1
|
Al Otaibi A, Al Shaikh Mubarak S, Al Hejji F, Almasaud A, Al Jami H, Iqbal J, Al Qarni A, Harbi NKA, Bakillah A. Thapsigargin and Tunicamycin Block SARS-CoV-2 Entry into Host Cells via Differential Modulation of Unfolded Protein Response (UPR), AKT Signaling, and Apoptosis. Cells 2024; 13:769. [PMID: 38727305 PMCID: PMC11083125 DOI: 10.3390/cells13090769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/05/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND SARS-Co-V2 infection can induce ER stress-associated activation of unfolded protein response (UPR) in host cells, which may contribute to the pathogenesis of COVID-19. To understand the complex interplay between SARS-Co-V2 infection and UPR signaling, we examined the effects of acute pre-existing ER stress on SARS-Co-V2 infectivity. METHODS Huh-7 cells were treated with Tunicamycin (TUN) and Thapsigargin (THA) prior to SARS-CoV-2pp transduction (48 h p.i.) to induce ER stress. Pseudo-typed particles (SARS-CoV-2pp) entry into host cells was measured by Bright GloTM luciferase assay. Cell viability was assessed by cell titer Glo® luminescent assay. The mRNA and protein expression was evaluated by RT-qPCR and Western Blot. RESULTS TUN (5 µg/mL) and THA (1 µM) efficiently inhibited the entry of SARS-CoV-2pp into host cells without any cytotoxic effect. TUN and THA's attenuation of virus entry was associated with differential modulation of ACE2 expression. Both TUN and THA significantly reduced the expression of stress-inducible ER chaperone GRP78/BiP in transduced cells. In contrast, the IRE1-XBP1s and PERK-eIF2α-ATF4-CHOP signaling pathways were downregulated with THA treatment, but not TUN in transduced cells. Insulin-mediated glucose uptake and phosphorylation of Ser307 IRS-1 and downstream p-AKT were enhanced with THA in transduced cells. Furthermore, TUN and THA differentially affected lipid metabolism and apoptotic signaling pathways. CONCLUSIONS These findings suggest that short-term pre-existing ER stress prior to virus infection induces a specific UPR response in host cells capable of counteracting stress-inducible elements signaling, thereby depriving SARS-Co-V2 of essential components for entry and replication. Pharmacological manipulation of ER stress in host cells might provide new therapeutic strategies to alleviate SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Abeer Al Otaibi
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Ahsa 31982, Saudi Arabia; (A.A.O.); (S.A.S.M.); (F.A.H.); (J.I.); (A.A.Q.)
- Biomedical Research Department, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Ahsa 36428, Saudi Arabia
- King Abdulaziz Hospital, Ministry of National Guard-Health Affairs (MNG-HA), Al Ahsa 36428, Saudi Arabia
| | - Sindiyan Al Shaikh Mubarak
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Ahsa 31982, Saudi Arabia; (A.A.O.); (S.A.S.M.); (F.A.H.); (J.I.); (A.A.Q.)
- Biomedical Research Department, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Ahsa 36428, Saudi Arabia
- King Abdulaziz Hospital, Ministry of National Guard-Health Affairs (MNG-HA), Al Ahsa 36428, Saudi Arabia
| | - Fatimah Al Hejji
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Ahsa 31982, Saudi Arabia; (A.A.O.); (S.A.S.M.); (F.A.H.); (J.I.); (A.A.Q.)
| | - Abdulrahman Almasaud
- Vaccine Development Unit, Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia; (A.A.); (H.A.J.); (N.K.A.H.)
| | - Haya Al Jami
- Vaccine Development Unit, Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia; (A.A.); (H.A.J.); (N.K.A.H.)
| | - Jahangir Iqbal
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Ahsa 31982, Saudi Arabia; (A.A.O.); (S.A.S.M.); (F.A.H.); (J.I.); (A.A.Q.)
- Biomedical Research Department, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Ahsa 36428, Saudi Arabia
- King Abdulaziz Hospital, Ministry of National Guard-Health Affairs (MNG-HA), Al Ahsa 36428, Saudi Arabia
| | - Ali Al Qarni
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Ahsa 31982, Saudi Arabia; (A.A.O.); (S.A.S.M.); (F.A.H.); (J.I.); (A.A.Q.)
- Biomedical Research Department, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Ahsa 36428, Saudi Arabia
- King Abdulaziz Hospital, Ministry of National Guard-Health Affairs (MNG-HA), Al Ahsa 36428, Saudi Arabia
| | - Naif Khalaf Al Harbi
- Vaccine Development Unit, Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia; (A.A.); (H.A.J.); (N.K.A.H.)
| | - Ahmed Bakillah
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Ahsa 31982, Saudi Arabia; (A.A.O.); (S.A.S.M.); (F.A.H.); (J.I.); (A.A.Q.)
