. Microbiologists are increasingly being asked to apply their scientific knowledge to respond to these threats. The recent pandemic caused by the severe acute respiratory syndrome (SARS) coronavirus illustrated not only how a newly evolved pathogen can rapidly spread throughout the world but also how the global community can unite to identify the causative agent and control its spread. Rapid response research mechanisms, such as those used by the SARS Accelerated Vaccine Initiative (SAVI), have shown that the application of emergency management techniques, together with rapid response research, can be highly effective when applied appropriately to new infectious diseases.
Collapse
Affiliation(s)
- B Brett Finlay
- Biotechnology Laboratory and Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada.
| | | | | |
Collapse
|
130
|
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
|
131
|
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: 165] [Impact Index Per Article: 7.9] [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
|
132
|
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: 1.0] [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
|
133
|
Low JGH, Lee CC, Leo YS, Low JGH, Lee CC, Leo YS. Severe acute respiratory syndrome and pulmonary tuberculosis. Clin Infect Dis 2004; 38:e123-5. [PMID: 15227635 PMCID: PMC7107818 DOI: 10.1086/421396] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2004] [Accepted: 02/19/2004] [Indexed: 11/05/2022] Open
Abstract
In Singapore, of 236 patients with probable severe acute respiratory syndrome (SARS), 2 were coinfected with tuberculosis, a phenomenon not previously reported. The patients' tuberculosis episodes only came to light after full recovery from SARS, when they presented with persistent respiratory symptoms and/or worsening chest radiography findings.
Collapse
Affiliation(s)
- J G H Low
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore.
| | | | | | | | | | | |
Collapse
|
134
|
Lam CWK, Chan MHM, Wong CK. Severe acute respiratory syndrome: clinical and laboratory manifestations. Clin Biochem Rev 2004; 25:121-132. [PMID: 18458712 PMCID: PMC1904416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Severe acute respiratory syndrome (SARS) is a recently emerged infectious disease with significant morbidity and mortality. An epidemic in 2003 affected 8,098 patients in 29 countries with 774 deaths. The aetiological agent is a new coronavirus spread by droplet transmission. Clinical and general laboratory manifestations included fever, chills, rigor, myalgia, malaise, diarrhoea, cough, dyspnoea, pneumonia, lymphopenia, neutrophilia, thrombocytopenia, and elevated serum lactate dehydrogenase (LD), alanine aminotransferase (ALT) and creatine kinase (CK) activities. Treatment has been empirical; initial potent antibiotic cover, followed by simultaneous ribavirin and corticosteroids, with or without pulse high-dose methylprednisolone, have been used. The postulated disease progression comprises (1) active viral infection, (2) hyperactive immune response, and (3) recovery or pulmonary destruction and death. We investigated serum LD isoenzymes and blood lymphocyte subsets of SARS patients, and found LD1 activity as the best biochemical prognostic indicator for death, while CD3+, CD4+, CD8+ and natural killer cell counts were promising predictors for intensive care unit (ICU) admission. Plasma cytokine and chemokine profiles showed markedly elevated Th1 cytokine interferon (IFN)-gamma, inflammatory cytokines interleukin (IL)-1beta, IL-6 and IL-12, neutrophil chemokine IL-8, monocyte chemoattractant protein-1 (MCP-1), and Th1 chemokine IFN-gamma-inducible protein-10 (IP-10) for at least two weeks after disease onset, but there was no significant elevation of inflammatory cytokine tumor necrosis factor (TNF)-alpha and anti-inflammatory cytokine IL-10. Corticosteroid reduced IL-8, MCP-1 and IP-10 concentrations from 5-8 days after treatment. Measurement of biochemical markers of bone metabolism demonstrated significant but transient increase in bone resorption from Day 28-44 after onset of fever, when pulse steroid was most frequently given. With tapering down of steroid therapy, there was a decrease in bone resorption marker together with an increase in bone formation markers round Day 50, suggesting that some of the bone loss might be reversed. Our research studies on the chemical pathology and clinical immunology of SARS should have implications for the pathophysiology and therapy of this potentially lethal infection.
Collapse
Affiliation(s)
- Christopher W K Lam
- Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong.
