101
|
Handrejk K, Schmitz KS, Veldhuis Kroeze EJB, van Dijk LLA, van Run P, Haagmans B, Moscona A, Porotto M, de Swart RL, de Vries RD, Rissmann M. Characterization of a SARS-CoV-2 Omicron BA.5 direct-contact transmission model in hamsters. NPJ VIRUSES 2024; 2:52. [PMID: 39512864 PMCID: PMC11537969 DOI: 10.1038/s44298-024-00061-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/20/2024] [Indexed: 11/15/2024]
Abstract
As SARS-CoV-2 continues to evolve antigenically to escape vaccine- or infection-induced immunity, suitable animal models are needed to study novel interventions against viral variants. Syrian hamsters are often used because of their high susceptibility to SARS-CoV-2 and associated tissue damage in the respiratory tract. Here, we established a direct-contact transmission model for SARS-CoV-2 Omicron BA.5 in hamsters. First, we determined whether 103 or 104 TCID50 in a low-volume inoculum led to reproducible infection and viral shedding in male and female hamsters. Next, we determined the optimal co-housing timing and duration between donor and recipient hamsters required for consistent direct-contact transmission. Finally, we compared viral loads and histopathological lesions in the respiratory tissues of donor and recipient hamsters. Intranasal inoculation of hamsters with 103 TCID50 and 104 TCID50 Omicron BA.5 in 10 µl per nostril led to reproducible infection. Viral loads in the throat measured by RT-qPCR were comparable between male and female hamsters. Notably, the shedding of infectious virus was significantly higher in male hamsters. Compared to SARS-CoV-2 D614G, Omicron BA.5 infection reached lower viral loads, had a delayed peak of virus replication, and induced limited body weight loss. To ensure consistent direct-contact transmission from inoculated donor hamsters to naïve recipients, a co-housing duration of 24 h starting 20 h post-infection of the donors was optimal. We detected mild inflammation in the respiratory tract of donor and recipient hamsters, and viral loads were higher and peaked earlier in donor hamsters compared to recipient hamsters. Taken together, we developed a robust Omicron BA.5 direct-contact transmission model in hamsters, that provides a valuable tool to study novel interventions.
Collapse
Affiliation(s)
- Kim Handrejk
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | | | | | | | - Peter van Run
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Bart Haagmans
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Anne Moscona
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY USA
- Center for Host–Pathogen Interaction, Columbia University Vagelos College of Physicians and Surgeons, New York, NY USA
- Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY USA
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY USA
| | - Matteo Porotto
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY USA
- Center for Host–Pathogen Interaction, Columbia University Vagelos College of Physicians and Surgeons, New York, NY USA
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Rik L. de Swart
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Rory D. de Vries
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Melanie Rissmann
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| |
Collapse
|
102
|
Zhu J, Xing F, Li Y, Wu C, Li S, Wang Q, Huang J, Zhang Y, Zheng X, Liu Z, Rao J, Hong R, Tian S, Xiong S, Tan L, Chen X, Li Y, He W, Hong X, Xia J, Zhou Q, Zhang Z. Exploring the causes of variability in quality of oropharyngeal swab sampling for SARS-CoV-2 nucleic acid testing and proposed improvement measures: a multicenter, double-blind study. Microbiol Spectr 2024; 12:e0156724. [PMID: 39382280 PMCID: PMC11537049 DOI: 10.1128/spectrum.01567-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 09/09/2024] [Indexed: 10/10/2024] Open
Abstract
Although coronavirus disease 2019 (COVID-19) has not been considered a public health emergency of international concern since last year, intermittent regional impacts still persist, and accurate testing remains crucial. Ribonuclease P protein subunit P30 (RPP30) RNA, known for its broad and stable expression in tissue cells, was used to evaluate samples from 10 hospitals with over 3,000 negative nucleic acid tests. The results revealed that the overall pass rate for the collected samples was consistently low and exhibited significant heterogeneity. After reassessing the evaluative effectiveness of RPP30 RNA Ct values from the samples of 132 positive individuals under quarantine observation, it was used to identify factors affecting sampling quality. These factors included different stages ranging from sample collection to PCR processing, various characteristics of both samplers and individuals being sampled, as well as sampling season and location. The results indicated that post-sampling handling had minimal impact, winter and fever clinic samples showed higher quality, whereas children's samples had lower quality. The key finding was that the characteristics of samplers were closely related to sampling quality, emphasizing the role of subjectivity. Quality control warnings led to substantial improvements, confirming this finding. Consequently, although there are various factors during the testing process, the most critical aspect is to improve, supervise, and maintain standardized practices among sampling staff.IMPORTANCEThis study further confirmed the reliability of internal references (IRs) in assessing sample quality, and utilized a large sample IR data to comprehensively and multidimensionally identify significant interference factors in nucleic acid test results. By further reminding and intervening in the subjective practices of specimen collectors, good results could be achieved.
Collapse
Affiliation(s)
- Jie Zhu
- Institute of Clinical Virology, Department of Infectious Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fanfan Xing
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yunzhu Li
- Department of Infectious Diseases, The Third Affiliated Hospital of Anhui Medical University, The First People's Hospital of Hefei, Hefei, China
| | - Chunchen Wu
- Department of Laboratory Medicine, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shasha Li
- Division of Liver Disease, The Second People's Hospital of Fuyang City, Fuyang, China
| | - Qin Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jinyue Huang
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yafei Zhang
- Institute of Clinical Virology, Department of Infectious Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaowei Zheng
- Department of Infectious Diseases, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Zhenjun Liu
- Department of Infectious Diseases, Anqing Municipal Hospital, Anqing, China
| | - Jianguo Rao
- Department of Infectious Diseases, The Lu'an Affiliated Hospital of Anhui Medical University, Lu'an People's Hospital, Lu'an, China
| | - Rui Hong
- Department of Infectious Diseases, Tongling Municipal Hospital, Tongling, China
| | - Shuilin Tian
- Division of Liver Disease, Traditional Chinese Hospital of LuAn, Anhui University of Traditional Chinese Medicine, Lu'an, China
| | - Shuangyun Xiong
- Department of Infectious Diseases, Funan County People's Hospital, Fuyang, China
| | - Lin Tan
- Division of Liver Disease, The Second People's Hospital of Fuyang City, Fuyang, China
| | - Xinlei Chen
- Institute of Clinical Virology, Department of Infectious Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yanwu Li
- Institute of Clinical Virology, Department of Infectious Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei He
- Division of Liver Disease, Traditional Chinese Hospital of LuAn, Anhui University of Traditional Chinese Medicine, Lu'an, China
| | - Xiaodan Hong
- Institute of Clinical Virology, Department of Infectious Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jianbo Xia
- Department of Laboratory Medicine, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiang Zhou
- Department of Clinical Laboratory, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhenhua Zhang
- Institute of Clinical Virology, Department of Infectious Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
103
|
da Silva LL, Leon LAA, da Cruz Moreira O, da Costa Nunes Pimentel Coelho WL, da Costa VD, Ivantes CAP, Pollo-Flores P, Lewis-Ximenez LL, de Paula VS, Villar LM. Serum microRNA 143 and 223 Gene Expression Profiles as Potential Biomarkers in Individuals with Hepatitis and COVID-19. Viruses 2024; 16:1734. [PMID: 39599849 PMCID: PMC11598994 DOI: 10.3390/v16111734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
MicroRNAs (miRNAs) can act as biomarkers and descriptors of the association between infections and other diseases, such as hepatitis and COVID-19. This study aims to investigate the role of miRNA serum expression according to laboratory data concerning hepatitis and COVID-19. Seventy individuals recruited in Southern and Southeastern Brazil donated serum samples and were divided into four groups: (i) 20 negative subjects, (ii) 20 presenting hepatitis, (iii) 19 with COVID-19 and (iv) 11 with hepatitis and COVID-19. Three miRNAs (miR-122, miR-143 and miR-223) were evaluated using real-time PCR. Hematological and biochemical markers were also analyzed. MiR-143 and miR-223 were downregulated among the hepatitis/COVID-19 group (p < 0.05). A positive correlation was observed between miR-223 and lymphocytes. There was a negative correlation between alanine transaminase (ALT) and aspartate transaminase (AST) for miR-143 and miR-223 and gamma-glutamyl transferase (GGT), alkaline phosphatase (AP) and neutrophil/lymphocyte ratio (NLR) only for miR-223 (p < 0.05). For hepatic fibrosis (FIB-4), miR-122 and miR-143 had a greater association and miR-223 was more associated with a history of vaccination against COVID-19. MicroRNAs 143 and 223 could be useful as biomarkers for hepatitis coinfection with COVID-19.