- Biomedical Research Department, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Ahsa 36428, Saudi Arabia
- King Abdulaziz Hospital, Ministry of National Guard-Health Affairs (MNG-HA), Al Ahsa 36428, Saudi Arabia
| |
Collapse
|
2
|
Samman N, El-Boubbou K, Al-Muhalhil K, Ali R, Alaskar A, Alharbi NK, Nehdi A. MICaFVi: A Novel Magnetic Immuno-Capture Flow Virometry Nano-Based Diagnostic Tool for Detection of Coronaviruses. BIOSENSORS 2023; 13:553. [PMID: 37232914 PMCID: PMC10216117 DOI: 10.3390/bios13050553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/03/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023]
Abstract
COVID-19 has resulted in a pandemic that aggravated the world's healthcare systems, economies, and education, and caused millions of global deaths. Until now, there has been no specific, reliable, and effective treatment to combat the virus and its variants. The current standard tedious PCR-based tests have limitations in terms of sensitivity, specificity, turnaround time, and false negative results. Thus, an alternative, rapid, accurate, and sensitive diagnostic tool that can detect viral particles, without the need for amplification or viral replication, is central to infectious disease surveillance. Here, we report MICaFVi (Magnetic Immuno-Capture Flow Virometry), a novel precise nano-biosensor diagnostic assay for coronavirus detection which combines the MNP-based immuno-capture of viruses for enrichment followed by flow-virometry analysis, enabling the sensitive detection of viral particles and pseudoviruses. As proof of concept, virus-mimicking spike-protein-coated silica particles (VM-SPs) were captured using anti-spike-antibody-conjugated MNPs (AS-MNPs) followed by detection using flow cytometry. Our results showed that MICaFVi can successfully detect viral MERS-CoV/SARS-CoV-2-mimicking particles as well as MERS-CoV pseudoviral particles (MERSpp) with high specificity and sensitivity, where a limit of detection (LOD) of 3.9 µg/mL (20 pmol/mL) was achieved. The proposed method has great potential for designing practical, specific, and point-of-care testing for rapid and sensitive diagnoses of coronavirus and other infectious diseases.
Collapse
Affiliation(s)
- Nosaibah Samman
- Medical Research Core Facility and Platforms (MRCFP), King Abdullah International Medical Research Center (KAIMRC) & King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia
| | - Kheireddine El-Boubbou
- King Abdullah International Medical Research Center (KAIMRC) & King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia
- Nanomaterials for Bioimaging Group (nanoBIG), Facultad de Ciencias, Departamento de Física de Materiales, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Department of Chemistry, College of Science, University of Bahrain, Sakhir 32038, Bahrain
| | - Khawlah Al-Muhalhil
- Medical Research Core Facility and Platforms (MRCFP), King Abdullah International Medical Research Center (KAIMRC) & King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia
| | - Rizwan Ali
- Medical Research Core Facility and Platforms (MRCFP), King Abdullah International Medical Research Center (KAIMRC) & King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia
| | - Ahmed Alaskar
- Medical Research Core Facility and Platforms (MRCFP), King Abdullah International Medical Research Center (KAIMRC) & King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia
- Department of Oncology, King Abdulaziz Medical City, College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia
| | - Naif Khalaf Alharbi
- King Abdullah International Medical Research Center (KAIMRC) & King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia
| | - Atef Nehdi
- Medical Research Core Facility and Platforms (MRCFP), King Abdullah International Medical Research Center (KAIMRC) & King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia
- Department of Life Sciences, Faculty of Sciences of Gabes, University of Gabes, Gabes 6029, Tunisia
| |
Collapse
|
3
|
Application of Pseudotyped Viruses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1407:45-60. [PMID: 36920691 DOI: 10.1007/978-981-99-0113-5_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Highly pathogenic emerging and reemerging viruses have serious public health and socioeconomic implications. Although conventional live virus research methods can more reliably investigate disease pathogenicity and evaluate antiviral products, they usually depend on high-level biosafety laboratories and skilled researchers; these requirements hinder in vitro assessments of efficacy, as well as efforts to test vaccines and antibody drugs. In contrast, pseudotyped viruses (i.e., single-round infectious viruses that mimic the membrane structures of various live viruses) are widely used in studies of highly pathogenic viruses because they can be handled in biosafety level 2 facilities. This chapter provides a concise overview of various aspects of pseudotyped virus technologies, including (1) exploration of the mechanisms of viral infection; (2) evaluation of the efficacies of vaccines and monoclonal antibodies based on pseudovirion-based neutralization assay; (3) assessment of antiviral agents (i.e., antibody-based drugs and inhibitors); (4) establishment of animal models of pseudotyped virus infection in vivo; (5) investigation of the evolution, infectivity, and antigenicity of viral variants and viral glycosylation; and (6) prediction of antibody-dependent cell-mediated cytotoxic activity.