| | | | | |
Collapse
|
135
|
Chan MHM, Wong VWS, Wong CK, Chan PKS, Chu CM, Hui DSC, Suen MWM, Sung JJY, Chung SSC, Lam CWK. Serum LD1 isoenzyme and blood lymphocyte subsets as prognostic indicators for severe acute respiratory syndrome. J Intern Med 2004; 255:512-8. [PMID: 15049886 PMCID: PMC7167120 DOI: 10.1111/j.1365-2796.2004.01323.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND The pathophysiology of severe acute respiratory syndrome (SARS) is at present poorly understood, but advanced age and serum total lactate dehydrogenase (LD) activity >300 U L(-1) have been associated with adverse clinical outcomes. Blood leucocytes and lymphocyte subsets were reported to decrease, respectively, in 47% and up to 100% of 38 patients in Beijing. However, their prognostic implications have not been thoroughly investigated. OBJECTIVE To investigate serum total LD, LD isoenzymes, and other parameters including blood lymphocyte subsets as prognostic indicators in SARS patients for adverse clinical outcomes in terms of admission to intensive care unit (ICU) and death. DESIGN Retrospective analysis. SUBJECTS AND METHODS A total of 109 patients with a clinical diagnosis of SARS according to the modified World Health Organization case definition of SARS were recruited from two major acute hospitals in Hong Kong. They were either involved in the initial outbreak of SARS, or cases from the community outbreak of Amoy Gardens between 10 March and 5 May 2003. The clinical diagnosis was subsequently confirmed by serological test and/or molecular analysis. Serum total LD and LD isoenzyme activities, complete blood picture with total leucocyte count and differential counts, absolute counts of CD3+, CD4+, CD8+, natural killer cells and B lymphocytes were measured daily upon admission. Receiver operating characteristic curve analysis was used to determine and compare different cut-offs for various biochemical and immunological parameters at peak serum total LD concentration in predicting adverse clinical outcomes. RESULTS Of a total of 109 patients, 41 were admitted to ICU and 42 died. Of 42 fatal patients, 24 died in ICU and 18 died in general medical wards. Age was found to be an independent prognostic indicator for death with an area under curve (AUC) of 0.96 [95% confidence interval (CI) = 0.90-0.99] but not for admission to ICU [AUC = 0.61 (CI = 0.51-0.70)]. Whilst serum total LD could only achieve AUC of 0.68 (CI = 0.59-0.77) for predicting death, LD1 isoenzyme was found to be the best biochemical prognostic indicator with AUC of 0.84 (CI = 0.75-0.90), sensitivity of 62% (CI = 46-76%), specificity of 93% (CI = 83-98%) at cut-off activity of > or =80 U L(-1). CD3+, CD4+, CD8+ and natural killer cell counts were promising immunological prognostic indicators for predicting admission to ICU with AUC of 0.94 (CI = 0.86-0.98), 0.91 (CI = 0.81-0.96), 0.93 (CI = 0.85-0.98), and 0.87 (CI = 0.76-0.94), respectively. CONCLUSIONS Apart from age, serum LD1 activity was the best prognostic indicator for predicting death in patients with SARS compared with serum total LD activity, haemoglobin concentration, leucocyte and lymphocyte counts. Its release could possibly be from blood erythrocytes and body tissues other than the myocardium. Blood CD3+, CD4+, CD8+ and natural killer cell counts were found to be good prognostic indicators for predicting admission to ICU in patients with SARS compared with age, leucocyte count and LD isoenzymes. The suppressed CD3+, CD4+, CD8+, and natural killer cell counts were also implicated in the pathophysiology of SARS. Patients with increased serum LD1 should be closely monitored to ensure prompt management, and preparation for admission to ICU could be planned ahead for patients with suppressed lymphocyte subsets.
Collapse
Affiliation(s)
- M H M Chan
- Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong
| | | | | | | | | | | | | | | | | | | |
Collapse
|
136
|
Cui W. Reply. Clin Infect Dis 2004. [PMCID: PMC7107917 DOI: 10.1086/382080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Wei Cui
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Beijing, People's Republic of China
- Reprints or correspondence: Dr. Wei Cui, Dept. of Clinical Laboratory, Peking Union Medical College Hospital, 1 Shuaifuyan Wangfujing, Beijing 100730, China ()
| |
Collapse
|
137
|
Zhou YH, Chen Z. Is the immune system impaired in patients with severe acute respiratory syndrome? Clin Infect Dis 2004; 38:921-2; author reply 922. [PMID: 14999642 PMCID: PMC7107854 DOI: 10.1086/382081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Affiliation(s)
- Yi-Hua Zhou
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
- Reprints or correspondence: Dr. Yi-Hua Zhou, Bldg. 50, Rm. 6535, 50 S. Dr. MSC-8009, LID/NIAID/NIH, Bethesda, MD 20892 ()
| | - Zhaochun Chen
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
138
|
Abstract
UNLABELLED INFECTIOUS AGENT: The severe acute respiratory syndrome (SARS) is a febrile pneumonia initially observed in China at the end of 2002. The infectious agent has rapidly been identified as a new coronavirus, baptised SARS-associated coronavirus (CoV-SARS). Transmission is inter-human, via respiratory particles mainly. CLINICAL PRESENTATION AND TREATMENT The clinical presentation is highly variable, from a mild fever to an acute respiratory distress syndrome. There is no specific treatment. Ribavirin associated with steroids have been used with success in numerous cases. EPIDEMIOLOGY During the first half of 2003, the spreading of the virus has been very fast, with a pandemic mode of evolution. More than 8,000 people were infected and 774 died. The reservoir of the virus, which may be animal, is still unknown. The epidemic seems to be controlled, but sporadic or epidemic re-emergences may occur and have been observed in China during January 2004.
Collapse
Affiliation(s)
- Amélie Guihot
- Service de maladies infectieuses et tropicales, hôpital Pitié-Salpêtrière, Paris (75)
| | - François Bricaire
- Service de maladies infectieuses et tropicales, hôpital Pitié-Salpêtrière, Paris (75)
| | - Taisheng Li
- Department of Infectious Diseases, Peking union medical college hospital, Chinese academy of medical sciences Pékin, Chine
| | - Philippe Bossi
- Service de maladies infectieuses et tropicales, hôpital Pitié-Salpêtrière, Paris (75)
- Correspondance : Philippe Bossi, Service de maladies infectieuses et tropicales hôpital Pitié-Salpêtrière 47-83, bd de l’Hôpital, 75013 Paris. Tél. : 01 42 16 01 01, Fax : 01 42 16 01 65.
| |
Collapse
|