Collapse
Affiliation(s)
- Lucas Lima da Silva
- National Reference Laboratory for Viral Hepatitis, Institute Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, RJ, Brazil; (V.D.d.C.); (L.L.L.-X.)
| | - Luciane Almeida Amado Leon
- Technological Development Laboratory, Institute Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, RJ, Brazil; (L.A.A.L.); (W.L.d.C.N.P.C.)
| | - Otacílio da Cruz Moreira
- Molecular Virology and Parasitology Laboratory, Institute Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, RJ, Brazil; (O.d.C.M.); (V.S.d.P.)
| | | | - Vanessa Duarte da Costa
- National Reference Laboratory for Viral Hepatitis, Institute Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, RJ, Brazil; (V.D.d.C.); (L.L.L.-X.)
| | | | - Priscila Pollo-Flores
- Department of Clinical Medicine, Fluminense Federal University, Niterói 24220-000, RJ, Brazil;
| | - Lia Laura Lewis-Ximenez
- National Reference Laboratory for Viral Hepatitis, Institute Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, RJ, Brazil; (V.D.d.C.); (L.L.L.-X.)
| | - Vanessa Salete de Paula
- Molecular Virology and Parasitology Laboratory, Institute Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, RJ, Brazil; (O.d.C.M.); (V.S.d.P.)
| | - Livia Melo Villar
- National Reference Laboratory for Viral Hepatitis, Institute Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, RJ, Brazil; (V.D.d.C.); (L.L.L.-X.)
| |
Collapse
|
104
|
Polat SE, Arıkan FM, Gençoğlu MY, Akyan ŞS, Uytun S, Tabakçı SÖ, Bilgiç I, Çakar MK, Ademhan Tural D, Tuğcu GD, Cinel G. Experience with flexible bronchoscopy for noncoronavirus disease of 2019 indications in pediatric patients during the coronavirus disease of 2019 pandemic. Pediatr Pulmonol 2024; 59:2867-2874. [PMID: 38896067 DOI: 10.1002/ppul.27144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/06/2024] [Accepted: 06/11/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND AND AIM Flexible bronchoscopy (FB) poses a risk of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) transmission due to aerosol generation. This study aimed to assess the utilization, indications, outcomes, and safety of FB in pediatric patients for noncoronavirus disease of 2019 (COVID-19) reasons during the pandemic. MATERIALS AND METHODS We retrospectively analyzed pediatric patients who underwent FB for non-COVID-19 indications at a tertiary children's hospital's pulmonary clinic during the COVID-19 pandemic. Patients showed no COVID-19 symptoms and tested negative for SARS-CoV-2 by real-time polymerase chain reaction (PCR) of nasopharyngeal and throat swabs within 24 h before the procedure. FBs were conducted in the operating room, with healthcare professionals (HCPs) wearing personal protective equipment, including medical N95 masks, gloves, gowns, and eye protection. RESULTS Between March 2020 and April 2022, 167 pediatric patients underwent FB for non-COVID-19 indications. Common indications included foreign body aspiration (22.7%), stridor (10.1%), and atelectasis (8.9%). No COVID-19 symptoms were observed in patients on the 1st and 10th days post-FB. During the 1-month follow-up, 52 patients underwent SARSCoV-2 PCR testing, and one patient tested positive in the third week after the procedure. None of the HCPs in the FB team experienced COVID-19 symptoms or tested positive for SARS-CoV-2. CONCLUSION A bronchoscopy protocol with safety precautions minimized the risk of COVID-19 transmission, allowing safe FB performance for non-COVID-19 indications in pediatric patients during the pandemic. The experience gained in FB during COVID-19 is valuable for similar situations in the future.
Collapse
Affiliation(s)
| | | | | | - Şule Selin Akyan
- Division of Pediatric Pulmonology, Ankara City Hospital, Ankara, Turkey
| | - Salih Uytun
- Division of Pediatric Pulmonology, Ankara City Hospital, Ankara, Turkey
| | | | - Işıl Bilgiç
- Division of Pediatric Pulmonology, Ankara City Hospital, Ankara, Turkey
| | | | | | - Gökçen D Tuğcu
- Division of Pediatric Pulmonology, Ankara City Hospital, Ankara, Turkey
| | - Güzin Cinel
- Division of Pediatric Pulmonology, Ankara City Hospital, Ankara, Turkey
- Division of Pediatric Pulmonology, Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| |
Collapse
|
105
|
Yang Y, Tan J, Wang F, Sun W, Shi H, Cheng Z, Xie Y, Zhou X. Preconcentration and detection of SARS-CoV-2 in wastewater: A comprehensive review. Biosens Bioelectron 2024; 263:116617. [PMID: 39094290 DOI: 10.1016/j.bios.2024.116617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/17/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
Severe acute respiratory syndrome coronaviruses 2 (SARS-CoV-2) causing coronavirus disease 2019 (COVID-19) affected the health of human beings and the global economy. The patients with SARS-CoV-2 infection had viral RNA or live infectious viruses in feces. Thus, the possible transmission of SARS-CoV-2 through wastewater received great attentions. Moreover, SARS-CoV-2 in wastewater can serve as an early indicator of the infection within communities. We summarized the preconcentration and detection technology of SARS-CoV-2 in wastewater aiming at the complex matrices of wastewater and low virus concentration and compared their performance characteristics. We described the emerging tests that would be possible to realize the rapid detection of SARS-CoV-2 in fields and encourage academics to advance their technologies beyond conception. We concluded with a brief discussion on the outlook for integrating preconcentration and the detection of SARS-CoV-2 with emerging technologies.
Collapse
Affiliation(s)
- Yihan Yang
- School of Environment, Tsinghua University, Beijing, 100084, China
| | - Jisui Tan
- School of Environment, Tsinghua University, Beijing, 100084, China
| | - Fan Wang
- School of Environment, Tsinghua University, Beijing, 100084, China
| | - Weiming Sun
- School of Environment, Tsinghua University, Beijing, 100084, China
| | - Hanchang Shi
- School of Environment, Tsinghua University, Beijing, 100084, China
| | - Zhao Cheng
- School of Environment, Tsinghua University, Beijing, 100084, China
| | - Yangcun Xie
- Chinese Academy of Environmental Planning, Beijing, 100043, China.
| | - Xiaohong Zhou
- School of Environment, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
106
|
Düker U, Nogueira R, Carpio-Vallejo E, Joost I, Hüppe K, Suchenwirth R, Saathoff Y, Wallner M. Sewer system sampling for wastewater-based disease surveillance: Is the work worth it? JOURNAL OF WATER AND HEALTH 2024; 22:2218-2232. [PMID: 39611680 DOI: 10.2166/wh.2024.301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/11/2024] [Indexed: 11/30/2024]
Abstract
Wastewater treatment plant (WWTP) influent sampling is commonly used in wastewater-based disease surveillance to assess the circulation of pathogens in the population aggregated in a catchment area. However, the signal can be lost within the sewer network due to adsorption, degradation, and dilution processes. The present work aimed to investigate the dynamics of SARS-CoV-2 concentration in three sub-catchments of the sewer system in the city of Hildesheim, Germany, characterised by different levels of urbanisation and presence/absence of industry, and to evaluate the benefit of sub-catchment sampling compared to WWTP influent sampling. Our study shows that sampling and analysis of virus concentrations in sub-catchments with particular settlement structures allows the identification of high concentrations of the virus at a local level in the wastewater, which are lower in samples collected at the inlet of the treatment plant covering the whole catchment. Higher virus concentrations per inhabitant were found in the sub-catchments in comparison to the inlet of the WWTP. Additionally, sewer sampling provides spatially resolved concentrations of SARS-CoV-2 in the catchment area, which is important for detecting local high incidences of COVID-19.