Collapse
|
4
|
Lipid Raft Integrity and Cellular Cholesterol Homeostasis Are Critical for SARS-CoV-2 Entry into Cells. Nutrients 2022; 14:nu14163417. [PMID: 36014919 PMCID: PMC9415163 DOI: 10.3390/nu14163417] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 12/20/2022] Open
Abstract
Lipid rafts in cell plasma membranes play a critical role in the life cycle of many viruses. However, the involvement of membrane cholesterol-rich lipid rafts in the entry of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) into target cells is not well known. In this study, we investigated whether the presence of cholesterol-rich microdomains is required for the entry of SARS-CoV-2 into host cells. Our results show that depletion of cholesterol in the rafts by methyl-beta-cyclodextrin (MβCD) treatment impaired the expression of the cell surface receptor angiotensin-converting enzyme 2 (ACE2), resulting in a significant increase in SARS-CoV-2 entry into cells. The effects exerted by MβCD could be substantially reversed by exogenous cholesterol replenishment. In contrast, disturbance of intracellular cholesterol homeostasis by statins or siRNA knockdown of key genes involved in the cholesterol biosynthesis and transport pathways reduced SARS-CoV-2 entry into cells. Our study also reveals that SREBP2-mediated cholesterol biosynthesis is involved in the process of SARS-CoV-2 entry in target cells. These results suggest that the host membrane cholesterol-enriched lipid rafts and cellular cholesterol homeostasis are essential for SARS-CoV-2 entry into cells. Pharmacological manipulation of intracellular cholesterol might provide new therapeutic strategies to alleviate SARS-CoV-2 entry into cells.
Collapse
|
5
|
Alharbi NK, Aljamaan F, Aljami HA, Alenazi MW, Albalawi H, Almasoud A, Alharthi FJ, Azhar EI, Barhoumi T, Bosaeed M, Gilbert SC, Hashem AM. Immunogenicity of High-Dose MVA-Based MERS Vaccine Candidate in Mice and Camels. Vaccines (Basel) 2022; 10:vaccines10081330. [PMID: 36016218 PMCID: PMC9413082 DOI: 10.3390/vaccines10081330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 11/16/2022] Open
Abstract
The Middle East respiratory syndrome coronavirus (MERS-CoV) is a zoonotic pathogen that can transmit from dromedary camels to humans, causing severe pneumonia, with a 35% mortality rate. Vaccine candidates have been developed and tested in mice, camels, and humans. Previously, we developed a vaccine based on the modified vaccinia virus Ankara (MVA) viral vector, encoding a full-length spike protein of MERS-CoV, MVA-MERS. Here, we report the immunogenicity of high-dose MVA-MERS in prime–boost vaccinations in mice and camels. Methods: Three groups of mice were immunised with MVA wild-type (MVA-wt) and MVA-MERS (MVA-wt/MVA-MERS), MVA-MERS/MVA-wt, or MVA-MERS/MVA-MERS. Camels were immunised with two doses of PBS, MVA-wt, or MVA-MERS. Antibody (Ab) responses were evaluated using ELISA and MERS pseudovirus neutralisation assays. Results: Two high doses of MVA-MERS induced strong Ab responses in both mice and camels, including neutralising antibodies. Anti-MVA Ab responses did not affect the immune responses to the vaccine antigen (MERS-CoV spike). Conclusions: MVA-MERS vaccine, administered in a homologous prime–boost regimen, induced high levels of neutralising anti-MERS-CoV antibodies in mice and camels. This could be considered for further development and evaluation as a dromedary vaccine to reduce MERS-CoV transmission to humans.
