46 results on '"Fielding BC"'
Search Results
2. SARS-CoV-2 spike protein diversity at an intra-host level, among SARS-CoV-2 infected individuals in South Africa, 2020 to 2022.
- Author
-
Subramoney K, Mtileni N, Davis A, Giandhari J, Tegally H, Wilkinson E, Naidoo Y, Ramphal Y, Pillay S, Ramphal U, Simane A, Reddy B, Mashishi B, Mbenenge N, de Oliveira T, Fielding BC, and Treurnicht FK
- Subjects
- Humans, South Africa epidemiology, Spike Glycoprotein, Coronavirus genetics, SARS-CoV-2 genetics, COVID-19 epidemiology
- Abstract
Intra-host diversity studies are used to characterise the mutational heterogeneity of SARS-CoV-2 infections in order to understand the impact of virus-host adaptations. This study investigated the frequency and diversity of the spike (S) protein mutations within SARS-CoV-2 infected South African individuals. The study included SARS-CoV-2 respiratory samples, from individuals of all ages, received at the National Health Laboratory Service at Charlotte Maxeke Johannesburg Academic hospital, Gauteng, South Africa, from June 2020 to May 2022. Single nucleotide polymorphism (SNP) assays and whole genome sequencing were performed on a random selection of SARS-CoV-2 positive samples. The allele frequency (AF) was determined using TaqMan Genotyper software for SNP PCR analysis and galaxy.eu for analysis of FASTQ reads from sequencing. The SNP assays identified 5.3% (50/948) of Delta cases with heterogeneity at delY144 (4%; 2/50), E484Q (6%; 3/50), N501Y (2%; 1/50) and P681H (88%; 44/50), however only heterogeneity for E484Q and delY144 were confirmed by sequencing. From sequencing we identified 9% (210/2381) of cases with Beta, Delta, Omicron BA.1, BA.2.15, and BA.4 lineages that had heterogeneity in the S protein. Heterogeneity was primarily identified at positions 19 (1.4%) with T19IR (AF 0.2-0.7), 371 (92.3%) with S371FP (AF 0.1-1.0), and 484 (1.9%) with E484AK (0.2-0.7), E484AQ (AF 0.4-0.5) and E484KQ (AF 0.1-0.4). Mutations at heterozygous amino acid positions 19, 371 and 484 are known antibody escape mutations, however the impact of the combination of multiple substitutions identified at the same position is unknown. Therefore, we hypothesise that intra-host SARS-CoV-2 quasispecies with heterogeneity in the S protein facilitate competitive advantage of variants that can completely/partially evade host's natural and vaccine-induced immune responses., Competing Interests: The authors declare no conflicts of interest., (Copyright: © 2023 Subramoney et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2023
- Full Text
- View/download PDF
3. Molecular Epidemiology of SARS-CoV-2 during Five COVID-19 Waves and the Significance of Low-Frequency Lineages.
- Author
-
Subramoney K, Mtileni N, Giandhari J, Naidoo Y, Ramphal Y, Pillay S, Ramphal U, Maharaj A, Tshiabuila D, Tegally H, Wilkinson E, de Oliveira T, Fielding BC, and Treurnicht FK
- Subjects
- Humans, Animals, Molecular Epidemiology, Databases, Factual, Drug Resistance, Viral, Mutation, Pangolins, Spike Glycoprotein, Coronavirus, SARS-CoV-2 genetics, COVID-19 epidemiology
- Abstract
SARS-CoV-2 lineages and variants of concern (VOC) have gained more efficient transmission and immune evasion properties with time. We describe the circulation of VOCs in South Africa and the potential role of low-frequency lineages on the emergence of future lineages. Whole genome sequencing was performed on SARS-CoV-2 samples from South Africa. Sequences were analysed with Nextstrain pangolin tools and Stanford University Coronavirus Antiviral & Resistance Database. In 2020, 24 lineages were detected, with B.1 (3%; 8/278), B.1.1 (16%; 45/278), B.1.1.348 (3%; 8/278), B.1.1.52 (5%; 13/278), C.1 (13%; 37/278) and C.2 (2%; 6/278) circulating during the first wave. Beta emerged late in 2020, dominating the second wave of infection. B.1 and B.1.1 continued to circulate at low frequencies in 2021 and B.1.1 re-emerged in 2022. Beta was outcompeted by Delta in 2021, which was thereafter outcompeted by Omicron sub-lineages during the 4th and 5th waves in 2022. Several significant mutations identified in VOCs were also detected in low-frequency lineages, including S68F (E protein); I82T (M protein); P13L, R203K and G204R/K (N protein); R126S (ORF3a); P323L (RdRp); and N501Y, E484K, D614G, H655Y and N679K (S protein). Low-frequency variants, together with VOCs circulating, may lead to convergence and the emergence of future lineages that may increase transmissibility, infectivity and escape vaccine-induced or natural host immunity.
- Published
- 2023
- Full Text
- View/download PDF
4. Editorial: Human coronavirus research: 20 years since the SARS-CoV outbreak.
- Author
-
Fielding BC
- Abstract
Competing Interests: The author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2022
- Full Text
- View/download PDF
5. The Flexible, Extended Coil of the PDZ-Binding Motif of the Three Deadly Human Coronavirus E Proteins Plays a Role in Pathogenicity.
- Author
-
Schoeman D, Cloete R, and Fielding BC
- Subjects
- Humans, SARS-CoV-2, Virulence, COVID-19, Coronavirus 229E, Human, Coronavirus NL63, Human, Coronavirus OC43, Human, Middle East Respiratory Syndrome Coronavirus
- Abstract
The less virulent human (h) coronaviruses (CoVs) 229E, NL63, OC43, and HKU1 cause mild, self-limiting respiratory tract infections, while the more virulent SARS-CoV-1, MERS-CoV, and SARS-CoV-2 have caused severe outbreaks. The CoV envelope (E) protein, an important contributor to the pathogenesis of severe hCoV infections, may provide insight into this disparate severity of the disease. We, therefore, generated full-length E protein models for SARS-CoV-1 and -2, MERS-CoV, HCoV-229E, and HCoV-NL63 and docked C-terminal peptides of each model to the PDZ domain of the human PALS1 protein. The PDZ-binding motif (PBM) of the SARS-CoV-1 and -2 and MERS-CoV models adopted a more flexible, extended coil, while the HCoV-229E and HCoV-NL63 models adopted a less flexible alpha helix. All the E peptides docked to PALS1 occupied the same binding site and the more virulent hCoV E peptides generally interacted more stably with PALS1 than the less virulent ones. We hypothesize that the increased flexibility of the PBM in the more virulent hCoVs facilitates more stable binding to various host proteins, thereby contributing to more severe disease. This is the first paper to model full-length 3D structures for both the more virulent and less virulent hCoV E proteins, providing novel insights for possible drug and/or vaccine development.
- Published
- 2022
- Full Text
- View/download PDF
6. Identification of SARS-CoV-2 Omicron variant using spike gene target failure and genotyping assays, Gauteng, South Africa, 2021.
- Author
-
Subramoney K, Mtileni N, Bharuthram A, Davis A, Kalenga B, Rikhotso M, Maphahlele M, Giandhari J, Naidoo Y, Pillay S, Ramphal U, Ramphal Y, Tegally H, Wilkinson E, Mohale T, Ismail A, Mashishi B, Mbenenge N, de Oliveira T, Makatini Z, Fielding BC, and Treurnicht FK
- Subjects
- Genotype, Humans, South Africa, Spike Glycoprotein, Coronavirus genetics, COVID-19 diagnosis, SARS-CoV-2 genetics
- Abstract
The circulation of Omicron BA.1 led to the rapid increase in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) cases in South Africa in November 2021, which warranted the use of more rapid detection methods. We, therefore, assessed the ability to detect Omicron BA.1 using genotyping assays to identify specific mutations in SARS-CoV-2 positive samples, Gauteng province, South Africa. The TaqPath™ COVID-19 real-time polymerase chain reaction assay was performed on all samples selected to identify spike gene target failure (SGTF). SARS-CoV-2 genotyping assays were used for the detection of del69/70 and K417N mutation. Whole-genome sequencing was performed on a subset of genotyped samples to confirm these findings. Of the positive samples received, 11.0% (175/1589) were randomly selected to assess if SGTF and genotyping assays, that detect del69/70 and K417N mutations, could identify Omicron BA.1. We identified SGTF in 98.9% (173/175) of samples, of which 88.0% (154/175) had both the del69/70 and K417N mutation. The genotyped samples (45.7%; 80/175) that were sequenced confirmed Omicron BA.1 (97.5%; 78/80). Our data show that genotyping for the detection of the del69/70 and K417N coupled with SGTF is efficient to exclude Alpha and Beta variants and rapidly detect Omicron BA.1. However, we still require assays for the detection of unique mutations that will allow for the differentiation between other Omicron sublineages. Therefore, the use of genotyping assays to detect new dominant or emerging lineages of SARS-CoV-2 will be beneficial in limited-resource settings., (© 2022 The Authors. Journal of Medical Virology published by Wiley Periodicals LLC.)
- Published
- 2022
- Full Text
- View/download PDF
7. HPLC-MS identification and expression of Candida drug-resistance proteins from African HIV-infected patients.
- Author
-
Abrantes PMDS, Fisher R, Bouic PJD, McArthur CP, Fielding BC, and Africa CWJ
- Abstract
The objective of this study was to elucidate the proteomic mechanisms of drug resistance in HIV-infected African patients. Cell membrane fractions from forty oral Candida isolates isolated from African HIV-positive patients were analysed using HPLC-MS with the aim of identifying proteins associated with their pathogenicity and drug resistance. Heat shock proteins that mediate the fungicidal activity of salivary peptides were found in all tested Candida fractions, with pH-responsive proteins associated with increased pathogenicity only being present in the three most commonly isolated species. ABC multidrug transporter efflux pumps and estrogen binding proteins were only found in C. albicans fractions, while ergosterol biosynthesis proteins were identified in four species. The combination of various adherence, invasion, upregulation and efflux pump mechanisms appear to be instrumental for the Candida host colonization and drug resistance emergence in HIV-infected individuals., Competing Interests: Conflict of interest: All authors declare no conflicts of interest in this paper., (© 2021 the Author(s), licensee AIMS Press.)