Collapse
Affiliation(s)
- Urda Düker
- Leibniz University Hannover, Welfengarten 1, 30459 Hannover, Germany
| | - Regina Nogueira
- Leibniz University Hannover, Welfengarten 1, 30459 Hannover, Germany
| | | | - Ingeborg Joost
- Ostfalia University of Applied Sciences, Campus Suderburg, Herbert-Meyer-Str. 7, 29556 Suderburg, Germany
| | - Katharina Hüppe
- Local Health Authority Hildesheim, Ludolfingerstr. 2, 31137 Hildesheim, Germany
| | - Roland Suchenwirth
- Public Health Agency of Lower Saxony, Roesebeckstr. 4-6, 30449 Hannover, Germany
| | - Yvonne Saathoff
- Public Health Agency of Lower Saxony, Roesebeckstr. 4-6, 30449 Hannover, Germany
| | - Markus Wallner
- Ostfalia University of Applied Sciences, Campus Suderburg, Herbert-Meyer-Str. 7, 29556 Suderburg, Germany E-mail:
| |
Collapse
|
107
|
Hall GJ, Page EJ, Rhee M, Hay C, Krause A, Langenbacher E, Ruth A, Grenier S, Duran AP, Kamara I, Iskander JK, Alsayyid F, Thomas DL, Bock E, Porta N, Pharo J, Osterink BA, Zelmanowitz S, Fleischmann CM, Liyanage D, Gray JP. Wastewater Surveillance of US Coast Guard Installations and Seagoing Military Vessels to Mitigate the Risk of COVID-19 Outbreaks, March 2021-August 2022. Public Health Rep 2024; 139:699-707. [PMID: 38561999 PMCID: PMC11504356 DOI: 10.1177/00333549241236644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024] Open
Abstract
OBJECTIVES Military training centers and seagoing vessels are often environments at high risk for the spread of COVID-19 and other contagious diseases, because military trainees and personnel arrive after traveling from many parts of the country and live in congregate settings. We examined whether levels of SARS-CoV-2 genetic material in wastewater correlated with SARS-CoV-2 infections among military personnel living in communal barracks and vessels at US Coast Guard training centers in the United States. METHODS The Coast Guard developed and established 3 laboratories with wastewater testing capability at Coast Guard training centers from March 2021 through August 2022. We analyzed wastewater from barracks housing trainees and from 4 Coast Guard vessels for the presence of SARS-CoV-2 genes N and E and quantified the results relative to levels of a fecal indicator virus, pepper mild mottle virus. We compared quantified data with the timing of medically diagnosed COVID-19 infection among (1) military personnel who had presented with symptoms or had been discovered through contact tracing and had medical tests and (2) military personnel who had been discovered through routine surveillance by positive SARS-CoV-2 antigen or polymerase chain reaction test results. RESULTS Levels of viral genes in wastewater at Coast Guard locations were best correlated with diagnosed COVID-19 cases when wastewater testing was performed twice weekly with passive samplers deployed for the entire week; such testing detected ≥1 COVID-19 case 69.8% of the time and ≥3 cases 88.3% of the time. Wastewater assessment in vessels did not continue because of logistical constraints. CONCLUSION Wastewater testing is an effective tool for measuring the presence and patterns of SARS-CoV-2 infections among military populations. Success with wastewater testing for SARS-CoV-2 infections suggests that other diseases may be assessed with similar approaches.
Collapse
Affiliation(s)
- Gregory J. Hall
- Department of Chemical and Environmental Sciences, US Coast Guard Academy, New London, CT, USA
| | - Eric J. Page
- Department of Physics, US Coast Guard Academy, New London, CT, USA
| | - Min Rhee
- Department of Chemical and Environmental Sciences, US Coast Guard Academy, New London, CT, USA
| | - Clara Hay
- Department of Chemical and Environmental Sciences, US Coast Guard Academy, New London, CT, USA
| | - Amelia Krause
- Department of Chemical and Environmental Sciences, US Coast Guard Academy, New London, CT, USA
| | - Emma Langenbacher
- Department of Chemical and Environmental Sciences, US Coast Guard Academy, New London, CT, USA
| | - Allison Ruth
- Department of Chemical and Environmental Sciences, US Coast Guard Academy, New London, CT, USA
| | - Steve Grenier
- Department of Civil and Environmental Engineering, US Coast Guard Academy, New London, CT, USA
| | - Alexander P. Duran
- Office of Environmental Safety, US Coast Guard Academy, New London, CT, USA
| | - Ibrahim Kamara
- Occupational Medicine and Quality Improvement Division, US Coast Guard Headquarters, Washington, DC, USA
| | - John K. Iskander
- Preventive Medicine and Population Health, US Coast Guard Headquarters, Washington, DC, USA
| | - Fahad Alsayyid
- Coast Guard Medical Directorate, US Coast Guard, Cape May, NJ, USA
| | - Dana L. Thomas
- Health, Safety, and Work-Life Service Center, US Coast Guard Headquarters, Washington, DC, USA
| | - Edward Bock
- Health, Safety, and Work-Life Service Center, US Coast Guard, Norfolk, VA, USA
| | - Nicholas Porta
- Health, Safety, and Work-Life Service Center, US Coast Guard, Norfolk, VA, USA
| | - Jessica Pharo
- Health, Safety, and Work-Life Service Center, US Coast Guard, Norfolk, VA, USA
| | - Beth A. Osterink
- Health, Safety, and Work-Life Service Center, US Coast Guard, Norfolk, VA, USA
| | - Sharon Zelmanowitz
- Department of Civil and Environmental Engineering, US Coast Guard Academy, New London, CT, USA
| | - Corinna M. Fleischmann
- Department of Civil and Environmental Engineering, US Coast Guard Academy, New London, CT, USA
| | - Dilhara Liyanage
- Department of Chemical and Environmental Sciences, US Coast Guard Academy, New London, CT, USA
| | - Joshua P. Gray
- Department of Chemical and Environmental Sciences, US Coast Guard Academy, New London, CT, USA
| |
Collapse
|
108
|
Dang H, Joung Y, Yang JY, Lee SH, Lee S, Joo SW, Park SG, Choo J. Advancing COVID-19 Diagnosis: Enhancement in SERS-PCR with 30-nm Au Nanoparticle-Internalized Nanodimpled Substrates. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403672. [PMID: 38970560 DOI: 10.1002/smll.202403672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/16/2024] [Indexed: 07/08/2024]
Abstract
Real-time polymerase chain reaction (RT-PCR) with fluorescence detection is the gold standard for diagnosing coronavirus disease 2019 (COVID-19) However, the fluorescence detection in RT-PCR requires multiple amplification steps when the initial deoxyribonucleic acid (DNA) concentration is low. Therefore, this study has developed a highly sensitive surface-enhanced Raman scattering-based PCR (SERS-PCR) assay platform using the gold nanoparticle (AuNP)-internalized gold nanodimpled substrate (AuNDS) plasmonic platform. By comparing different sizes of AuNPs, it is observed that using 30 nm AuNPs improves the detection limit by approximately ten times compared to 70 nm AuNPs. Finite-difference time-domain (FDTD) simulations show that multiple hotspots are formed between AuNPs and the cavity surface and between AuNPs when 30 nm AuNPs are internalized in the cavity, generating a strong electric field. With this 30 nm AuNPs-AuNDS SERS platform, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ribonucleic acid (RNA)-dependent RNA polymerase (RdRp) can be detected in only six amplification cycles, significantly improving over the 25 cycles required for RT-PCR. These findings pave the way for an amplification-free molecular diagnostic system based on SERS.