Collapse
Affiliation(s)
- Naif Khalaf Alharbi
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
- Correspondence:
| | - Fahad Aljamaan
- Animal Facilities, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
| | - Haya A. Aljami
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
| | - Mohammed W. Alenazi
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
| | - Hind Albalawi
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
| | - Abdulrahman Almasoud
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
| | - Fatima J. Alharthi
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
| | - Esam I. Azhar
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 22254, Saudi Arabia
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 22254, Saudi Arabia
| | - Tlili Barhoumi
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
| | - Mohammad Bosaeed
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
- Department of Medicine, King Abdulaziz Medical City, Riyadh 12746, Saudi Arabia
| | | | - Anwar M. Hashem
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah 22254, Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 22254, Saudi Arabia
| |
Collapse
|
6
|
Alharbi NK, Samman N, Alhayli S, Alghoribi MF, Almasoud A, Nehdi A. Development and Evaluation of Enzyme-Linked Viral Immune Capture Assay for Detection of SARS-CoV-2. Front Bioeng Biotechnol 2022; 10:898726. [PMID: 36003542 PMCID: PMC9393230 DOI: 10.3389/fbioe.2022.898726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/15/2022] [Indexed: 12/05/2022] Open
Abstract
The pandemic of COVID-19 was caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in 2019 and it has prompted unprecedented research activities for vaccines, therapeutics, and diagnostics. The real-time reverse transcriptase-polymerase chain reaction (RT-PCR) is the gold standard method of diagnosis; however, immune-based assays offer cost-effective, deployable, easy-to-read solutions for diagnosis and surveillance. Here, we present the development, optimization, and testing of an enzyme-linked viral immune capture assay (ELVICA). It utilizes the spike antigen as the detected target of the virus and antibody-coated beads to capture the virus and enrich the detection. This method can be readout by luminescent and colorimetric equipment. It can also be visualized by the imaging system, offering a variety of detection approaches. ELVICA showed specificity to SARS-CoV-2-pseudotyped viruses as compared to MERS-CoV-pseudotyped viruses. As compared to RT-PCR, ELVICA showed high compatibility in detecting the virus in patient respiratory samples, especially for samples that are below a Ct value of 32 in RT-PCR. This assay is readily adaptable for detecting other pathogens and serves as a quick and affordable diagnostic tool.
Collapse
Affiliation(s)
- Naif Khalaf Alharbi
- King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Science (KSAU-HS), Riyadh, Saudi Arabia
| | - Nosaibah Samman
- King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Science (KSAU-HS), Riyadh, Saudi Arabia
| | - Sadeem Alhayli
- King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Science (KSAU-HS), Riyadh, Saudi Arabia
| | - Majed F. Alghoribi
- King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Science (KSAU-HS), Riyadh, Saudi Arabia
| | - Abdulrahman Almasoud
- King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Science (KSAU-HS), Riyadh, Saudi Arabia
| | - Atef Nehdi
- King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Science (KSAU-HS), Riyadh, Saudi Arabia
- Department of Life Sciences, Faculty of Sciences of Gabes, University of Gabes, Gabes, Tunisia
| |
Collapse
|
7
|
Neerukonda SN, Vassell R, Weiss CD, Wang W. Measuring Neutralizing Antibodies to SARS-CoV-2 Using Lentiviral Spike-Pseudoviruses. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2452:305-314. [PMID: 35554914 DOI: 10.1007/978-1-0716-2111-0_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Assays measuring neutralizing antibodies (nAbs) against SARS-CoV-2 are used to evaluate serological responses after SARS-CoV-2 infection and the potency of therapeutic antibodies and preventive vaccines. It is therefore imperative that neutralization assays be sensitive, specific, quantitative, and scalable for high throughput. Pseudoviruses are excellent surrogates for highly pathogenic viruses such as SARS-CoV-2 because they can be safely used to measure nAbs in a biosafety level-2 laboratory. In addition, pseudoviruses allow for easy introduction of mutations to study the effect of amino acid changes in the spike protein. In this chapter, we describe a recently optimized assay for measuring neutralizing antibodies to SARS-CoV-2 that uses a HIV-based lentiviral vector pseudotyped with the spike glycoprotein of SARS-CoV-2 to infect 293T cells stably expressing ACE2 and TMPRSS2.
Collapse
Affiliation(s)
- Sabari Nath Neerukonda
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research and Review, US Food and Drug Administration, Silver Spring, MD, USA
| | - Russell Vassell
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research and Review, US Food and Drug Administration, Silver Spring, MD, USA
| | - Carol D Weiss
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research and Review, US Food and Drug Administration, Silver Spring, MD, USA.
| | - Wei Wang
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research and Review, US Food and Drug Administration, Silver Spring, MD, USA.