- Published
- 2021
- Full Text
- View/download PDF
8. Insult to Injury-Potential Contribution of Coronavirus Disease-19 to Neuroinflammation and the Development of HIV-Associated Neurocognitive Disorders.
- Author
-
Williams ME and Fielding BC
- Subjects
- AIDS-Associated Nephropathy virology, COVID-19 virology, Humans, Inflammation virology, Neurocognitive Disorders virology, SARS-CoV-2 pathogenicity, AIDS-Associated Nephropathy complications, COVID-19 complications, Inflammation complications, Neurocognitive Disorders complications
- Abstract
Severe acute respiratory syndrome (SARS)-coronavirus (CoV)-2 is responsible for a new coronavirus disease known as coronavirus disease-19 (COVID-19). SARS-CoV-2 reports neurotropic properties and may have neurological implications, and this creates another health burden for people living with HIV. As yet, the impact of COVID-19 on (neuro)inflammation and the development of HIV-associated neurocognitive disorders (HAND) is not fully known. Here, we reviewed preliminary evidence that provides clues that COVID-19 may exacerbate inflammatory mechanisms related to the development of HAND.
- Published
- 2021
- Full Text
- View/download PDF
9. Human Coronaviruses: Counteracting the Damage by Storm.
- Author
-
Schoeman D and Fielding BC
- Subjects
- Animals, COVID-19 epidemiology, COVID-19 virology, Coronavirus Infections epidemiology, Coronavirus Infections virology, Cytokines immunology, Humans, Middle East Respiratory Syndrome Coronavirus genetics, Middle East Respiratory Syndrome Coronavirus physiology, Pandemics, Severe acute respiratory syndrome-related coronavirus genetics, Severe acute respiratory syndrome-related coronavirus physiology, SARS-CoV-2 genetics, SARS-CoV-2 physiology, Severe Acute Respiratory Syndrome epidemiology, Severe Acute Respiratory Syndrome virology, COVID-19 immunology, Coronavirus Infections immunology, Severe Acute Respiratory Syndrome immunology
- Abstract
Over the past 18 years, three highly pathogenic human (h) coronaviruses (CoVs) have caused severe outbreaks, the most recent causative agent, SARS-CoV-2, being the first to cause a pandemic. Although much progress has been made since the COVID-19 pandemic started, much about SARS-CoV-2 and its disease, COVID-19, is still poorly understood. The highly pathogenic hCoVs differ in some respects, but also share some similarities in clinical presentation, the risk factors associated with severe disease, and the characteristic immunopathology associated with the progression to severe disease. This review aims to highlight these overlapping aspects of the highly pathogenic hCoVs-SARS-CoV, MERS-CoV, and SARS-CoV-2-briefly discussing the importance of an appropriately regulated immune response; how the immune response to these highly pathogenic hCoVs might be dysregulated through interferon (IFN) inhibition, antibody-dependent enhancement (ADE), and long non-coding RNA (lncRNA); and how these could link to the ensuing cytokine storm. The treatment approaches to highly pathogenic hCoV infections are discussed and it is suggested that a greater focus be placed on T-cell vaccines that elicit a cell-mediated immune response, using rapamycin as a potential agent to improve vaccine responses in the elderly and obese, and the potential of stapled peptides as antiviral agents.
- Published
- 2021
- Full Text
- View/download PDF
10. Leptin Deficiency, Caused by Malnutrition, Makes You Susceptible to SARS-CoV-2 Infection but Could Offer Protection from Severe COVID-19.
- Author
-
Schoeman D and Fielding BC
- Subjects
- Antibody Formation, Body Mass Index, COVID-19 Vaccines immunology, Cytokine Release Syndrome etiology, Cytokine Release Syndrome prevention & control, Developing Countries, Disease Susceptibility, Humans, Immunity, Cellular, Immunogenicity, Vaccine, Immunologic Deficiency Syndromes etiology, Leptin physiology, Lymphocyte Activation, Malnutrition immunology, Models, Biological, Obesity complications, Protein-Energy Malnutrition complications, Protein-Energy Malnutrition immunology, Risk, Severity of Illness Index, T-Lymphocytes immunology, COVID-19 complications, Leptin deficiency, Malnutrition complications, SARS-CoV-2
- Abstract
In much of the developing world, severe malnutrition is the most prevalent cause of immunodeficiency and affects up to 50% of the population in some impoverished communities. As yet, we do not know how severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) will behave in populations with immunodeficiency caused by malnourishment. Interestingly, researchers are now speculating that, in some instances, a defective cellular immune system could paradoxically be a protective factor against severe disease in certain patients contracting SARS-CoV and SARS-CoV-2. This could be linked to the absence of T-cell activation. Based on available information presented here, it is plausible that the hyperimmune response, and subsequent cytokine storm often associated with severe coronavirus disease 2019 (COVID-19), could be "counteracted" by the defective immune response seen in individuals with malnutrition-induced leptin deficiency. In this paper, we proposed a theory that although those with malnutrition-linked leptin deficiency are at risk of SARS-CoV-2 infection, they are at lower risk of developing severe COVID-19., (Copyright © 2021 Schoeman and Fielding.)
- Published
- 2021
- Full Text
- View/download PDF
11. Alkaloids: Therapeutic Potential against Human Coronaviruses.
- Author
-
Fielding BC, da Silva Maia Bezerra Filho C, Ismail NSM, and Sousa DP
- Subjects
- Alkaloids therapeutic use, Antiviral Agents therapeutic use, COVID-19 virology, Humans, Pandemics, SARS-CoV-2 chemistry, SARS-CoV-2 drug effects, Spike Glycoprotein, Coronavirus chemistry, Alkaloids chemistry, Antiviral Agents chemistry, Spike Glycoprotein, Coronavirus antagonists & inhibitors, COVID-19 Drug Treatment
- Abstract
Alkaloids are a class of natural products known to have wide pharmacological activity and have great potential for the development of new drugs to treat a wide array of pathologies. Some alkaloids have antiviral activity and/or have been used as prototypes in the development of synthetic antiviral drugs. In this study, eleven anti-coronavirus alkaloids were identified from the scientific literature and their potential therapeutic value against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is discussed. In this study, in silico studies showed an affinity of the alkaloids for binding to the receptor-binding domain of the SARS-CoV-2 spike protein, putatively preventing it from binding to the host cell. Lastly, several mechanisms for the known anti-coronavirus activity of alkaloids were discussed, showing that the alkaloids are interesting compounds with potential use as bioactive agents against SARS-CoV-2.
- Published
- 2020
- Full Text
- View/download PDF
12. Is There a Link Between the Pathogenic Human Coronavirus Envelope Protein and Immunopathology? A Review of the Literature.
- Author
-
Schoeman D and Fielding BC
- Abstract
Since the severe acute respiratory syndrome (SARS) outbreak in 2003, human coronaviruses (hCoVs) have been identified as causative agents of severe acute respiratory tract infections. Two more hCoV outbreaks have since occurred, the most recent being SARS-CoV-2, the causative agent of coronavirus disease 2019 (COVID-19). The clinical presentation of SARS and MERS is remarkably similar to COVID-19, with hyperinflammation causing a severe form of the disease in some patients. Previous studies show that the expression of the SARS-CoV E protein is associated with the hyperinflammatory response that could culminate in acute respiratory distress syndrome (ARDS), a potentially fatal complication. This immune-mediated damage is largely caused by a cytokine storm, which is induced by significantly elevated levels of inflammatory cytokines interleukin (IL)-1β and IL-6, which are partly mediated by the expression of the SARS-CoV E protein. The interaction between the SARS-CoV E protein and the host protein, syntenin, as well as the viroporin function of SARS-CoV E, are linked to this cytokine dysregulation. This review aims to compare the clinical presentation of virulent hCoVs with a specific focus on the cause of the immunopathology. The review also proposes that inhibition of IL-1β and IL-6 in severe cases can improve patient outcome., (Copyright © 2020 Schoeman and Fielding.)
- Published
- 2020
- Full Text
- View/download PDF
13. HIV and Human Coronavirus Coinfections: A Historical Perspective.
- Author
-
Makoti P and Fielding BC
- Subjects
- Betacoronavirus isolation & purification, COVID-19, Coinfection epidemiology, Coinfection immunology, Coinfection therapy, Coronavirus Infections epidemiology, Coronavirus Infections immunology, Coronavirus Infections therapy, HIV Infections epidemiology, HIV Infections immunology, HIV Infections therapy, Humans, Pandemics, Pneumonia, Viral epidemiology, Pneumonia, Viral immunology, Pneumonia, Viral therapy, SARS-CoV-2, Treatment Outcome, Coinfection virology, Coronavirus Infections virology, HIV Infections virology, Pneumonia, Viral virology
- Abstract
Seven human coronaviruses (hCoVs) are known to infect humans. The most recent one, SARS-CoV-2, was isolated and identified in January 2020 from a patient presenting with severe respiratory illness in Wuhan, China. Even though viral coinfections have the potential to influence the resultant disease pattern in the host, very few studies have looked at the disease outcomes in patients infected with both HIV and hCoVs. Groups are now reporting that even though HIV-positive patients can be infected with hCoVs, the likelihood of developing severe CoV-related diseases in these patients is often similar to what is seen in the general population. This review aimed to summarize the current knowledge of coinfections reported for HIV and hCoVs. Moreover, based on the available data, this review aimed to theorize why HIV-positive patients do not frequently develop severe CoV-related diseases.