Collapse
Affiliation(s)
- Hajun Dang
- Department of Chemistry, Chung-Ang University, Seoul, 06974, South Korea
| | - Younju Joung
- Department of Chemistry, Chung-Ang University, Seoul, 06974, South Korea
| | - Jun-Yeong Yang
- Nano-Bio Convergence Department, Korea Institute of Materials Science (KIMS), Changwon, 51508, South Korea
| | - Soo Hyun Lee
- Nano-Bio Convergence Department, Korea Institute of Materials Science (KIMS), Changwon, 51508, South Korea
| | - Seunghun Lee
- Nano-Bio Convergence Department, Korea Institute of Materials Science (KIMS), Changwon, 51508, South Korea
| | - Sang-Woo Joo
- Department of Chemistry, Soongsil University, Seoul, 06978, South Korea
| | - Sung-Gyu Park
- Nano-Bio Convergence Department, Korea Institute of Materials Science (KIMS), Changwon, 51508, South Korea
| | - Jaebum Choo
- Department of Chemistry, Chung-Ang University, Seoul, 06974, South Korea
| |
Collapse
|
109
|
Kumar P, Zhang X, Shaha R, Kschischo M, Dobbelstein M. Identification of antibody-resistant SARS-CoV-2 mutants via N4-Hydroxycytidine mutagenesis. Antiviral Res 2024; 231:106006. [PMID: 39293594 DOI: 10.1016/j.antiviral.2024.106006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 08/31/2024] [Accepted: 09/12/2024] [Indexed: 09/20/2024]
Abstract
Monoclonal antibodies targeting the Spike protein of SARS-CoV-2 are effective against COVID-19 and might mitigate future pandemics. However, their efficacy is challenged by the emergence of antibody-resistant virus variants. We developed a method to efficiently identify such resistant mutants based on selection from mutagenized virus pools. By inducing mutations with the active compound of Molnupiravir, N4-hydroxycytidine (NHC), and subsequently passaging the virus in the presence of antibodies, we identified specific Spike mutations linked to resistance. Validation of these mutations was conducted using pseudotypes and immunofluorescence analysis. From a Wuhan-like strain of SARS-CoV-2, we identified the following mutations conferring strong resistance towards the corresponding antibodies: Bamlanivimab - E484K, F490S and S494P; Sotrovimab - E340K; Cilgavimab - K444R/E and N450D. From the Omicron B.1.1.529 variant, the strongly selected mutations were: Bebtelovimab - V445A; Sotrovimab - E340K and K356M; Cilgavimab - K444R, V445A and N450D. We also identified escape mutations in the Wuhan-like Spike for the broadly neutralizing antibodies S2K146 - combined G485S and Q493R - and S2H97 - D428G, K462E and S514F. Structural analysis revealed that the selected mutations occurred at antibody-binding residues within the receptor-binding domains of the Spike protein. Most of the selected mutants largely maintained ACE2 binding and infectivity. Notably, many of the identified resistance-conferring mutations are prevalent in real-world SARS-CoV-2 variants, but some of them (G485S, D428G, and K462E) have not yet been observed in circulating strains. Our approach offers a strategy for predicting the therapeutic efficacy of antibodies against emerging virus variants.
Collapse
MESH Headings
- SARS-CoV-2/genetics
- SARS-CoV-2/immunology
- SARS-CoV-2/drug effects
- Cytidine/analogs & derivatives
- Cytidine/pharmacology
- Cytidine/genetics
- Humans
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Drug Resistance, Viral/genetics
- Mutation
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- Mutagenesis
- COVID-19/virology
- COVID-19/immunology
- Antiviral Agents/pharmacology
- COVID-19 Drug Treatment
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/pharmacology
- Hydroxylamines
Collapse
Affiliation(s)
- Priya Kumar
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, 37077, Göttingen, Germany
| | - Xiaoxiao Zhang
- Department of Mathematics and Technology, University of Applied Sciences Koblenz, 53424, Remagen, Germany; Department of Informatics, Technical University of Munich, 81675, Munich, Germany
| | - Rahul Shaha
- Department of Molecular Enzymology, Göttingen Center of Molecular Biosciences (GZMB), University of Göttingen, 37077, Göttingen, Germany
| | - Maik Kschischo
- Department of Mathematics and Technology, University of Applied Sciences Koblenz, 53424, Remagen, Germany
| | - Matthias Dobbelstein
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, 37077, Göttingen, Germany; Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077 Göttingen, Germany.
| |
Collapse
|
110
|
Oscorbin IP, Novikova LM, Khrapov EA, Filipenko ML. PI primers increase the efficacy of LAMP and RT-LAMP for SARS-CoV-2 and MS2 phage detection. Diagn Microbiol Infect Dis 2024; 110:116449. [PMID: 39133998 DOI: 10.1016/j.diagmicrobio.2024.116449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 06/15/2024] [Accepted: 07/15/2024] [Indexed: 09/04/2024]
Abstract
LAMP (Loop-mediated isothermal amplification) is a popular method for the molecular diagnostics of numerous pathogens, specifically useful for point-of-care testing. However, the efficacy and sensitivity of LAMP still need to be maximised for the best performance in clinical settings. Adding a novel fourth primer pair is a promising way to accelerate the LAMP speed. Here, we report PI primers that are part of inner primers and can be used in LAMP without a specific design. PI primers were tested in quantitative LAMP detecting SARS-CoV-2 and MS2. The new primers have increased the speed and sensitivity of quantitative LAMP, RT-LAMP, and duplex LAMP with artificial templates and RNA samples from nasal swabs. Adding PI primers could become a valuable option for LAMP optimisation, especially when a desirable LAMP target is a highly variable DNA sequence with a few conservative sites for primers.
Collapse
Affiliation(s)
- Igor P Oscorbin
- Laboratory of Pharmacogenomics, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 8 Lavrentiev Avenue, 630090 Novosibirsk, Russia.
| | - Lidiya M Novikova
- Laboratory of Pharmacogenomics, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 8 Lavrentiev Avenue, 630090 Novosibirsk, Russia
| | - Evgeniy A Khrapov
- Laboratory of Pharmacogenomics, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 8 Lavrentiev Avenue, 630090 Novosibirsk, Russia
| | - Maxim L Filipenko
- Laboratory of Pharmacogenomics, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 8 Lavrentiev Avenue, 630090 Novosibirsk, Russia
| |
Collapse
|
111
|
Staniszewski F, Schilder A, Osinkina L, Westenthanner M, Kataeva N, Posch B, Gillitschka Y, Stoecker K, Silberreis K, Coen S, Cannas A, Matusali G, Schmidleithner C, Stehr J, Buersgens F, Peham JR. Rapid detection of SARS-CoV-2 with a mobile device based on pulse controlled amplification. Biosens Bioelectron 2024; 263:116626. [PMID: 39116633 DOI: 10.1016/j.bios.2024.116626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024]
Abstract
In the past, vast research has been conducted on biosensors and point-of-care (PoC) diagnostics. Despite rapid advances especially during the SARS-CoV-2 pandemic in this research field a low-cost molecular biosensor exhibiting the user-friendliness of a rapid antigen test, and also the sensitivity and specificity of a PCR test, has not been developed yet. To this end we developed a novel microfluidics based and handheld PoC device, that facilitates viral detection at PCR sensitivity and specificity in less than 40 min, including 15 min sample preparation. This was attained by incorporation of pulse controlled amplification (PCA), a method which uses short electrical pulses to rapidly increase the temperature of a small fraction of the sample volume. In this work, we present a low-cost PCA device with a microfluidic consumable intended for the use in a decentralized or home-setting. We used finite element analysis (FEA) simulations to display the fundamental principle and highlight the critical parameter dependency of PCA, such as pulse length and resistor shape. Furthermore, we integrated a simple and fast workflow for sample preparation and evaluated the limit of detection (LoD) for SARS-CoV-2 viral RNA, which is 0.88 copies/μL (=44 copies/reaction), and thus, comparable to conventional RT-qPCR. Additionally, target specificity of the device was validated. Our device and PCA approach enables cost-effective, rapid and mobile molecular diagnostics while remaining highly sensitive and specific.