| |
Collapse
|
8
|
Bosaeed M, Balkhy HH, Almaziad S, Aljami HA, Alhatmi H, Alanazi H, Alahmadi M, Jawhary A, Alenazi MW, Almasoud A, Alanazi R, Bittaye M, Aboagye J, Albaalharith N, Batawi S, Folegatti P, Ramos Lopez F, Ewer K, Almoaikel K, Aljeraisy M, Alothman A, Gilbert SC, Khalaf Alharbi N. Safety and immunogenicity of ChAdOx1 MERS vaccine candidate in healthy Middle Eastern adults (MERS002): an open-label, non-randomised, dose-escalation, phase 1b trial. THE LANCET. MICROBE 2022; 3:e11-e20. [PMID: 34751259 PMCID: PMC8565931 DOI: 10.1016/s2666-5247(21)00193-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
BACKGROUND ChAdOx1-vectored vaccine candidates against several pathogens have been developed and tested in clinical trials and ChAdOx1 nCoV-19 has now been licensed for emergency use for COVID-19. We assessed the safety and immunogenicity of the ChAdOx1 MERS vaccine in a phase 1b trial in healthy Middle Eastern adults. METHOD MERS002 is an open-label, non-randomised, dose-escalation, phase 1b trial. Healthy Middle Eastern adults aged 18-50 years were included in the study. ChAdOx1 MERS was administered as a single intramuscular injection into the deltoid muscle of the non-dominant arm at three different dose groups: 5·0 × 109 viral particles in a low-dose group, 2·5 × 1010 viral particles in an intermediate-dose group, and 5·0 × 1010 viral particles in a high-dose group. The primary objective was to assess the safety and tolerability of ChAdOx1 MERS, measured by the occurrence of solicited and unsolicited adverse events after vaccination for up to 28 days and occurrence of serious adverse events up to 6 months. The study is registered with ClinicalTrials.gov, NCT04170829. FINDINGS Between Dec 17, 2019, and June 1, 2020, 24 participants were enrolled (six to the low-dose, nine to the intermediate-dose, and nine to the high-dose group) and received a dose; 23 were available for follow-up at 6 months. The one dose of ChAdOx1 MERS vaccine was well tolerated with no serious adverse event reported during the 6 months of follow-up. Most adverse events were mild (67, 74%) and moderate (17, 19%). Six (7%) severe adverse events were reported by two participants in the intermediate-dose group (two feverish, two headache, one joint pain, and one muscle pain). Pain at the injection site was the most common local and overall adverse event, reported by 15 (63%) of the 24 participants. The most common systemic adverse event was headache, reported by 14 (58%), followed by muscle pain reported by 13 (54%). The vaccine induced both antibody and T cell immune responses in all volunteers; antibodies peaked at day 28 and T cell responses peaked at day 14; and continued until the end of follow-up at 6 months. INTERPRETATION The acceptable safety and immunogenicity data from this phase 1b trial of ChAdOx1 MERS vaccine candidate in Healthy Middle Eastern adults, combined with previous safety and immunogenicity data from a trial in the UK, support selecting the ChAdOx1 MERS vaccine for advancement into phase 2 clinical evaluation. FUNDING UK Department of Health and Social Care, using UK Aid funding, managed by the UK National Institute for Health Research; and King Abdullah International Medical Research Center.
Collapse
Affiliation(s)
- Mohammad Bosaeed
- Clinical Trial Services, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Department of Medicine, King Abdulaziz Medical City in Riyadh, Riyadh, Saudi Arabia
| | | | - Sultan Almaziad
- Department of Medicine, King Abdulaziz Medical City in Riyadh, Riyadh, Saudi Arabia
| | - Haya A Aljami
- Vaccine Development Unit, Infectious Disease Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Hind Alhatmi
- Department of Medicine, King Abdulaziz Medical City in Riyadh, Riyadh, Saudi Arabia
| | - Hala Alanazi
- Clinical Trial Services, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Mashael Alahmadi
- Vaccine Development Unit, Infectious Disease Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Ayah Jawhary
- Clinical Trial Services, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Mohammed W Alenazi
- Vaccine Development Unit, Infectious Disease Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Abdulrahman Almasoud
- Vaccine Development Unit, Infectious Disease Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Rawan Alanazi
- Clinical Trial Services, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Mustapha Bittaye
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Jeremy Aboagye
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Nahla Albaalharith
- Department of Nursing, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Sarah Batawi
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Pedro Folegatti
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Fernando Ramos Lopez
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Katie Ewer
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Khalid Almoaikel
- Clinical Trial Services, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Majed Aljeraisy
- Clinical Trial Services, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Adel Alothman
- Clinical Trial Services, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Department of Medicine, King Abdulaziz Medical City in Riyadh, Riyadh, Saudi Arabia
| | - Sarah C Gilbert
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Naif Khalaf Alharbi
- Vaccine Development Unit, Infectious Disease Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
9
|
An X, Martinez-Paniagua M, Rezvan A, Sefat SR, Fathi M, Singh S, Biswas S, Pourpak M, Yee C, Liu X, Varadarajan N. Single-dose intranasal vaccination elicits systemic and mucosal immunity against SARS-CoV-2. iScience 2021; 24:103037. [PMID: 34462731 PMCID: PMC8388188 DOI: 10.1016/j.isci.2021.103037] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 05/21/2021] [Accepted: 08/23/2021] [Indexed: 12/18/2022] Open
Abstract
Despite remarkable progress in the development and authorization of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), there is a need to validate vaccine platforms for broader application. The current intramuscular vaccines are designed to elicit systemic immunity without conferring mucosal immunity in the nasal compartment, which is the first barrier that SARS-CoV-2 virus breaches before dissemination to the lung. We report the development of an intranasal subunit vaccine that uses lyophilized spike protein and liposomal STING agonist as an adjuvant. This vaccine induces systemic neutralizing antibodies, IgA in the lung and nasal compartments, and T-cell responses in the lung of mice. Single-cell RNA sequencing confirmed the coordinated activation of T/B-cell responses in a germinal center-like manner within the nasal-associated lymphoid tissues, confirming its role as an inductive site to enable durable immunity. The ability to elicit immunity in the respiratory tract can prevent the establishment of infection in individuals and prevent disease transmission.