- Published
- 2020
- Full Text
- View/download PDF
14. Natural Antioxidants: A Review of Studies on Human and Animal Coronavirus.
- Author
-
Diniz LRL, Bezerra Filho CDSM, Fielding BC, and de Sousa DP
- Subjects
- Animals, Antioxidants therapeutic use, Coronavirus pathogenicity, Coronavirus Infections virology, Humans, Antioxidants pharmacology, Coronavirus drug effects, Coronavirus Infections drug therapy
- Abstract
The outbreaks of viruses with wide spread and mortality in the world population have motivated the research for new therapeutic approaches. There are several viruses that cause a biochemical imbalance in the infected cell resulting in oxidative stress. These effects may be associated with the development of pathologies and worsening of symptoms. Therefore, this review is aimed at discussing natural compounds with both antioxidant and antiviral activities, specifically against coronavirus infection, in an attempt to contribute to global researches for discovering effective therapeutic agents in the treatment of coronavirus infection and its severe clinical complications. The contribution of the possible action of these compounds on metabolic modulation associated with antiviral properties, in addition to other mechanisms of action, is presented., Competing Interests: No potential conflict of interest was reported by the authors., (Copyright © 2020 Lúcio Ricardo Leite Diniz et al.)
- Published
- 2020
- Full Text
- View/download PDF
15. Coronavirus envelope protein: current knowledge.
- Author
-
Schoeman D and Fielding BC
- Subjects
- Animals, Coronavirus genetics, Coronavirus pathogenicity, Coronavirus Infections virology, Humans, Severe acute respiratory syndrome-related coronavirus chemistry, Severe acute respiratory syndrome-related coronavirus genetics, Severe acute respiratory syndrome-related coronavirus pathogenicity, Severe Acute Respiratory Syndrome virology, Viral Envelope Proteins genetics, Zoonoses transmission, Zoonoses virology, Coronavirus chemistry, Viral Envelope Proteins chemistry
- Abstract
Background: Coronaviruses (CoVs) primarily cause enzootic infections in birds and mammals but, in the last few decades, have shown to be capable of infecting humans as well. The outbreak of severe acute respiratory syndrome (SARS) in 2003 and, more recently, Middle-East respiratory syndrome (MERS) has demonstrated the lethality of CoVs when they cross the species barrier and infect humans. A renewed interest in coronaviral research has led to the discovery of several novel human CoVs and since then much progress has been made in understanding the CoV life cycle. The CoV envelope (E) protein is a small, integral membrane protein involved in several aspects of the virus' life cycle, such as assembly, budding, envelope formation, and pathogenesis. Recent studies have expanded on its structural motifs and topology, its functions as an ion-channelling viroporin, and its interactions with both other CoV proteins and host cell proteins., Main Body: This review aims to establish the current knowledge on CoV E by highlighting the recent progress that has been made and comparing it to previous knowledge. It also compares E to other viral proteins of a similar nature to speculate the relevance of these new findings. Good progress has been made but much still remains unknown and this review has identified some gaps in the current knowledge and made suggestions for consideration in future research., Conclusions: The most progress has been made on SARS-CoV E, highlighting specific structural requirements for its functions in the CoV life cycle as well as mechanisms behind its pathogenesis. Data shows that E is involved in critical aspects of the viral life cycle and that CoVs lacking E make promising vaccine candidates. The high mortality rate of certain CoVs, along with their ease of transmission, underpins the need for more research into CoV molecular biology which can aid in the production of effective anti-coronaviral agents for both human CoVs and enzootic CoVs.
- Published
- 2019
- Full Text
- View/download PDF
16. Acute toxicity studies of the South African medicinal plant Galenia africana .
- Author
-
Ng'uni T, Klaasen JA, and Fielding BC
- Abstract
Background: Medicinal plants are used by a large proportion of the global population as complementary and alternative medicines. However, little is known about their toxicity. G. africana has been used to treat wounds, coughs and skin diseases and is used in cosmetic formulations such as lotions and shampoos., Methods: The acute oral and dermal toxicity potential of G. africana was analyzed after a single administration of 300 and 2000 mg/kgbw for acute oral toxicity and 2000 mg/kgbw for acute dermal toxicity. Female Sprague-Dawley rats were used for the acute oral toxicity study whereas both male and female Sprague-Dawley rats were used for the acute dermal toxicity study. In the Episkin skin irritation test, the irritation potential of G. africana (concentrate) and G. africana (in-use dilution) extracts were assessed using the Episkin reconstituted human epidermis. In the dermal sensitization study, female CBA/Ca mice were treated with G. africana concentrations of 50, 100 and 200 mg/ml respectively. The vehicle of choice was dimethylformamide which acted as a control., Results: The results of the acute oral and dermal toxicity studies revealed that the median lethal dosage (LD
50 ) for G. africana extract in Sprague-Dawley rats was considered to exceed 2000 mg/kgbw. In the irritation test, the G. africana (concentrate) and G. africana (in-use dilution) extracts were non-irritant on the Episkin reconstituted human epidermis. In the dermal sensitization study, the stimulation index (SI) values for the mice treated with the G. africana extract at concentrations of 50, 100 and 200 mg/ml/kgbw, when compared to the control group, were 1.3, 0.9 and 1.3 respectively. The open application of the extract at the various concentrations did not result in a SI of ≥ 3 in any group. Hence, it did not elicit a hypersensitivity response., Conclusion: These findings demonstrate that the acute toxicity profile for G. africana is acceptable and can subsequently be used for single use in the pharmaceutical and cosmetic industries.- Published
- 2018
- Full Text
- View/download PDF
17. MERS-CoV: Understanding the Latest Human Coronavirus Threat.
- Author
-
Chafekar A and Fielding BC
- Subjects
- Animals, Antiviral Agents therapeutic use, Coronavirus Infections prevention & control, Coronavirus Infections therapy, Coronavirus Infections transmission, Disease Models, Animal, Genome, Viral, Humans, Middle East Respiratory Syndrome Coronavirus genetics, Middle East Respiratory Syndrome Coronavirus immunology, Respiratory Tract Infections virology, Vaccines isolation & purification, Viral Proteins, Coronavirus Infections diagnosis, Middle East Respiratory Syndrome Coronavirus pathogenicity, Respiratory Tract Infections diagnosis
- Abstract
Human coronaviruses cause both upper and lower respiratory tract infections in humans. In 2012, a sixth human coronavirus (hCoV) was isolated from a patient presenting with severe respiratory illness. The 60-year-old man died as a result of renal and respiratory failure after admission to a hospital in Jeddah, Saudi Arabia. The aetiological agent was eventually identified as a coronavirus and designated Middle East respiratory syndrome coronavirus (MERS-CoV). MERS-CoV has now been reported in more than 27 countries across the Middle East, Europe, North Africa and Asia. As of July 2017, 2040 MERS-CoV laboratory confirmed cases, resulting in 712 deaths, were reported globally, with a majority of these cases from the Arabian Peninsula. This review summarises the current understanding of MERS-CoV, with special reference to the (i) genome structure; (ii) clinical features; (iii) diagnosis of infection; and (iv) treatment and vaccine development., Competing Interests: The authors declare no conflict of interest.
- Published
- 2018
- Full Text
- View/download PDF
18. Molecular Detection of Tick-Borne Pathogen Diversities in Ticks from Livestock and Reptiles along the Shores and Adjacent Islands of Lake Victoria and Lake Baringo, Kenya.
- Author
-
Omondi D, Masiga DK, Fielding BC, Kariuki E, Ajamma YU, Mwamuye MM, Ouso DO, and Villinger J
- Abstract
Although diverse tick-borne pathogens (TBPs) are endemic to East Africa, with recognized impact on human and livestock health, their diversity and specific interactions with tick and vertebrate host species remain poorly understood in the region. In particular, the role of reptiles in TBP epidemiology remains unknown, despite having been implicated with TBPs of livestock among exported tortoises and lizards. Understanding TBP ecologies, and the potential role of common reptiles, is critical for the development of targeted transmission control strategies for these neglected tropical disease agents. During the wet months (April-May; October-December) of 2012-2013, we surveyed TBP diversity among 4,126 ticks parasitizing livestock and reptiles at homesteads along the shores and islands of Lake Baringo and Lake Victoria in Kenya, regions endemic to diverse neglected tick-borne diseases. After morphological identification of 13 distinct Rhipicephalus, Amblyomma , and Hyalomma tick species, ticks were pooled (≤8 individuals) by species, host, sampling site, and collection date into 585 tick pools. By supplementing previously established molecular assays for TBP detection with high-resolution melting analysis of PCR products before sequencing, we identified high frequencies of potential disease agents of ehrlichiosis (12.48% Ehrlichia ruminantium , 9.06% Ehrlichia canis ), anaplasmosis (6.32% Anaplasma ovis , 14.36% Anaplasma platys , and 3.08% Anaplasma bovis ,), and rickettsiosis (6.15% Rickettsia africae , 2.22% Rickettsia aeschlimannii , 4.27% Rickettsia rhipicephali , and 4.95% Rickettsia spp.), as well as Paracoccus sp. and apicomplexan hemoparasites (0.51% Theileria sp., 2.56% Hepatozoon fitzsimonsi , and 1.37% Babesia caballi ) among tick pools. Notably, we identified E. ruminantium in both Amblyomma and Rhipicephalus pools of ticks sampled from livestock in both study areas as well as in Amblyomma falsomarmoreum (66.7%) and Amblyomma nuttalli (100%) sampled from tortoises and Amblyomma sparsum (63.6%) sampled in both cattle and tortoises at Lake Baringo. Similarly, we identified E. canis in rhipicephaline ticks sampled from livestock and dogs in both regions and Amblyomma latum (75%) sampled from monitor lizards at Lake Victoria. These novel tick-host-pathogen interactions have implications on the risk of disease transmission to humans and domestic animals and highlight the complexity of TBP ecologies, which may include reptiles as reservoir species, in sub-Saharan Africa.