Collapse
Affiliation(s)
- Filip Staniszewski
- AIT Austrian Institute of Technology, Center for Health and Bioresources, Competence Unit Molecular Diagnostics, Giefinggasse 4, 1210, Vienna, Austria
| | - Alexandra Schilder
- AIT Austrian Institute of Technology, Center for Health and Bioresources, Competence Unit Molecular Diagnostics, Giefinggasse 4, 1210, Vienna, Austria
| | - Lidiya Osinkina
- HP Health Solutions Germany GmbH, Am Klopferspitz 19, 82152, Planegg, Germany
| | | | - Nadezhda Kataeva
- AIT Austrian Institute of Technology, Center for Health and Bioresources, Competence Unit Molecular Diagnostics, Giefinggasse 4, 1210, Vienna, Austria
| | - Barbara Posch
- AIT Austrian Institute of Technology, Center for Health and Bioresources, Competence Unit Molecular Diagnostics, Giefinggasse 4, 1210, Vienna, Austria
| | - Yasmin Gillitschka
- AIT Austrian Institute of Technology, Center for Health and Bioresources, Competence Unit Molecular Diagnostics, Giefinggasse 4, 1210, Vienna, Austria
| | - Kilian Stoecker
- Bundeswehr Institute of Microbiology, Neuherbergstr. 11, 80937, Munich, Germany
| | - Kim Silberreis
- Bundeswehr Institute of Microbiology, Neuherbergstr. 11, 80937, Munich, Germany
| | - Sabrina Coen
- Laboratory of Virology, National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, Via Portuense 292, 00149, Rome, Italy
| | - Angela Cannas
- Laboratory of Microbiology, National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, Via Portuense 292, 00149, Rome, Italy
| | - Giulia Matusali
- Laboratory of Virology, National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, Via Portuense 292, 00149, Rome, Italy
| | - Christina Schmidleithner
- AIT Austrian Institute of Technology, Center for Health and Bioresources, Competence Unit Molecular Diagnostics, Giefinggasse 4, 1210, Vienna, Austria
| | - Joachim Stehr
- HP Health Solutions Germany GmbH, Am Klopferspitz 19, 82152, Planegg, Germany
| | - Federico Buersgens
- HP Health Solutions Germany GmbH, Am Klopferspitz 19, 82152, Planegg, Germany
| | - Johannes R Peham
- AIT Austrian Institute of Technology, Center for Health and Bioresources, Competence Unit Molecular Diagnostics, Giefinggasse 4, 1210, Vienna, Austria.
| |
Collapse
|
112
|
Roth A, Gehre L, Gerke J, Lutz M, Manafa G, Schmitz T, Lambio C, Zhuang S, Butler J, Lakes T, Savaskan N. Epidemiology of COVID-19 in Berlin-Neukölln nursing homes. J Infect Public Health 2024; 17:102546. [PMID: 39321605 DOI: 10.1016/j.jiph.2024.102546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/10/2024] [Accepted: 09/15/2024] [Indexed: 09/27/2024] Open
Abstract
BACKGROUND The COVID-19 pandemic has affected various urban population groups in different ways. Earlier studies have shown that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) disproportionally impacts nursing home residents by increasing morbidity and mortality following viral exposure. However, little is known about the epidemiology of this disease in detail. Therefore, the objective of this study is to analyze the development of the COVID-19 pandemic in 14 nursing homes across Berlin-Neukölln, Germany, during pandemic waves 1 to 5 (Feb 2020 - May 2022). METHODS Reporting data to the Neukölln Department of Public Health on COVID-19 cases in connection with nursing homes were extracted from the SORMAS database. The case fatality rates (CFRs) and odds ratios (ORs) of demographic parameters, prevalent variants of concern (VOCs) and vaccine availability were calculated. In addition, the temporal course in waves 1-5 in Neukölln and the relevant government measures were examined. RESULTS Data collected from nursing homes providing age-dependent physical care revealed that 1.9 % of the total 108,600 cases registered in Berlin-Neukölln during the study period were related one of the 14 facilities. Compared to the general population in Neukölln, nursing homes exhibited a 20-fold increase in the CFR. Notably, nursing homes with higher bed capacities displayed a greater CFR than did smaller nursing homes. Similarly, elderly residents living in nursing homes faced a much greater mortality rate than did their counterparts living outside of medical settings (OR = 3.5). The original wild-type SARS-CoV-2 strain had the most severe direct impact, with a CFR of 16.7 %, compared to the alpha (CFR = 6.9 %), delta (CFR = 10.2 %) and omicron (CFR = 2.8 %) variants in nursing homes. Interestingly, the number of infections increased following vaccination campaigns, but this trend was accompanied by a decrease in the number of deaths from 2.6 to 1.1 per week. As a result, the CFR significantly decreased from 18.4 to 5.5, while still exceeding the mean CFR compared to that of the general population of Neukölln. CONCLUSIONS Our findings reveal the changing patterns of outbreak frequency and severity across the five pandemic waves. They highlight the crucial role of rapid vaccination programs for residents, staff, visitors, and third-party services in safeguarding nursing homes. Additionally, improvements in containment and cluster strategies are essential in prevaccination scenarios to prevent future infection traps for elderly individuals in long-term care facilities. The presented data highlight the importance of tailored protection measures for one of the most vulnerable populations in our society.
Collapse
Affiliation(s)
- Alexandra Roth
- Public Health Authority Neukölln, District Office Neukölln of Berlin, Blaschkoallee 32, 12359 Berlin, Germany; Institut für Gesundheitswissenschaften, FB Hebammenwissenschaft, University of Lübeck, Lübeck, Germany.
| | - Lena Gehre
- Public Health Authority Neukölln, District Office Neukölln of Berlin, Blaschkoallee 32, 12359 Berlin, Germany
| | - Jonas Gerke
- Public Health Authority Neukölln, District Office Neukölln of Berlin, Blaschkoallee 32, 12359 Berlin, Germany
| | - Maja Lutz
- Public Health Authority Neukölln, District Office Neukölln of Berlin, Blaschkoallee 32, 12359 Berlin, Germany
| | - Georgianna Manafa
- Public Health Authority Neukölln, District Office Neukölln of Berlin, Blaschkoallee 32, 12359 Berlin, Germany; Geography Department, Humboldt-Universität zu Berlin, 10099 Berlin, Germany
| | - Tillman Schmitz
- Geography Department, Humboldt-Universität zu Berlin, 10099 Berlin, Germany
| | - Christoph Lambio
- Geography Department, Humboldt-Universität zu Berlin, 10099 Berlin, Germany
| | - Sida Zhuang
- Geography Department, Humboldt-Universität zu Berlin, 10099 Berlin, Germany
| | - Jeffrey Butler
- Geography Department, Humboldt-Universität zu Berlin, 10099 Berlin, Germany
| | - Tobia Lakes
- Geography Department, Humboldt-Universität zu Berlin, 10099 Berlin, Germany
| | - Nicolai Savaskan
- Public Health Authority Neukölln, District Office Neukölln of Berlin, Blaschkoallee 32, 12359 Berlin, Germany.
| |
Collapse
|
113
|
Takata M, Nakamoto M, Kitaura T, Okada K, Endou H, Ma'arif AS, Nishikawa Y, Mukuda K, Morishita S, Murota H, Yamasaki A, Kageyama S, Burioka N, Chikumi H. An Optimal Transport Medium for SARS-CoV-2 Detection in the Direct Method of Rapid Microfluidic PCR System. Yonago Acta Med 2024; 67:293-302. [PMID: 39583763 PMCID: PMC11584235 DOI: 10.33160/yam.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/30/2024] [Indexed: 11/26/2024]
Abstract
Background Recently developed rapid real-time reverse transcription PCR (RT-PCR) systems adopting microfluidic thermal cycling technology are ideal for point-of-care (POC) testing for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Because the RNA extraction step before real-time RT-PCR is rate-limiting, a direct RNA extraction method (direct method) that adopts chemical viral lysis and eliminates RNA purification steps is preferable for rapid real-time RT-PCR. In the direct method, selecting the transport medium is essential because it may be introduced into subsequent real-time RT-PCR steps, but might inhibit PCR. However, the influence of transport medium on the combination of the direct method and rapid real-time RT-PCR has been yet unstudied. In the present study, we examined the influence of various transport mediums when combining the direct method and rapid real-time RT-PCR of GeneSoC® (GeneSoC® RT-PCR), the recently developed compact PCR system that adapts novel microfluidic thermal cycling technology. Methods To explore the influence of the transport medium on the GeneSoC® RT-PCR, the concordance of the RNA extraction and direct method was evaluated in the clinical samples collected in viral transport medium (VTM) or eSwab®. The sensitivity of GeneSoC® RT-PCR combined with the direct method was assessed using spiked samples in generic (H2O and PBS) or commercially available transport media (VTM and eSwab®). Analytical sensitivity was examined using clinical specimens collected from the VTM and eSwab®. The inhibitory effect of PCR inhibitors on clinical specimens was assessed using clinical samples diluted 1,000 times. Results While only 1 copy/reaction of RNA was detected in H2O and eSwab® of the spiked samples, a minimum of 5 copies/reaction was detected in PBS (-) and VTM. Among the clinical specimens tested using the direct method, the detection of viral RNA was unstable in the samples containing less than 100 copies/reaction viral RNA in VTM, whereas less than 10 copies/reaction viral RNA were detected in eSwab®. The positive, negative, and overall concordance between the RNA extraction and the direct method was 84%, 100%, and 85%, respectively, in eSwab® samples, whereas the values were 35%, 100%, and 38%, respectively, in VTM samples. When the clinical samples were diluted 1,000 times, GeneSoC® RT-PCR could detect as low as 1.15 copies/reaction RNA using direct method, and the sensitivity was comparable to that of RNA extraction. Conclusion The combination of the direct method and microfluidic rapid PCR machine GeneSoC® has a high sensitivity for detecting SARS-CoV-2 RNA in clinical samples with eSwab® transport medium.