Collapse
Affiliation(s)
- Xingyue An
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77204, USA
| | - Melisa Martinez-Paniagua
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77204, USA
| | - Ali Rezvan
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77204, USA
| | - Samiur Rahman Sefat
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77204, USA
| | - Mohsen Fathi
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77204, USA
| | - Shailbala Singh
- Department of Melanoma Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Sujit Biswas
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | | | - Cassian Yee
- Department of Melanoma Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Xinli Liu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Navin Varadarajan
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
10
|
Alserehi HA, Alqunaibet AM, Al-Tawfiq JA, Alharbi NK, Alshukairi AN, Alanazi KH, Bin Saleh GM, Alshehri AM, Almasoud A, Hashem AM, Alruwaily AR, Alaswad RH, Al-Mutlaq HM, Almudaiheem AA, Othman FM, Aldakeel SA, Abu Ghararah MR, Jokhdar HA, Algwizani AR, Almudarra SS, Albarrag AM. Seroprevalence of SARS-CoV-2 (COVID-19) among healthcare workers in Saudi Arabia: comparing case and control hospitals. Diagn Microbiol Infect Dis 2020; 99:115273. [PMID: 33296851 PMCID: PMC7677039 DOI: 10.1016/j.diagmicrobio.2020.115273] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 11/08/2020] [Accepted: 11/15/2020] [Indexed: 01/27/2023]
Abstract
Healthcare workers (HCWs) stand at the frontline for fighting coronavirus disease 2019 (COVID-19) pandemic. This puts them at higher risk of acquiring the infection than other individuals in the community. Defining immunity status among health care workers is therefore of interest since it helps to mitigate the exposure risk. This study was conducted between May 20th and 30th, 2020. Eighty-five hospitals across Kingdom of Saudi Arabia were divided into 2 groups: COVID-19 referral hospitals are those to which RT-PCR-confirmed COVID-19 patients were admitted or referred for management (Case-hospitals). COVID-19 nonaffected hospitals where no COVID-19 patients had been admitted or managed and no HCW outbreak (Control hospitals). Next, seroprevalence of severe acute respiratory syndrome coronavirus 2 among HCWs was evaluated; there were 12,621 HCWs from the 85 hospitals. There were 61 case-hospitals with 9379 (74.3%) observations, and 24 control-hospitals with 3242 (25.7%) observations. The overall positivity rate by the immunoassay was 299 (2.36%) with a significant difference between the case-hospital (2.9%) and the control-group (0.8%) (P value <0.001). There was a wide variation in the positivity rate between regions and/or cities in Saudi Arabia, ranging from 0% to 6.31%. Of the serology positive samples, 100 samples were further tested using the SAS2pp neutralization assay; 92 (92%) samples showed neutralization activity. The seropositivity rate in Kingdom of Saudi Arabia is low and varies across different regions with higher positivity in case-hospitals than control-hospitals. The lack of neutralizing antibodies (NAb) in 8% of the tested samples could mean that assay is a more sensitive assay or that neutralization assay has a lower detection limits; or possibly that some samples had cross-reaction to spike protein of other coronaviruses in the assay, but these were not specific to neutralize severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
Collapse
Affiliation(s)
- Haleema Ali Alserehi
- Executive Department of Global Health, Epidemiology, surveillance and preparedness affairs, Executive Department of Research, Saudi Center for Disease Prevention and Control, Riyadh, Saudi Arabia.