- Published
- 2017
- Full Text
- View/download PDF
19. Potential Broad Spectrum Inhibitors of the Coronavirus 3CLpro: A Virtual Screening and Structure-Based Drug Design Study.
- Author
-
Berry M, Fielding BC, and Gamieldien J
- Subjects
- Antiviral Agents chemistry, Antiviral Agents pharmacology, Coronavirus 3C Proteases, Cysteine Endopeptidases chemistry, Cysteine Proteinase Inhibitors chemistry, Cysteine Proteinase Inhibitors pharmacology, Humans, Molecular Docking Simulation, Molecular Dynamics Simulation, Viral Proteins chemistry, Antiviral Agents isolation & purification, Coronavirus drug effects, Cysteine Proteinase Inhibitors isolation & purification, Drug Design, Drug Evaluation, Preclinical methods, Viral Proteins antagonists & inhibitors
- Abstract
Human coronaviruses represent a significant disease burden; however, there is currently no antiviral strategy to combat infection. The outbreak of severe acute respiratory syndrome (SARS) in 2003 and Middle East respiratory syndrome (MERS) less than 10 years later demonstrates the potential of coronaviruses to cross species boundaries and further highlights the importance of identifying novel lead compounds with broad spectrum activity. The coronavirus 3CL(pro) provides a highly validated drug target and as there is a high degree of sequence homology and conservation in main chain architecture the design of broad spectrum inhibitors is viable. The ZINC drugs-now library was screened in a consensus high-throughput pharmacophore modeling and molecular docking approach by Vina, Glide, GOLD and MM-GBSA. Molecular dynamics further confirmed results obtained from structure-based techniques. A highly defined hit-list of 19 compounds was identified by the structure-based drug design methodologies. As these compounds were extensively validated by a consensus approach and by molecular dynamics, the likelihood that at least one of these compounds is bioactive is excellent. Additionally, the compounds segregate into 15 significantly dissimilar (p < 0.05) clusters based on shape and features, which represent valuable scaffolds that can be used as a basis for future anti-coronaviral inhibitor discovery experiments. Importantly though, the enriched subset of 19 compounds identified from the larger library has to be validated experimentally.
- Published
- 2015
- Full Text
- View/download PDF
20. Unraveling Host-Vector-Arbovirus Interactions by Two-Gene High Resolution Melting Mosquito Bloodmeal Analysis in a Kenyan Wildlife-Livestock Interface.
- Author
-
Omondi D, Masiga DK, Ajamma YU, Fielding BC, Njoroge L, and Villinger J
- Subjects
- Animals, Arbovirus Infections veterinary, Blood, Humans, Kenya, Animals, Wild genetics, Arbovirus Infections transmission, Arboviruses pathogenicity, Culicidae virology, Host-Pathogen Interactions, Insect Vectors, Livestock genetics
- Abstract
The blood-feeding patterns of mosquitoes are directly linked to the spread of pathogens that they transmit. Efficient identification of arthropod vector bloodmeal hosts can identify the diversity of vertebrate species potentially involved in disease transmission cycles. While molecular bloodmeal analyses rely on sequencing of cytochrome b (cyt b) or cytochrome oxidase 1 gene PCR products, recently developed bloodmeal host identification based on high resolution melting (HRM) analyses of cyt b PCR products is more cost-effective. To resolve the diverse vertebrate hosts that mosquitoes may potentially feed on in sub-Saharan Africa, we utilized HRM profiles of both cyt b and 16S ribosomal RNA genes. Among 445 blood-fed Aedeomyia, Aedes, Anopheles, Culex, Mansonia, and Mimomyia mosquitoes from Kenya's Lake Victoria and Lake Baringo regions where many mosquito-transmitted pathogens are endemic, we identified 33 bloodmeal hosts including humans, eight domestic animal species, six peridomestic animal species and 18 wildlife species. This resolution of vertebrate host species was only possible by comparing profiles of both cyt b and 16S markers, as melting profiles of some pairs of species were similar for either marker but not both. We identified mixed bloodmeals in a Culex pipiens from Mbita that had fed on a goat and a human and in two Mansonia africana mosquitoes from Baringo that each had fed on a rodent (Arvicanthis niloticus) in addition to a human or baboon. We further detected Sindbis and Bunyamwera viruses in blood-fed mosquito homogenates by Vero cell culture and RT-PCR in Culex, Aedeomyia, Anopheles and Mansonia mosquitoes from Baringo that had fed on humans and livestock. The observed mosquito feeding on both arbovirus amplifying hosts (including sheep and goats) and possible arbovirus reservoirs (birds, porcupine, baboons, rodents) informs arbovirus disease epidemiology and vector control strategies.
- Published
- 2015
- Full Text
- View/download PDF
21. Identification of new respiratory viruses in the new millennium.
- Author
-
Berry M, Gamieldien J, and Fielding BC
- Subjects
- Global Health, Humans, Viruses classification, Communicable Diseases, Emerging epidemiology, Communicable Diseases, Emerging virology, Respiratory Tract Infections epidemiology, Respiratory Tract Infections virology, Virus Diseases epidemiology, Virus Diseases virology, Viruses isolation & purification
- Abstract
The rapid advancement of molecular tools in the past 15 years has allowed for the retrospective discovery of several new respiratory viruses as well as the characterization of novel emergent strains. The inability to characterize the etiological origins of respiratory conditions, particularly in children, led several researchers to pursue the discovery of the underlying etiology of disease. In 2001, this led to the discovery of human metapneumovirus (hMPV) and soon following that the outbreak of Severe Acute Respiratory Syndrome coronavirus (SARS-CoV) promoted an increased interest in coronavirology and the latter discovery of human coronavirus (HCoV) NL63 and HCoV-HKU1. Human bocavirus, with its four separate lineages, discovered in 2005, has been linked to acute respiratory tract infections and gastrointestinal complications. Middle East Respiratory Syndrome coronavirus (MERS-CoV) represents the most recent outbreak of a completely novel respiratory virus, which occurred in Saudi Arabia in 2012 and presents a significant threat to human health. This review will detail the most current clinical and epidemiological findings to all respiratory viruses discovered since 2001.
- Published
- 2015
- Full Text
- View/download PDF
22. The coronavirus nucleocapsid is a multifunctional protein.
- Author
-
McBride R, van Zyl M, and Fielding BC
- Subjects
- Coronavirus Nucleocapsid Proteins, Protein Transport, Coronavirus physiology, Nucleocapsid Proteins metabolism, Virus Replication
- Abstract
The coronavirus nucleocapsid (N) is a structural protein that forms complexes with genomic RNA, interacts with the viral membrane protein during virion assembly and plays a critical role in enhancing the efficiency of virus transcription and assembly. Recent studies have confirmed that N is a multifunctional protein. The aim of this review is to highlight the properties and functions of the N protein, with specific reference to (i) the topology; (ii) the intracellular localization and (iii) the functions of the protein.
- Published
- 2014
- Full Text
- View/download PDF
23. The role of severe acute respiratory syndrome (SARS)-coronavirus accessory proteins in virus pathogenesis.
- Author
-
McBride R and Fielding BC
- Subjects
- Animals, Coronavirus pathogenicity, Humans, Open Reading Frames, Severe Acute Respiratory Syndrome epidemiology, Severe Acute Respiratory Syndrome transmission, Coronavirus genetics, Coronavirus metabolism, Severe Acute Respiratory Syndrome virology, Viral Regulatory and Accessory Proteins genetics, Viral Regulatory and Accessory Proteins metabolism
- Abstract
A respiratory disease caused by a novel coronavirus, termed the severe acute respiratory syndrome coronavirus (SARS-CoV), was first reported in China in late 2002. The subsequent efficient human-to-human transmission of this virus eventually affected more than 30 countries worldwide, resulting in a mortality rate of ~10% of infected individuals. The spread of the virus was ultimately controlled by isolation of infected individuals and there has been no infections reported since April 2004. However, the natural reservoir of the virus was never identified and it is not known if this virus will re-emerge and, therefore, research on this virus continues. The SARS-CoV genome is about 30 kb in length and is predicted to contain 14 functional open reading frames (ORFs). The genome encodes for proteins that are homologous to known coronavirus proteins, such as the replicase proteins (ORFs 1a and 1b) and the four major structural proteins: nucleocapsid (N), spike (S), membrane (M) and envelope (E). SARS-CoV also encodes for eight unique proteins, called accessory proteins, with no known homologues. This review will summarize the current knowledge on SARS-CoV accessory proteins and will include: (i) expression and processing; (ii) the effects on cellular processes; and (iii) functional studies.