Collapse
Affiliation(s)
- Miyako Takata
- Department of Pathobiological Science and Technology, Graduate School of Medical Science, School of Health Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Masaki Nakamoto
- Division of Infectious Diseases, School of Medicine, Faculty of Medicine, Tottori university, Yonago 683-8503, Japan
| | - Tsuyoshi Kitaura
- Division of Infectious Diseases, School of Medicine, Faculty of Medicine, Tottori university, Yonago 683-8503, Japan
| | - Kensaku Okada
- Division of Infectious Diseases, School of Medicine, Faculty of Medicine, Tottori university, Yonago 683-8503, Japan
| | - Hiroko Endou
- Watarase Research Center Kyorin Pharmaceutical Co., Ltd., Shimotsuga 329-0114, Japan
| | - Athok Shofiudin Ma'arif
- Division of Infectious Diseases, School of Medicine, Faculty of Medicine, Tottori university, Yonago 683-8503, Japan
| | - Yukari Nishikawa
- Division of Infectious Diseases, School of Medicine, Faculty of Medicine, Tottori university, Yonago 683-8503, Japan
| | - Kengo Mukuda
- Division of Infectious Diseases, School of Medicine, Faculty of Medicine, Tottori university, Yonago 683-8503, Japan
| | - Shota Morishita
- Division of Clinical Laboratory, Tottori University Hospital, Yonago 683-8504, Japan
| | - Hiromi Murota
- Division of Clinical Laboratory, Tottori University Hospital, Yonago 683-8504, Japan
| | - Akira Yamasaki
- Division of Respiratory medicine and Rheumatology, Graduate School of Medical Sciences, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Seiji Kageyama
- Division of virology, Department of Microbiology and Immunology, School of Medicine, Faculty of Medicine, Tottori university, Yonago 683-8503, Japan
| | - Naoto Burioka
- Department of Pathobiological Science and Technology, Graduate School of Medical Science, School of Health Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
- Department of Internal Medicine, National Hospital Organization Yonago Medical Center, Yonago 683-0006, Japan
| | - Hiroki Chikumi
- Division of Infectious Diseases, School of Medicine, Faculty of Medicine, Tottori university, Yonago 683-8503, Japan
| |
Collapse
|
114
|
Mohl BP, Blaurock C, Breithaupt A, Riek A, Speakman JR, Hambly C, Bokelmann M, Pei G, Sadeghi B, Dorhoi A, Balkema-Buschmann A. Increased Susceptibility of Rousettus aegyptiacus Bats to Respiratory SARS-CoV-2 Challenge Despite Its Distinct Tropism for Gut Epithelia in Bats. Viruses 2024; 16:1717. [PMID: 39599832 PMCID: PMC11598992 DOI: 10.3390/v16111717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Increasing evidence suggests bats are the ancestral hosts of the majority of coronaviruses. In general, coronaviruses primarily target the gastrointestinal system, while some strains, especially Betacoronaviruses with the most relevant representatives SARS-CoV, MERS-CoV, and SARS-CoV-2, also cause severe respiratory disease in humans and other mammals. We previously reported the susceptibility of Rousettus aegyptiacus (Egyptian fruit bats) to intranasal SARS-CoV-2 infection. Here, we compared their permissiveness to an oral infection versus respiratory challenge (intranasal or orotracheal) by assessing virus shedding, host immune responses, tissue-specific pathology, and physiological parameters. While respiratory challenge with a moderate infection dose of 1 × 104 TCID50 caused a systemic infection with oral and nasal shedding of replication-competent virus, the oral challenge only induced nasal shedding of low levels of viral RNA. Even after a challenge with a higher infection dose of 1 × 106 TCID50, no replication-competent virus was detectable in any of the samples of the orally challenged bats. We postulate that SARS-CoV-2 is inactivated by HCl and digested by pepsin in the stomach of R. aegyptiacus, thereby decreasing the efficiency of an oral infection. Therefore, fecal shedding of RNA seems to depend on systemic dissemination upon respiratory infection. These findings may influence our general understanding of the pathophysiology of coronavirus infections in bats.
Collapse
Affiliation(s)
- Björn-Patrick Mohl
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Suedufer 10, 17493 Greifswald-Insel Riems, Germany; (B.-P.M.); (C.B.); (M.B.); (B.S.)
| | - Claudia Blaurock
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Suedufer 10, 17493 Greifswald-Insel Riems, Germany; (B.-P.M.); (C.B.); (M.B.); (B.S.)
| | - Angele Breithaupt
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Suedufer 10, 17493 Greifswald-Insel Riems, Germany;
| | - Alexander Riek
- Institute of Animal Welfare and Animal Husbandry, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Doernbergstraße 25, 29223 Celle, Germany;
| | - John R. Speakman
- School of Biological Sciences, University of Aberdeen, Aberdeen AB24 2TZ, UK; (J.R.S.); (C.H.)
| | - Catherine Hambly
- School of Biological Sciences, University of Aberdeen, Aberdeen AB24 2TZ, UK; (J.R.S.); (C.H.)
| | - Marcel Bokelmann
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Suedufer 10, 17493 Greifswald-Insel Riems, Germany; (B.-P.M.); (C.B.); (M.B.); (B.S.)
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Nordufer 20, 13353 Berlin, Germany
| | - Gang Pei
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Suedufer 10, 17493 Greifswald-Insel Riems, Germany; (G.P.); (A.D.)
| | - Balal Sadeghi
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Suedufer 10, 17493 Greifswald-Insel Riems, Germany; (B.-P.M.); (C.B.); (M.B.); (B.S.)
| | - Anca Dorhoi
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Suedufer 10, 17493 Greifswald-Insel Riems, Germany; (G.P.); (A.D.)
| | - Anne Balkema-Buschmann
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Suedufer 10, 17493 Greifswald-Insel Riems, Germany; (B.-P.M.); (C.B.); (M.B.); (B.S.)
| |
Collapse
|
115
|
Lett MJ, Otte F, Hauser D, Schön J, Kipfer ET, Hoffmann D, Halwe NJ, Breithaupt A, Ulrich L, Britzke T, Kochmann J, Corleis B, Zhang Y, Urda L, Cmiljanovic V, Lang C, Beer M, Mittelholzer C, Klimkait T. High protection and transmission-blocking immunity elicited by single-cycle SARS-CoV-2 vaccine in hamsters. NPJ Vaccines 2024; 9:206. [PMID: 39472701 PMCID: PMC11522273 DOI: 10.1038/s41541-024-00992-z] [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: 03/05/2024] [Accepted: 10/10/2024] [Indexed: 11/02/2024] Open
Abstract
Vaccines have played a central role in combating the COVID-19 pandemic, but newly emerging SARS-CoV-2 variants are increasingly evading first-generation vaccine protection. To address this challenge, we designed "single-cycle infection SARS-CoV-2 viruses" (SCVs) that lack essential viral genes, possess distinctive immune-modulatory features, and exhibit an excellent safety profile in the Syrian hamster model. Animals intranasally vaccinated with an Envelope-gene-deleted vaccine candidate were fully protected against an autologous challenge with the SARS-CoV-2 virus through systemic and mucosal humoral immune responses. Additionally, the deletion of immune-downregulating viral genes in the vaccine construct prevented challenge virus transmission to contact animals. Moreover, vaccinated animals displayed neither tissue inflammation nor lung damage. Consequently, SCVs hold promising potential to induce potent protection against COVID-19, surpassing the immunity conferred by natural infection, as demonstrated in human immune cells.