| | - Ada Mohammed Alqunaibet
- Executive Department of Global Health, Epidemiology, surveillance and preparedness affairs, Executive Department of Research, Saudi Center for Disease Prevention and Control, Riyadh, Saudi Arabia
| | - Jaffar A Al-Tawfiq
- Infectious Disease Unit, Specialty Internal Medicine, and Quality and Patient Safety Departement, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia; Infectious Disease Division, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA; Infectious Disease Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Naif Khalaf Alharbi
- Vaccine Development Unit, Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Abeer Nizar Alshukairi
- Department of medicine, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | | | | | - Amer Mohammed Alshehri
- Public Health Lab, Saudi Center for Disease Prevention and Control, Riyadh, Saudi Arabia
| | - Abdulrahman Almasoud
- Vaccine Development Unit, Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Anwar M Hashem
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia; Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Amaal Rabie Alruwaily
- Executive Department of Global Health, Epidemiology, surveillance and preparedness affairs, Executive Department of Research, Saudi Center for Disease Prevention and Control, Riyadh, Saudi Arabia
| | - Rehab Habeeb Alaswad
- Executive Department of Global Health, Epidemiology, surveillance and preparedness affairs, Executive Department of Research, Saudi Center for Disease Prevention and Control, Riyadh, Saudi Arabia
| | - Hind Mohammed Al-Mutlaq
- Executive Department of Global Health, Epidemiology, surveillance and preparedness affairs, Executive Department of Research, Saudi Center for Disease Prevention and Control, Riyadh, Saudi Arabia
| | - Abdulllah Ali Almudaiheem
- Executive Department of Global Health, Epidemiology, surveillance and preparedness affairs, Executive Department of Research, Saudi Center for Disease Prevention and Control, Riyadh, Saudi Arabia
| | - Fatmah Mahmoud Othman
- Research department, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | | | | | | | | | - Sami Saeed Almudarra
- Executive Department of Global Health, Epidemiology, surveillance and preparedness affairs, Executive Department of Research, Saudi Center for Disease Prevention and Control, Riyadh, Saudi Arabia
| | - Ahmed Mohammed Albarrag
- Public Health Lab, Saudi Center for Disease Prevention and Control, Riyadh, Saudi Arabia; Department of Pathology, School of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
11
|
Liu K, Zou R, Cui W, Li M, Wang X, Dong J, Li H, Li H, Wang P, Shao X, Su W, Chan HCS, Li H, Yuan S. Clinical HDAC Inhibitors Are Effective Drugs to Prevent the Entry of SARS-CoV2. ACS Pharmacol Transl Sci 2020; 3:1361-1370. [PMID: 34778724 PMCID: PMC7671100 DOI: 10.1021/acsptsci.0c00163] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Indexed: 12/13/2022]
Abstract
![]()
The
outbreak of COVID-19 by the end of 2019 has posed serious health
threats to humanity and jeopardized the global economy. However, no
effective drugs are available to treat COVID-19 currently and there
is a great demand to fight against it. Here, we combined computational
screening and an efficient cellular pseudotyped virus system, confirming
that clinical HDAC inhibitors can efficiently prevent SARS-CoV-2 and
potentially be used to fight against COVID-19.
Collapse
Affiliation(s)
- Ke Liu
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Rongfeng Zou
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Wenqiang Cui
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Meiqing Li
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xueying Wang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Junlin Dong
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Hongchun Li
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Hongpei Li
- College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Peihui Wang
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Ximing Shao
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Wu Su
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - H. C. Stephen Chan
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Hongchang Li
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Shuguang Yuan
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| |
Collapse
|
12
|
High Rate of Circulating MERS-CoV in Dromedary Camels at Slaughterhouses in Riyadh, 2019. Viruses 2020; 12:v12111215. [PMID: 33120981 PMCID: PMC7692456 DOI: 10.3390/v12111215] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 10/02/2020] [Accepted: 10/09/2020] [Indexed: 12/04/2022] Open
Abstract
MERS-CoV is a zoonotic virus that has emerged in humans in 2012 and caused severe respiratory illness with a mortality rate of 34.4%. Since its appearance, MERS-CoV has been reported in 27 countries and most of these cases were in Saudi Arabia. So far, dromedaries are considered to be the intermediate host and the only known source of human infection. This study was designed to determine the seroprevalence and the infection rate of MERS-CoV in slaughtered food-camels in Riyadh, Saudi Arabia. A total of 171 nasal swabs along with 161 serum samples were collected during the winter; from January to April 2019. Nasal swabs were examined by Rapid test and RT-PCR to detect MERS-CoV RNA, while serum samples were tested primarily using S1-based ELISA Kit to detect MERS-CoV (IgG) antibodies and subsequently by MERS pseudotyped viral particles (MERSpp) neutralization assay for confirmation. Genetic diversity of the positive isolates was determined based on the amplification and sequencing of the spike gene. Our results showed high prevalence (38.6%) of MERS-CoV infection in slaughtered camels and high seropositivity (70.8%) during the time of the study. These data indicate previous and ongoing MERS-CoV infection in camels. Phylogenic analysis revealed relatively low genetic variability among our isolated samples. When these isolates were aligned against published spike sequences of MERS-CoV, deposited in global databases, there was sequence similarity of 94%. High seroprevalence and high genetic stability of MERS-CoV in camels indicating that camels pose a public health threat. The widespread MERS-CoV infections in camels might lead to a risk of future zoonotic transmission into people with direct contact with these infected camels. This study confirms re-infections in camels, highlighting a challenge for vaccine development when it comes to protective immunity.