- Published
- 2012
- Full Text
- View/download PDF
24. Optimization and preclinical design of genetically engineered viruses for human oncolytic therapy.
- Author
-
Hiss DC and Fielding BC
- Subjects
- Animals, Genetic Engineering standards, Genetic Therapy methods, Genetic Therapy standards, Humans, Oncolytic Virotherapy standards, Viruses genetics, Genetic Engineering methods, Neoplasms genetics, Neoplasms therapy, Oncolytic Virotherapy methods, Oncolytic Viruses genetics
- Abstract
Introduction: Oncolytic viruses (OVs) occupy a strategic niche in the dynamic era of biological and gene therapy of human cancers. However, the use of OVs is the subject of close scrutiny due to impediments such as the insufficiency of patient generalizations posed by heterogeneous tumor responses to treatment, inherent or potentially lethal viral pathogenicities, unanticipated host- or immune-related adverse effects, and the emergence of virus-resistant cancer cells. These challenges can be overcome by the design and development of more definitive (optimized, targeted, and individualized) cancer virotherapeutics., Areas Covered: The translation of current knowledge and recent innovations into rational treatment prospects hinges on an iterative loop of variables pertaining to genetically engineered viral oncolytic efficacy and safety profiles, mechanism-of-action data, potencies of synergistic oncolytic viral combinations with conventional tumor, immuno-, chemo-, and radiation treatment modalities, optimization of the probabilities of treatment successes in heterogeneous (virus-sensitive and -resistant) tumor cell populations by mathematical modeling, and lessons learned from preclinical studies and human clinical trials., Expert Opinion: In recent years, it has become increasingly clear that proof-of-principle is critical for the preclinical optimization of oncolytic viruses to target heterogeneous forms of cancer and to prioritize current concerns related to the efficacy and safety of oncolytic virotherapy.
- Published
- 2012
- Full Text
- View/download PDF
25. The variable N-terminal region of DDX5 contains structural elements and auto-inhibits its interaction with NS5B of hepatitis C virus.
- Author
-
Dutta S, Gupta G, Choi YW, Kotaka M, Fielding BC, Song J, and Tan YJ
- Subjects
- Adenosine Triphosphate metabolism, Binding Sites, Cell Line, Crystallography, X-Ray, DEAD-box RNA Helicases genetics, Humans, Nuclear Magnetic Resonance, Biomolecular, Protein Conformation, Protein Structure, Tertiary, Structural Homology, Protein, DEAD-box RNA Helicases chemistry, DEAD-box RNA Helicases metabolism, Viral Nonstructural Proteins metabolism
- Abstract
RNA helicases of the DEAD (Asp-Glu-Ala-Asp)-box family of proteins are involved in many aspects of RNA metabolism from transcription to RNA decay, but most of them have also been shown to be multifunctional. The DEAD-box helicase DDX5 of host cells has been shown to interact with the RNA-dependent RNA polymerase (NS5B) of HCV (hepatitis C virus). In the present study, we report the presence of two independent NS5B-binding sites in DDX5, one located at the N-terminus and another at the C-terminus. The N-terminal fragment of DDX5, which consists of the first 305 amino acids and shall be referred as DDX5-N, was expressed and crystallized. The crystal structure shows that domain 1 (residues 79-303) of DDX5 contains the typical features found in the structures of other DEAD-box helicases. DDX5-N also contains the highly variable NTR (N-terminal region) of unknown function and the crystal structure reveals structural elements in part of the NTR, namely residues 52-78. This region forms an extensive loop and an α-helix. From co-immunoprecipitation experiments, the NTR of DDX5-N was observed to auto-inhibit its interaction with NS5B. Interestingly, the α-helix in NTR is essential for this auto-inhibition and seems to mediate the interaction between the highly flexible 1-51 residues in NTR and the NS5B-binding site in DDX5-N. Furthermore, NMR investigations reveal that there is a direct interaction between DDX5 and NS5B in vitro.
- Published
- 2012
- Full Text
- View/download PDF
26. Antimicrobial-resistant Klebsiella species isolated from free-range chicken samples in an informal settlement.
- Author
-
Fielding BC, Mnabisa A, Gouws PA, and Morris T
- Abstract
Introduction: Sub-therapeutic doses of antimicrobial agents are administered routinely to poultry to aid growth and to prevent disease, with prolonged exposure often resulting in bacterial resistance. Crossover of antibiotic resistant bacteria from poultry to humans poses a risk to human health., Material and Methods: In this study, 17 chicken samples collected from a vendor operating in an informal settlement in the Cape Town Metropolitan area, South Africa were screened for antimicrobial-resistant Gram-negative bacilli using the Kirby Bauer disk diffusion assay., Results: IN TOTAL, SIX ANTIBIOTICS WERE SCREENED: ampicillin, ciprofloxacin, gentamicin, nalidixic acid, tetracycline and trimethoprim. Surprisingly, Klebsiella ozaenae was identified in 96 and K. rhinoscleromatis in 6 (n=102) of the samples tested. Interestingly, ∼40% of the isolated Klebsiella spp. showed multiple resistance to at least three of the six antibiotics tested., Conclusions: Klebsiella ozaenae and K. rhinoscleromatis cause clinical chronic rhinitis and are almost exclusively associated with people living in areas of poor hygiene.
- Published
- 2012
- Full Text
- View/download PDF
27. Human coronavirus NL63: a clinically important virus?
- Author
-
Fielding BC
- Subjects
- Child, Preschool, Coronavirus Infections virology, Humans, Infant, Prevalence, Respiratory Tract Infections virology, Seasons, Coronavirus Infections epidemiology, Coronavirus Infections pathology, Coronavirus NL63, Human isolation & purification, Respiratory Tract Infections epidemiology, Respiratory Tract Infections pathology
- Abstract
Respiratory tract infection is a leading cause of morbidity and mortality worldwide, especially among young children. Human coronaviruses (HCoVs) have only recently been shown to cause both lower and upper respiratory tract infections. To date, five coronaviruses (HCoV-229E, HCoV-OC43, SARS-CoV, HCoV-NL63 and HCoV HKU-1) that infect humans have been identified, four of which (HCoV-229E, HCoV-OC43, HCoV-NL63 and HCoV-HKU-1) circulate continuously in the human population. Human coronavirus NL63 (HCoV-NL63) was first isolated from the aspirate from a 7-month-old baby in early 2004. Infection with HCoV-NL63 has since been shown to be a common worldwide occurrence and has been associated with many clinical symptoms and diagnoses, including severe lower respiratory tract infection, croup and bronchiolitis. HCoV-NL63 causes disease in children, the elderly and the immunocompromised, and has been detected in 1.0-9.3% of respiratory tract infections in children. In this article, the current knowledge of human coronavirus HCoV-NL63, with special reference to the clinical features, prevalence and seasonal incidence, and coinfection with other respiratory viruses, will be discussed.
- Published
- 2011
- Full Text
- View/download PDF
28. Understanding Human Coronavirus HCoV-NL63.
- Author
-
Abdul-Rasool S and Fielding BC
- Abstract
Even though coronavirus infection of humans is not normally associated with severe diseases, the identification of the coronavirus responsible for the outbreak of severe acute respiratory syndrome showed that highly pathogenic coronaviruses can enter the human population. Shortly thereafter, in Holland in 2004, another novel human coronavirus (HCoV-NL63) was isolated from a seven-month old infant suffering from respiratory symptoms. This virus has subsequently been identified in various countries, indicating a worldwide distribution. HCoV-NL63 has been shown to infect mainly children and the immunocommpromised, who presented with either mild upper respiratory symptoms (cough, fever and rhinorrhoea) or more serious lower respiratory tract involvement such as bronchiolitis and croup, which was observed mainly in younger children. In fact, HCoV-NL63 is the aetiological agent for up to 10% of all respiratory diseases. This review summarizes recent findings of human coronavirus HCoV-NL63 infections, including isolation and identification, phylogeny and taxonomy, genome structure and transcriptional regulation, transmission and pathogenesis, and detection and diagnosis.
- Published
- 2010
- Full Text
- View/download PDF
29. Expression, purification and preliminary crystallographic analysis of recombinant human DEAD-box polypeptide 5.
- Author
-
Choi YW, Dutta S, Fielding BC, and Tan YJ
- Subjects
- Crystallization, DEAD-box RNA Helicases genetics, DEAD-box RNA Helicases isolation & purification, Gene Expression, Humans, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins isolation & purification, X-Ray Diffraction, DEAD-box RNA Helicases chemistry
- Abstract
The DEAD-box RNA helicase DDX5 is involved in many aspects of RNA processing and has been implicated in a number of cellular processes involving alteration of RNA secondary structure. The N-terminal region of DDX5, which contains the conserved domain 1 of the DEAD-box helicases, has been cloned and expressed in Escherichia coli and purified. Here, the crystallization and preliminary diffraction analysis of this region is reported. X-ray diffraction data were processed to a resolution of 2.7 A. The crystals belonged to space group I222, with unit-cell parameters a = 66.18, b = 73.80, c = 104.00 A, alpha = beta = gamma = 90 degrees .
- Published
- 2010
- Full Text
- View/download PDF
30. Human coronavirus NL63 open reading frame 3 encodes a virion-incorporated N-glycosylated membrane protein.
- Author
-
Müller MA, van der Hoek L, Voss D, Bader O, Lehmann D, Schulz AR, Kallies S, Suliman T, Fielding BC, Drosten C, and Niedrig M
- Subjects
- Animals, Blotting, Western, Cell Line, Endoplasmic Reticulum chemistry, Golgi Apparatus chemistry, Humans, Lysosomes chemistry, Macaca mulatta, Microscopy, Confocal, Microscopy, Fluorescence, Models, Biological, Models, Molecular, Viral Structural Proteins analysis, Coronavirus genetics, Glycoproteins genetics, Glycoproteins metabolism, Open Reading Frames, Viral Matrix Proteins genetics, Viral Matrix Proteins metabolism, Virion chemistry
- Abstract
Background: Human pathogenic coronavirus NL63 (hCoV-NL63) is a group 1 (alpha) coronavirus commonly associated with respiratory tract infections. In addition to known non-structural and structural proteins all coronaviruses have one or more accessory proteins whose functions are mostly unknown. Our study focuses on hCoV-NL63 open reading frame 3 (ORF 3) which is a highly conserved accessory protein among coronaviruses., Results: In-silico analysis of the 225 amino acid sequence of hCoV-NL63 ORF 3 predicted a triple membrane-spanning protein. Expression in infected CaCo-2 and LLC-MK2 cells was confirmed by immunofluorescence and Western blot analysis. The protein was detected within the endoplasmatic reticulum/Golgi intermediate compartment (ERGIC) where coronavirus assembly and budding takes place. Subcellular localization studies using recombinant ORF 3 protein transfected in Huh-7 cells revealed occurrence in ERGIC, Golgi- and lysosomal compartments. By fluorescence microscopy of differently tagged envelope (E), membrane (M) and nucleocapsid (N) proteins it was shown that ORF 3 protein colocalizes extensively with E and M within the ERGIC. Using N-terminally FLAG-tagged ORF 3 protein and an antiserum specific to the C-terminus we verified the proposed topology of an extracellular N-terminus and a cytosolic C-terminus. By in-vitro translation analysis and subsequent endoglycosidase H digestion we showed that ORF 3 protein is N-glycosylated at the N-terminus. Analysis of purified viral particles revealed that ORF 3 protein is incorporated into virions and is therefore an additional structural protein., Conclusions: This study is the first extensive expression analysis of a group 1 hCoV-ORF 3 protein. We give evidence that ORF 3 protein is a structural N-glycosylated and virion-incorporated protein.