Collapse
Affiliation(s)
- Martin Joseph Lett
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Fabian Otte
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - David Hauser
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Jacob Schön
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institute, Greifswald - Isle of Riems, Greifswald, Germany
| | - Enja Tatjana Kipfer
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Donata Hoffmann
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institute, Greifswald - Isle of Riems, Greifswald, Germany
| | - Nico J Halwe
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institute, Greifswald - Isle of Riems, Greifswald, Germany
| | - Angele Breithaupt
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institute, Greifswald - Isle of Riems, Greifswald, Germany
| | - Lorenz Ulrich
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institute, Greifswald - Isle of Riems, Greifswald, Germany
| | - Tobias Britzke
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institute, Greifswald - Isle of Riems, Greifswald, Germany
| | - Jana Kochmann
- Institute of Immunology, Friedrich-Loeffler-Institute, Greifswald - Isle of Riems, Greifswald, Germany
| | - Björn Corleis
- Institute of Immunology, Friedrich-Loeffler-Institute, Greifswald - Isle of Riems, Greifswald, Germany
| | - Yuepeng Zhang
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Lorena Urda
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Christopher Lang
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institute, Greifswald - Isle of Riems, Greifswald, Germany
| | - Christian Mittelholzer
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
- RocketVax AG, Basel, Switzerland
| | - Thomas Klimkait
- Molecular Virology, Department of Biomedicine, University of Basel, Basel, Switzerland.
| |
Collapse
|
116
|
Hadi R, Poddar A, Sonnaila S, Bhavaraju VSM, Agrawal S. Advancing CRISPR-Based Solutions for COVID-19 Diagnosis and Therapeutics. Cells 2024; 13:1794. [PMID: 39513901 PMCID: PMC11545109 DOI: 10.3390/cells13211794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/19/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Since the onset of the COVID-19 pandemic, a variety of diagnostic approaches, including RT-qPCR, RAPID, and LFA, have been adopted, with RT-qPCR emerging as the gold standard. However, a significant challenge in COVID-19 diagnostics is the wide range of symptoms presented by patients, necessitating early and accurate diagnosis for effective management. Although RT-qPCR is a precise molecular technique, it is not immune to false-negative results. In contrast, CRISPR-based detection methods for SARS-CoV-2 offer several advantages: they are cost-effective, time-efficient, highly sensitive, and specific, and they do not require sophisticated instruments. These methods also show promise for scalability, enabling diagnostic tests. CRISPR technology can be customized to target any genomic region of interest, making it a versatile tool with applications beyond diagnostics, including therapeutic development. The CRISPR/Cas systems provide precise gene targeting with immense potential for creating next-generation diagnostics and therapeutics. One of the key advantages of CRISPR/Cas-based therapeutics is the ability to perform multiplexing, where different sgRNAs or crRNAs can target multiple sites within the same gene, reducing the likelihood of viral escape mutants. Among the various CRISPR systems, CRISPR/Cas13 and CARVER (Cas13-assisted restriction of viral expression and readout) are particularly promising. These systems can target a broad range of single-stranded RNA viruses, making them suitable for the diagnosis and treatment of various viral diseases, including SARS-CoV-2. However, the efficacy and safety of CRISPR-based therapeutics must be thoroughly evaluated in pre-clinical and clinical settings. While CRISPR biotechnologies have not yet been fully harnessed to control the current COVID-19 pandemic, there is an optimism that the limitations of the CRISPR/Cas system can be overcome soon. This review discusses how CRISPR-based strategies can revolutionize disease diagnosis and therapeutic development, better preparing us for future viral threats.
Collapse
Affiliation(s)
- Roaa Hadi
- Cell and Molecular Biology Program, Fulbright College of Arts and Sciences, University of Arkansas, Fayetteville, AR 72701, USA;
- Department of Chemical, Biochemical, and Environmental Engineering, University of Maryland, Baltimore County, Baltimore, MD 21250, USA
| | - Abhishek Poddar
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA;
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Shivakumar Sonnaila
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA;
| | | | - Shilpi Agrawal
- Department of Biomedical Engineering, College of Engineering, University of Arkansas, Fayetteville, AR 72701, USA
| |
Collapse
|
117
|
Fabbrini M, D’Amico F, van der Gun BTF, Barone M, Conti G, Roggiani S, Wold KI, Vincenti-Gonzalez MF, de Boer GC, Veloo ACM, van der Meer M, Righi E, Gentilotti E, Górska A, Mazzaferri F, Lambertenghi L, Mirandola M, Mongardi M, Tacconelli E, Turroni S, Brigidi P, Tami A. The gut microbiota as an early predictor of COVID-19 severity. mSphere 2024; 9:e0018124. [PMID: 39297639 PMCID: PMC11540175 DOI: 10.1128/msphere.00181-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/04/2024] [Indexed: 10/30/2024] Open
Abstract
Several studies reported alterations of the human gut microbiota (GM) during COVID-19. To evaluate the potential role of the GM as an early predictor of COVID-19 at disease onset, we analyzed gut microbial samples of 315 COVID-19 patients that differed in disease severity. We observed significant variations in microbial diversity and composition associated with increasing disease severity, as the reduction of short-chain fatty acid producers such as Faecalibacterium and Ruminococcus, and the growth of pathobionts as Anaerococcus and Campylobacter. Notably, we developed a multi-class machine-learning classifier, specifically a convolutional neural network, which achieved an 81.5% accuracy rate in predicting COVID-19 severity based on GM composition at disease onset. This achievement highlights its potential as a valuable early biomarker during the first week of infection. These findings offer promising insights into the intricate relationship between GM and COVID-19, providing a potential tool for optimizing patient triage and streamlining healthcare during the pandemic.IMPORTANCEEfficient patient triage for COVID-19 is vital to manage healthcare resources effectively. This study underscores the potential of gut microbiota (GM) composition as an early biomarker for COVID-19 severity. By analyzing GM samples from 315 patients, significant correlations between microbial diversity and disease severity were observed. Notably, a convolutional neural network classifier was developed, achieving an 81.5% accuracy in predicting disease severity based on GM composition at disease onset. These findings suggest that GM profiling could enhance early triage processes, offering a novel approach to optimizing patient management during the pandemic.