Collapse
|
13
|
Almahboub SA, Algaissi A, Alfaleh MA, ElAssouli MZ, Hashem AM. Evaluation of Neutralizing Antibodies Against Highly Pathogenic Coronaviruses: A Detailed Protocol for a Rapid Evaluation of Neutralizing Antibodies Using Vesicular Stomatitis Virus Pseudovirus-Based Assay. Front Microbiol 2020; 11:2020. [PMID: 33013745 PMCID: PMC7498578 DOI: 10.3389/fmicb.2020.02020] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 07/30/2020] [Indexed: 12/13/2022] Open
Abstract
Emerging highly pathogenic human coronaviruses (CoVs) represent a serious ongoing threat to the public health worldwide. The spike (S) proteins of CoVs are surface glycoproteins that facilitate viral entry into host cells via attachment to their respective cellular receptors. The S protein is believed to be a major immunogenic component of CoVs and a target for neutralizing antibodies (nAbs) and most candidate vaccines. Development of a safe and convenient assay is thus urgently needed to determine the prevalence of CoVs nAbs in the population, to study immune response in infected individuals, and to aid in vaccines and viral entry inhibitor evaluation. While live virus-based neutralization assays are used as gold standard serological methods to detect and measure nAbs, handling of highly pathogenic live CoVs requires strict bio-containment conditions in biosafety level-3 (BSL-3) laboratories. On the other hand, use of replication-incompetent pseudoviruses bearing CoVs S proteins could represent a safe and useful method to detect nAbs in serum samples under biosafety level-2 (BSL-2) conditions. Here, we describe a detailed protocol of a safe and convenient assay to generate vesicular stomatitis virus (VSV)-based pseudoviruses to evaluate and measure nAbs against highly pathogenic CoVs. The protocol covers methods to produce VSV pseudovirus bearing the S protein of the Middle East respiratory syndrome-CoV (MERS-CoV) and the severe acute respiratory syndrome-CoV-2 (SARS-CoV-2), pseudovirus titration, and pseudovirus neutralization assay. Such assay could be adapted by different laboratories and researchers working on highly pathogenic CoVs without the need to handle live viruses in the BSL-3 environment.
Collapse
Affiliation(s)
- Sarah A. Almahboub
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdullah Algaissi
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
- Medical Research Center, Jazan University, Jazan, Saudi Arabia
| | - Mohamed A. Alfaleh
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - M-Zaki ElAssouli
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Anwar M. Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
14
|
An X, Martinez-Paniagua M, Rezvan A, Fathi M, Singh S, Biswas S, Pourpak M, Yee C, Liu X, Varadarajan N. Single-dose intranasal vaccination elicits systemic and mucosal immunity against SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 32743568 DOI: 10.1101/2020.07.23.212357] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A safe and durable vaccine is urgently needed to tackle the COVID19 pandemic that has infected >15 million people and caused >620,000 deaths worldwide. As with other respiratory pathogens, the nasal compartment is the first barrier that needs to be breached by the SARS-CoV-2 virus before dissemination to the lung. Despite progress at remarkable speed, current intramuscular vaccines are designed to elicit systemic immunity without conferring mucosal immunity. We report the development of an intranasal subunit vaccine that contains the trimeric or monomeric spike protein and liposomal STING agonist as adjuvant. This vaccine induces systemic neutralizing antibodies, mucosal IgA responses in the lung and nasal compartments, and T-cell responses in the lung of mice. Single-cell RNA-sequencing confirmed the concomitant activation of T and B cell responses in a germinal center-like manner within the nasal-associated lymphoid tissues (NALT), confirming its role as an inductive site that can lead to long-lasting immunity. The ability to elicit immunity in the respiratory tract has can prevent the initial establishment of infection in individuals and prevent disease transmission across humans.
Collapse
|