- Published
- 2010
- Full Text
- View/download PDF
31. The nonstructural protein 8 (nsp8) of the SARS coronavirus interacts with its ORF6 accessory protein.
- Author
-
Kumar P, Gunalan V, Liu B, Chow VT, Druce J, Birch C, Catton M, Fielding BC, Tan YJ, and Lal SK
- Subjects
- Animals, Cell Line, Chlorocebus aethiops, Cloning, Molecular, Cytoplasm chemistry, Immunoprecipitation, Microscopy, Confocal, Microscopy, Fluorescence, Protein Binding, Two-Hybrid System Techniques, Viral Nonstructural Proteins genetics, Severe acute respiratory syndrome-related coronavirus metabolism, Viral Nonstructural Proteins metabolism
- Abstract
Severe acute respiratory syndrome (SARS) coronavirus (SARS-CoV) caused a severe outbreak in several regions of the world in 2003. The SARS-CoV genome is predicted to contain 14 functional open reading frames (ORFs). The first ORF (1a and 1b) encodes a large polyprotein that is cleaved into nonstructural proteins (nsp). The other ORFs encode for four structural proteins (spike, membrane, nucleocapsid and envelope) as well as eight SARS-CoV-specific accessory proteins (3a, 3b, 6, 7a, 7b, 8a, 8b and 9b). In this report we have cloned the predicted nsp8 gene and the ORF6 gene of the SARS-CoV and studied their abilities to interact with each other. We expressed the two proteins as fusion proteins in the yeast two-hybrid system to demonstrate protein-protein interactions and tested the same using a yeast genetic cross. Further the strength of the interaction was measured by challenging growth of the positive interaction clones on increasing gradients of 2-amino trizole. The interaction was then verified by expressing both proteins separately in-vitro in a coupled-transcription translation system and by coimmunoprecipitation in mammalian cells. Finally, colocalization experiments were performed in SARS-CoV infected Vero E6 mammalian cells to confirm the nsp8-ORF6 interaction. To the best of our knowledge, this is the first report of the interaction between a SARS-CoV accessory protein and nsp8 and our findings suggest that ORF6 protein may play a role in virus replication.
- Published
- 2007
- Full Text
- View/download PDF
32. Over-expression of severe acute respiratory syndrome coronavirus 3b protein induces both apoptosis and necrosis in Vero E6 cells.
- Author
-
Khan S, Fielding BC, Tan TH, Chou CF, Shen S, Lim SG, Hong W, and Tan YJ
- Subjects
- Animals, Cell Nucleolus chemistry, Cell Nucleus chemistry, Chlorocebus aethiops, DNA Fragmentation, Microscopy, Fluorescence, Sequence Deletion, Time Factors, Vero Cells, Viral Nonstructural Proteins genetics, Viral Nonstructural Proteins metabolism, bcl-2-Associated X Protein physiology, Apoptosis, Necrosis, Severe acute respiratory syndrome-related coronavirus pathogenicity, Viral Nonstructural Proteins physiology
- Abstract
The genome of the severe acute respiratory syndrome coronavirus encodes for eight accessory viral proteins with no known homologues in other coronaviruses. One of these is the 3b protein, which is encoded by the second open reading frame in subgenomic RNA 3 and contains 154 amino acids. Here, a detailed time-course study was performed to compare the apoptosis and necrosis profiles induced by full-length 3b, a 3b mutant that was deleted by 30 amino acids from the C terminus (3bDelta124-154) and the classical apoptosis inducer, Bax. Our results showed that Vero E6 cells transfected with a construct for expressing 3b underwent necrosis as early as 6h after transfection and underwent simultaneous necrosis and apoptosis at later time-points. At all the time-points analysed, the apoptosis induced by the expression of 3b was less than the level induced by Bax but the level of necrosis was comparable. The 3bDelta124-154 mutant behaves in a similar manner indicating that the localization of the 3b protein does not seems to be important for the cell-death pathways since full-length 3b is localized predominantly to the nucleolus, while the mutant is found to be concentrated in the peri-nuclear regions. To our knowledge, this is the first report of the induction of necrosis by a SARS-CoV protein.
- Published
- 2006
- Full Text
- View/download PDF
33. ACE2 orthologues in non-mammalian vertebrates (Danio, Gallus, Fugu, Tetraodon and Xenopus).
- Author
-
Chou CF, Loh CB, Foo YK, Shen S, Fielding BC, Tan TH, Khan S, Wang Y, Lim SG, Hong W, Tan YJ, and Fu J
- Subjects
- Angiotensin-Converting Enzyme 2, Animals, Animals, Genetically Modified, Base Sequence, Chickens genetics, Chromosome Mapping, DNA, Complementary genetics, Exons, Green Fluorescent Proteins genetics, Humans, Introns, Molecular Sequence Data, Peptidyl-Dipeptidase A chemistry, Phylogeny, Promoter Regions, Genetic, Recombinant Proteins genetics, Takifugu genetics, Tetraodontiformes genetics, Xenopus genetics, Zebrafish genetics, Peptidyl-Dipeptidase A genetics, Vertebrates genetics
- Abstract
Angiotensin-converting enzyme 2 (ACE2), a newly identified member in the renin-angiotensin system (RAS), acts as a negative regulator of ACE. It is mainly expressed in cardiac blood vessels and the tubular epithelia of kidneys and abnormal expression has been implicated in diabetes, hypertension and heart failure. The mechanism and physiological function of this zinc metallopeptidase in mammals are not yet fully understood. Non-mammalian vertebrate models offer attractive and simple alternatives that could facilitate the exploration of ACE2 function. In this paper we report the in silico analysis of Ace2 genes from the Gallus (chicken), Xenopus (frog), Fugu and Tetraodon (pufferfish) genome assembly databases, and from the Danio (zebrafish) cDNA library. Exon ambiguities of Danio and Xenopus Ace2s were resolved by RT-PCR and 3'RACE. Analyses of the exon-intron structures, alignment, phylogeny and hydrophilicity plots, together with the conserved synteny among these vertebrates, support the orthologous relationship between mammalian and non-mammalian ACE2s. The putative promoters of Ace2 from human, Tetraodon and Xenopus tropicalis drove the expression of enhanced green fluorescent protein (EGFP) specifically in the heart tissue of transgenic Xenopus thus making it a suitable model for future functional genomic studies. Additionally, the search for conserved cis-elements resulted in the discovery of WGATAR motifs in all the putative Ace2 promoters from 7 different animals, suggesting a possible role of GATA family transcriptional factors in regulating the expression of Ace2.
- Published
- 2006
- Full Text
- View/download PDF
34. Severe acute respiratory syndrome coronavirus protein 7a interacts with hSGT.
- Author
-
Fielding BC, Gunalan V, Tan TH, Chou CF, Shen S, Khan S, Lim SG, Hong W, and Tan YJ
- Subjects
- Amino Acid Sequence, Animals, Carrier Proteins genetics, Chlorocebus aethiops, Humans, Molecular Chaperones, Molecular Sequence Data, Mutation, Protein Binding, Two-Hybrid System Techniques, Vero Cells, Carrier Proteins metabolism, Membrane Proteins metabolism, Severe acute respiratory syndrome-related coronavirus, Viral Envelope Proteins metabolism, Viral Matrix Proteins metabolism, Viral Proteins metabolism, Viral Structural Proteins metabolism
- Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV) 7a is an accessory protein with no known homologues. In this study, we report the interaction of a SARS-CoV 7a and small glutamine-rich tetratricopeptide repeat-containing protein (SGT). SARS-CoV 7a and human SGT interaction was identified using a two-hybrid system screen and confirmed with interaction screens in cell culture and cellular co-localization studies. The SGT domain of interaction was mapped by deletion mutant analysis and results indicated that tetratricopeptide repeat 2 (aa 125-158) was essential for interaction. We also showed that 7a interacted with SARS-CoV structural proteins M (membrane) and E (envelope), which have been shown to be essential for virus-like particle formation. Taken together, our results coupled with data from studies of the interaction between SGT and HIV-1 vpu indicated that SGT could be involved in the life-cycle, possibly assembly of SARS-CoV.
- Published
- 2006
- Full Text
- View/download PDF
35. Monoclonal antibodies targeting the HR2 domain and the region immediately upstream of the HR2 of the S protein neutralize in vitro infection of severe acute respiratory syndrome coronavirus.
- Author
-
Lip KM, Shen S, Yang X, Keng CT, Zhang A, Oh HL, Li ZH, Hwang LA, Chou CF, Fielding BC, Tan TH, Mayrhofer J, Falkner FG, Fu J, Lim SG, Hong W, and Tan YJ
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Monoclonal immunology, Cell Line, Epitopes immunology, Humans, Membrane Fusion immunology, Membrane Glycoproteins chemistry, Membrane Glycoproteins genetics, Molecular Sequence Data, Neutralization Tests, Protein Structure, Tertiary, Severe acute respiratory syndrome-related coronavirus metabolism, Severe Acute Respiratory Syndrome immunology, Spike Glycoprotein, Coronavirus, Viral Envelope Proteins chemistry, Viral Envelope Proteins genetics, Antibodies, Viral immunology, Membrane Glycoproteins immunology, Severe acute respiratory syndrome-related coronavirus immunology, Severe Acute Respiratory Syndrome virology, Viral Envelope Proteins immunology
- Abstract
We have previously shown that an Escherichia coli-expressed, denatured spike (S) protein fragment of the severe acute respiratory coronavirus, containing residues 1029 to 1192 which include the heptad repeat 2 (HR2) domain, was able to induce neutralizing polyclonal antibodies (C. T. Keng, A. Zhang, S. Shen, K. M. Lip, B. C. Fielding, T. H. Tan, C. F. Chou, C. B. Loh, S. Wang, J. Fu, X. Yang, S. G. Lim, W. Hong, and Y. J. Tan, J. Virol. 79:3289-3296, 2005). In this study, monoclonal antibodies (MAbs) were raised against this fragment to identify the linear neutralizing epitopes in the functional domain and to investigate the mechanisms involved in neutralization. Eighteen hybridomas secreting the S protein-specific MAbs were obtained. Binding sites of these MAbs were mapped to four linear epitopes. Two of them were located within the HR2 region and two immediately upstream of the HR2 domain. MAbs targeting these epitopes showed in vitro neutralizing activities and were able to inhibit cell-cell membrane fusion. These results provide evidence of novel neutralizing epitopes that are located in the HR2 domain and the spacer region immediately upstream of the HR2 of the S protein.
- Published
- 2006
- Full Text
- View/download PDF
36. The severe acute respiratory syndrome coronavirus 3a protein up-regulates expression of fibrinogen in lung epithelial cells.
- Author
-
Tan YJ, Tham PY, Chan DZ, Chou CF, Shen S, Fielding BC, Tan TH, Lim SG, and Hong W
- Subjects
- Animals, Blotting, Western, Cell Line, Chlorocebus aethiops, Epithelial Cells metabolism, Epithelial Cells virology, Fibrinogen genetics, Humans, Polymerase Chain Reaction, RNA, Messenger analysis, Up-Regulation, Vero Cells, Viral Envelope Proteins, Viroporin Proteins, Fibrinogen metabolism, Severe acute respiratory syndrome-related coronavirus, Viral Proteins metabolism
- Abstract
Here we analyzed the gene expression profile of cells that stably express the severe acute respiratory syndrome coronavirus (SARS-CoV) 3a protein to determine its effects on host functions. A lung epithelial cell-line, A549, was chosen for this study because the lung is the primary organ infected by SARS-CoV and fatalities resulted mainly from pulmonary complications. Our results showed that the expression of 3a up-regulates the mRNA levels of all three subunits, Aalpha, Bbeta, and gamma, of fibrinogen. Consequently, the intracellular levels as well as the secretion of fibrinogen were increased. We also observed increased fibrinogen levels in SARS-CoV-infected Vero E6 cells.
- Published
- 2005
- Full Text
- View/download PDF
37. Genetic lesions within the 3a gene of SARS-CoV.
- Author
-
Tan TH, Barkham T, Fielding BC, Chou CF, Shen S, Lim SG, Hong W, and Tan YJ
- Subjects
- Humans, RNA, Viral genetics, Viral Envelope Proteins, Viroporin Proteins, Frameshift Mutation, Severe acute respiratory syndrome-related coronavirus genetics, Severe acute respiratory syndrome-related coronavirus isolation & purification, Severe Acute Respiratory Syndrome virology, Viral Proteins genetics
- Abstract
A series of frameshift mutations within the 3a gene has been observed in culture-derived severe acute respiratory syndrome coronavirus (SARS-CoV). We report here that viral RNA from clinical samples obtained from SARS-CoV infected patients also contains a heterogeneous population of wild-type and mutant 3a transcripts.
- Published
- 2005
- Full Text
- View/download PDF
38. Amino acids 1055 to 1192 in the S2 region of severe acute respiratory syndrome coronavirus S protein induce neutralizing antibodies: implications for the development of vaccines and antiviral agents.
- Author
-
Keng CT, Zhang A, Shen S, Lip KM, Fielding BC, Tan TH, Chou CF, Loh CB, Wang S, Fu J, Yang X, Lim SG, Hong W, and Tan YJ
- Subjects
- Animals, Antibodies, Viral immunology, Blotting, Western, Cells, Cultured, Chlorocebus aethiops, Cloning, Molecular, Epitopes genetics, Epitopes immunology, Escherichia coli genetics, Fluorescent Antibody Technique, Immunoprecipitation, Membrane Glycoproteins genetics, Membrane Glycoproteins physiology, Neutralization Tests, Rabbits, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins immunology, Recombinant Fusion Proteins isolation & purification, Spike Glycoprotein, Coronavirus, Viral Envelope Proteins genetics, Viral Envelope Proteins physiology, Epitope Mapping, Membrane Glycoproteins immunology, Severe acute respiratory syndrome-related coronavirus immunology, Viral Envelope Proteins immunology
- Abstract
The spike (S) protein of the severe acute respiratory syndrome coronavirus (SARS-CoV) interacts with cellular receptors to mediate membrane fusion, allowing viral entry into host cells; hence it is recognized as the primary target of neutralizing antibodies, and therefore knowledge of antigenic determinants that can elicit neutralizing antibodies could be beneficial for the development of a protective vaccine. Here, we expressed five different fragments of S, covering the entire ectodomain (amino acids 48 to 1192), as glutathione S-transferase fusion proteins in Escherichia coli and used the purified proteins to raise antibodies in rabbits. By Western blot analysis and immunoprecipitation experiments, we showed that all the antibodies are specific and highly sensitive to both the native and denatured forms of the full-length S protein expressed in virus-infected cells and transfected cells, respectively. Indirect immunofluorescence performed on fixed but unpermeabilized cells showed that these antibodies can recognize the mature form of S on the cell surface. All the antibodies were also able to detect the maturation of the 200-kDa form of S to the 210-kDa form by pulse-chase experiments. When the antibodies were tested for their ability to inhibit SARS-CoV propagation in Vero E6 culture, it was found that the anti-SDelta10 antibody, which was targeted to amino acid residues 1029 to 1192 of S, which include heptad repeat 2, has strong neutralizing activities, suggesting that this region of S carries neutralizing epitopes and is very important for virus entry into cells.
- Published
- 2005
- Full Text
- View/download PDF
39. A novel cell-based binding assay system reconstituting interaction between SARS-CoV S protein and its cellular receptor.
- Author
-
Chou CF, Shen S, Tan YJ, Fielding BC, Tan TH, Fu J, Xu Q, Lim SG, and Hong W
- Subjects
- Animals, CHO Cells, Chlorocebus aethiops, Cricetinae, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Membrane Glycoproteins genetics, Spike Glycoprotein, Coronavirus, Vero Cells, Viral Envelope Proteins genetics, Membrane Glycoproteins metabolism, Receptors, Virus metabolism, Severe acute respiratory syndrome-related coronavirus metabolism, Viral Envelope Proteins metabolism
- Abstract
Severe acute respiratory syndrome (SARS), a life-threatening disease, is caused by the newly identified virus SARS coronavirus (SARS-CoV). In order to study the spike (S) protein of this highly contagious virus, we established a clonal cell-line, CHO-SG, from the Chinese hamster ovary cells that stably expresses C-terminally EGFP-tagged SARS-CoV S protein (S-EGFP). The ectodomain of the S glycoprotein is localized on the surface of CHO-SG cells with N-acetyl-glucosamine-terminated carbohydrate structure. CHO-SG cells associated tightly with Vero E6 cells, a SARS-CoV receptor (ACE2) expressing cell-line, and the interaction remained stable under highly stringent condition (1M NaCl). This interaction could be blocked by either the serum from a SARS convalescent patient or a goat anti-ACE2 antibody, indicating that the interaction is specific. A binding epitope with lesser degree of glycosylation and native conformation was localized by using rabbit anti-sera raised against five denatured recombinant S protein fragments expressed in Escherichia coli. One of the sera obtained from the fragment encompassing amino acids 48-358 significantly blocked the interaction between CHO-SG and Vero E6 cells. The region is useful for studying neutralizing antibodies in future vaccine development. This paper describes an easy and safe cell-based assay suitable for studying the binding between SARS-CoV S protein and its receptor.
- Published
- 2005
- Full Text
- View/download PDF
40. Overexpression of 7a, a protein specifically encoded by the severe acute respiratory syndrome coronavirus, induces apoptosis via a caspase-dependent pathway.
- Author
-
Tan YJ, Fielding BC, Goh PY, Shen S, Tan TH, Lim SG, and Hong W
- Subjects
- Animals, COS Cells, Caspase 3, Cell Line, Chlorocebus aethiops, HeLa Cells, Humans, Membrane Proteins genetics, Severe acute respiratory syndrome-related coronavirus genetics, Vero Cells, Viral Matrix Proteins, Viral Proteins genetics, Apoptosis, Caspases metabolism, Membrane Proteins metabolism, Severe acute respiratory syndrome-related coronavirus pathogenicity, Viral Proteins metabolism
- Abstract
Besides genes that are homologous to proteins found in other coronaviruses, the severe acute respiratory syndrome coronavirus genome also contains nine other potential open reading frames. Previously, we have characterized the expression and cellular localization of two of these "accessory" viral proteins, 3a (previously termed U274) and 7a (previously termed U122). In this study, we further examined whether they can induce apoptosis, which has been observed clinically. We showed that the overexpression of 7a, but not of 3a or the viral structural proteins, nucleocapsid, membrane, and envelope, induces apoptosis. 7a induces apoptosis via a caspase-dependent pathway and in cell lines derived from different organs, including lung, kidney, and liver.
- Published
- 2004
- Full Text
- View/download PDF
41. Characterization of a unique group-specific protein (U122) of the severe acute respiratory syndrome coronavirus.
- Author
-
Fielding BC, Tan YJ, Shuo S, Tan TH, Ooi EE, Lim SG, Hong W, and Goh PY
- Subjects
- Amino Acid Sequence, Animals, Chlorocebus aethiops, Endoplasmic Reticulum metabolism, Humans, Membrane Proteins chemistry, Molecular Sequence Data, Open Reading Frames genetics, Protein Sorting Signals, Severe acute respiratory syndrome-related coronavirus genetics, Severe acute respiratory syndrome-related coronavirus pathogenicity, Species Specificity, Transfection, Vero Cells, Viral Matrix Proteins, Viral Proteins chemistry, Membrane Proteins genetics, Membrane Proteins metabolism, Severe acute respiratory syndrome-related coronavirus metabolism, Viral Proteins genetics, Viral Proteins metabolism
- Abstract
A novel coronavirus (CoV) has been identified as the etiological agent of severe acute respiratory syndrome (SARS). The SARS-CoV genome encodes the characteristic essential CoV replication and structural proteins. Additionally, the genome contains six group-specific open reading frames (ORFs) larger than 50 amino acids, with no known homologues. As with the group-specific genes of the other CoVs, little is known about the SARS-CoV group-specific genes. SARS-CoV ORF7a encodes a putative unique 122-amino-acid protein, designated U122 in this study. The deduced sequence contains a probable cleaved signal sequence and a C-terminal transmembrane helix, indicating that U122 is likely to be a type I membrane protein. The C-terminal tail also contains a typical endoplasmic reticulum (ER) retrieval motif, KRKTE. U122 was expressed in SARS-CoV-infected Vero E6 cells, as it could be detected by Western blot and immunofluorescence analyses. U122 is localized to the perinuclear region of both SARS-CoV-infected and transfected cells and colocalized with ER and intermediate compartment markers. Mutational analyses showed that both the signal peptide sequence and ER retrieval motif were functional.
- Published
- 2004
- Full Text
- View/download PDF
42. A novel severe acute respiratory syndrome coronavirus protein, U274, is transported to the cell surface and undergoes endocytosis.
- Author
-
Tan YJ, Teng E, Shen S, Tan TH, Goh PY, Fielding BC, Ooi EE, Tan HC, Lim SG, and Hong W
- Subjects
- Amino Acid Sequence, Animals, Chlorocebus aethiops, Humans, Molecular Sequence Data, Protein Sorting Signals, Rabbits, Severe acute respiratory syndrome-related coronavirus genetics, Severe acute respiratory syndrome-related coronavirus metabolism, Transfection, Vero Cells, Viral Proteins chemistry, Viral Proteins genetics, Biological Transport, Endocytosis, Severe acute respiratory syndrome-related coronavirus pathogenicity, Viral Proteins metabolism
- 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.
- Published
- 2004
- Full Text
- View/download PDF
43. Profiles of antibody responses against severe acute respiratory syndrome coronavirus recombinant proteins and their potential use as diagnostic markers.
- Author
-
Tan YJ, Goh PY, Fielding BC, Shen S, Chou CF, Fu JL, Leong HN, Leo YS, Ooi EE, Ling AE, Lim SG, and Hong W
- Subjects
- Animals, Antibodies, Viral blood, Antibody Specificity, Antigens, Viral genetics, Antigens, Viral immunology, Cells, Cultured, Fluorescent Antibody Technique, Humans, Immunoglobulin A blood, Immunoglobulin G blood, Immunoglobulin M blood, Plasmids, Severe acute respiratory syndrome-related coronavirus isolation & purification, Sensitivity and Specificity, Biomarkers, Severe acute respiratory syndrome-related coronavirus genetics, Severe acute respiratory syndrome-related coronavirus immunology, Severe Acute Respiratory Syndrome diagnosis, Severe Acute Respiratory Syndrome immunology
- Abstract
A new coronavirus (severe acute respiratory syndrome coronavirus [SARS-CoV]) has been identified to be the etiological agent of severe acute respiratory syndrome. Given the highly contagious and acute nature of the disease, there is an urgent need for the development of diagnostic assays that can detect SARS-CoV infection. For determination of which of the viral proteins encoded by the SARS-CoV genome may be exploited as diagnostic antigens for serological assays, the viral proteins were expressed individually in mammalian and/or bacterial cells and tested for reactivity with sera from SARS-CoV-infected patients by Western blot analysis. A total of 81 sera, including 67 from convalescent patients and seven pairs from two time points of infection, were analyzed, and all showed immunoreactivity towards the nucleocapsid protein (N). Sera from some of the patients also showed immunoreactivity to U274 (59 of 81 [73%]), a protein that is unique to SARS-CoV. In addition, all of the convalescent-phase sera showed immunoreactivity to the spike (S) protein when analyzed by an immunofluorescence method utilizing mammalian cells stably expressing S. However, samples from the acute phase (2 to 9 days after the onset of illness) did not react with S, suggesting that antibodies to N may appear earlier than antibodies to S. Alternatively, this could be due to the difference in the sensitivities of the two methods. The immunoreactivities to these recombinant viral proteins are highly specific, as sera from 100 healthy donors did not react with any of them. These results suggest that recombinant N, S, and U274 proteins may be used as antigens for the development of serological assays for SARS-CoV.
- Published
- 2004
- Full Text
- View/download PDF
44. The genetic organization of a 2,966 basepair DNA fragment of a single capsid nucleopolyhedrovirus isolated from Trichoplusia ni.
- Author
-
Fielding BC, Khan S, Wang W, Kruger C, Abrahams R, and Davison S
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, DNA, Viral analysis, Molecular Sequence Data, Nucleopolyhedroviruses classification, Nucleopolyhedroviruses isolation & purification, Open Reading Frames, Sequence Alignment, Genome, Viral, Nucleocapsid Proteins genetics, Nucleopolyhedroviruses genetics, Spodoptera virology, Viral Proteins genetics
- Abstract
In order to investigate the genomic organization of the Trichoplusia ni Single Capsid Nucleopolyhedrovirus (TnSNPV), a 2,966 basepairs (bp) genomic fragment was sequenced. The fragment was found to contain five open reading frames (ORFs) homologous to baculovirus genes, including p26, fibrillin (p10), AcMNPV ORF-29, late expression factor 6 (lef-6) and the C-terminal portion of p74, on either strand of DNA. Predicted amino acid sequences for the ORFs were compared and identity values of between 12% and 54% were observed. TnSNPV has previously been tentatively identified as a member of the Group II NPVs. Clustering and arrangement of the TnSNPV genes were similar to the clustering reported for SeMNPV, confirming TnSNPV as a Group II NPV.
- Published
- 2002
- Full Text
- View/download PDF
45. Identification and characterization of the Trichoplusia ni single capsid nuclear polyhedrosis virus p10 gene.
- Author
-
Fielding BC and Davison S
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Molecular Sequence Data, Open Reading Frames, Sequence Alignment, Sequence Analysis, DNA, Moths virology, Nucleocapsid Proteins genetics, Nucleopolyhedroviruses genetics, Viral Proteins genetics
- Abstract
The p10 gene was identified and characterized from the Trichoplusia ni single capsid nuclear polyhedrosis virus (TniSNPV). The p10 open reading frame (ORF) sequence was identified following sequencing of the ends of the EcoRI-G clone. Subsequent sequencing of an EcoRI-SmaI subclone identified the entire p10 and a portion of a p26 homologue. The p10 ORF of 264 basepairs (bps), encoded a predicted protein of 88 amino acids (aas) with Mr 9527 Da. The putative late transcription initiation motif (TAAG) was found upstream of the translation initiation codon at position -46. Downstream of the translation stop codon, a putative poly(A) signal was identified. The p10 amino acid sequence contained the three conserved domains reported for all other p10 genes. The p10 amino acid sequence was most homologous (85% similarity and 67% identity) to that of Buzura suppresaria NPV p10 sequence.
- Published
- 2000
- Full Text
- View/download PDF
46. The characterization and phylogenetic relationship of the Trichoplusia ni single capsid nuclear polyhedrosis virus polyhedrin gene.
- Author
-
Fielding BC and Davison S
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Molecular Sequence Data, Occlusion Body Matrix Proteins, Phylogeny, Sequence Alignment, Viral Proteins chemistry, Viral Structural Proteins, Capsid genetics, Lepidoptera virology, Nucleopolyhedroviruses genetics, Viral Proteins genetics
- Abstract
The polyhedrin gene (polh) was identified from the Trichoplusia ni (Tni) single capsid nuclear polyhedrosis virus (SNPV). An EcoRI fragment containing the truncated polyhedrin gene was detected by hybridization with an AcMNPV expression vector probe; the remaining portion of the gene was amplified by reverse PCR. An open reading frame (ORF) of 741 nucleotides (nt), encoding a putative protein of 246 amino acids (a.a) with Mr 28,780 Da was identified. The 5'-noncoding region contained the putative late (TAAG) transcription initiation motif. The 3' end, downstream of the translation stop codon, lacked an obvious putative poly (A) signal. Nucleotide and amino acid homology are greater than 80% to that of Mamestra brassicae polyhedrin sequences. Results suggest that T. niSNPV is a member of the group II nuclear polyhedrosis viruses.
- Published
- 1999
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.