Collapse
Affiliation(s)
- Marco Fabbrini
- Unit of Microbiome
Science and Biotechnology, Department of Pharmacy and Biotechnology,
University of Bologna,
Bologna, Italy
- Human Microbiomics
Unit, Department of Medical and Surgical Sciences, University of
Bologna, Bologna,
Italy
| | - Federica D’Amico
- Human Microbiomics
Unit, Department of Medical and Surgical Sciences, University of
Bologna, Bologna,
Italy
| | - Bernardina T. F. van der Gun
- Department of Medical
Microbiology and Infection Prevention, University of Groningen,
University Medical Center Groningen,
Groningen, the Netherlands
| | - Monica Barone
- Human Microbiomics
Unit, Department of Medical and Surgical Sciences, University of
Bologna, Bologna,
Italy
| | - Gabriele Conti
- Unit of Microbiome
Science and Biotechnology, Department of Pharmacy and Biotechnology,
University of Bologna,
Bologna, Italy
- Human Microbiomics
Unit, Department of Medical and Surgical Sciences, University of
Bologna, Bologna,
Italy
| | - Sara Roggiani
- Unit of Microbiome
Science and Biotechnology, Department of Pharmacy and Biotechnology,
University of Bologna,
Bologna, Italy
- Human Microbiomics
Unit, Department of Medical and Surgical Sciences, University of
Bologna, Bologna,
Italy
| | - Karin I. Wold
- Department of Medical
Microbiology and Infection Prevention, University of Groningen,
University Medical Center Groningen,
Groningen, the Netherlands
| | - María F. Vincenti-Gonzalez
- Department of Medical
Microbiology and Infection Prevention, University of Groningen,
University Medical Center Groningen,
Groningen, the Netherlands
- Spatial Epidemiology
Lab (SpELL), Université Libre de Bruxelles
(ULB), Brussels,
Belgium
| | - Gerolf C. de Boer
- Department of Medical
Microbiology and Infection Prevention, University of Groningen,
University Medical Center Groningen,
Groningen, the Netherlands
| | - Alida C. M. Veloo
- Department of Medical
Microbiology and Infection Prevention, University of Groningen,
University Medical Center Groningen,
Groningen, the Netherlands
| | - Margriet van der Meer
- Department of Medical
Microbiology and Infection Prevention, University of Groningen,
University Medical Center Groningen,
Groningen, the Netherlands
| | - Elda Righi
- Department of
Diagnostics and Public Health, Infectious Diseases Department,
University of Verona,
Verona, Italy
| | - Elisa Gentilotti
- Department of
Diagnostics and Public Health, Infectious Diseases Department,
University of Verona,
Verona, Italy
| | - Anna Górska
- Department of
Diagnostics and Public Health, Infectious Diseases Department,
University of Verona,
Verona, Italy
| | - Fulvia Mazzaferri
- Department of
Diagnostics and Public Health, Infectious Diseases Department,
University of Verona,
Verona, Italy
| | - Lorenza Lambertenghi
- Department of
Diagnostics and Public Health, Infectious Diseases Department,
University of Verona,
Verona, Italy
| | - Massimo Mirandola
- Department of
Diagnostics and Public Health, Infectious Diseases Department,
University of Verona,
Verona, Italy
| | - Maria Mongardi
- Department of
Diagnostics and Public Health, Infectious Diseases Department,
University of Verona,
Verona, Italy
| | - Evelina Tacconelli
- Department of
Diagnostics and Public Health, Infectious Diseases Department,
University of Verona,
Verona, Italy
| | - Silvia Turroni
- Unit of Microbiome
Science and Biotechnology, Department of Pharmacy and Biotechnology,
University of Bologna,
Bologna, Italy
| | - Patrizia Brigidi
- Human Microbiomics
Unit, Department of Medical and Surgical Sciences, University of
Bologna, Bologna,
Italy
| | - Adriana Tami
- Department of Medical
Microbiology and Infection Prevention, University of Groningen,
University Medical Center Groningen,
Groningen, the Netherlands
| |
Collapse
|
118
|
Dong Y, Ritto AP, Damiano RF, Coli AG, Hadade R, Rocca CCDA, Serafim ADP, Guedes BF, Nitrini R, Imamura M, Forlenza OV, Busatto Filho G. Memory complaints after COVID-19: a potential indicator of primary cognitive impairment or a correlate of psychiatric symptoms? Transl Psychiatry 2024; 14:455. [PMID: 39461945 PMCID: PMC11513141 DOI: 10.1038/s41398-024-03154-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 09/02/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Cognitive impairment and symptoms of psychiatric disorders have been reported frequently as features of post-acute sequelae of SARS-CoV-2 infection. This study aims to investigate subjective memory complaints in COVID-19 survivors and determine if these are more strongly associated with objective cognitive impairment related to sequelae of SARS-CoV-2 infection or with symptoms of psychiatric conditions. A total of 608 COVID-19 survivors were evaluated in-person 6-11 months after hospitalization, with 377 patients assigned to a "no subjective memory complaint (SMC)" group and 231 patients assigned to an SMC group based on their Memory Complaint Scale scores. Follow-up evaluations included an objective cognitive battery and scale-based assessments of anxiety, depression, and post-traumatic stress symptoms. We found the perception of memory impairment in COVID-19 survivors to be more strongly associated to core symptoms of psychiatric conditions rather than to primary objective cognitive impairment. Univariate analysis indicated significant differences between the "no SMC" and SMC groups, both for the psychiatric symptom evaluations and for the cognitive evaluations (p < 0.05); however, the psychiatric symptoms all had large partial eta-squared values (ranging from 0.181 to 0.213), whereas the cognitive variables had small/medium partial eta-squared values (ranging from 0.002 to 0.024). Additionally, multiple regression analysis indicated that only female sex and depressive and post-traumatic stress symptoms were predictors of subjective memory complaints. These findings may help guide clinical evaluations for COVID-19 survivors presenting with memory complaints while also serving to expand our growing understanding of the relationship between COVID-19, subjective memory complaints, and the risk of cognitive decline.
Collapse
Affiliation(s)
- Yiling Dong
- The George Washington School of Medicine and Health Sciences, Washington, DC, USA
| | - Ana Paula Ritto
- Departamento de Fisioterapia, Fonoaudiologia e Terapia Ocupacional, Faculdade de Medicina FMUSP, São Paulo, Brazil
| | - Rodolfo Furlan Damiano
- Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo HCFMUSP, São Paulo, Brazil
| | - Amanda Goulart Coli
- Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo HCFMUSP, São Paulo, Brazil
| | - Rodrigo Hadade
- Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo HCFMUSP, São Paulo, Brazil
| | - Cristiana Castanho de Almeida Rocca
- Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo HCFMUSP, São Paulo, Brazil
| | - Antonio de Pádua Serafim
- Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo HCFMUSP, São Paulo, Brazil
| | - Bruno Fukelmann Guedes
- Departamento de Neurologia, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo HCFMUSP, São Paulo, Brazil
| | - Ricardo Nitrini
- Departamento de Neurologia, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo HCFMUSP, São Paulo, Brazil
| | - Marta Imamura
- Departamento de Medicina Legal, Bioética, Medicina do Trabalho e Medicina Física e Reabilitação, Faculdade de Medicina FMUSP, São Paulo, Brazil
| | - Orestes Vicente Forlenza
- Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo HCFMUSP, São Paulo, Brazil
| | - Geraldo Busatto Filho
- Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo HCFMUSP, São Paulo, Brazil.
| |
Collapse
|
119
|
Khodaie A, Heidarzadeh H. Evaluation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) using a high figure-of-merit plasmonic multimode refractive index optical sensor. Sci Rep 2024; 14:25499. [PMID: 39462024 PMCID: PMC11513005 DOI: 10.1038/s41598-024-77336-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024] Open
Abstract
In recent years, following the outbreak of the COVID-19 pandemic, there has been a significant increase in cases of SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) and related deaths worldwide. Despite the pandemic nearing its end due to the introduction of mass-produced vaccines against SARS-CoV-2, early detection and diagnosis of the virus remain crucial in preventing disease progression. This article explores the rapid identification of SARS-CoV-2 by implementing a multimode plasmonic refractive index (MMRI) optical sensor, developed based on the split ring resonator (SRR) design. The Finite Difference Time Domain (FDTD) numerical solution method simulates the sensor. The studied sensor demonstrates three resonance modes within the reflection spectrum ranging from 800 nm to 1400 nm. Its material composition and dimensional parameters are optimized to enhance the sensor's performance. The research indicates that all three resonance modes exhibit strong performance with high sensitivity and figures of merit. Notably, the first mode achieves an exceptional sensitivity of 557 nm/RIU, while the third mode exhibits a commendable sensitivity of 453 nm/RIU and a Figure of Merit (FOM) of 45 RIU-1. These findings suggest that the developed MMRI optical sensor holds significant potential for the early and accurate detection of SARS-CoV-2, contributing to improved disease management and control efforts.
Collapse
Affiliation(s)
- Ali Khodaie
- Department of Electrical and Computer Engineering, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Hamid Heidarzadeh
- Department of Electrical and Computer Engineering, University of Mohaghegh Ardabili, Ardabil, Iran.
| |
Collapse
|
120
|
Perdiguero B, Álvarez E, Marcos-Villar L, Sin L, López-Bravo M, Valverde JR, Sorzano CÓS, Falqui M, Coloma R, Esteban M, Guerra S, Gómez CE. B and T Cell Bi-Cistronic Multiepitopic Vaccine Induces Broad Immunogenicity and Provides Protection Against SARS-CoV-2. Vaccines (Basel) 2024; 12:1213. [PMID: 39591118 PMCID: PMC11598604 DOI: 10.3390/vaccines12111213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND The COVID-19 pandemic, caused by SARS-CoV-2, has highlighted the need for vaccines targeting both neutralizing antibodies (NAbs) and long-lasting cross-reactive T cells covering multiple viral proteins to provide broad and durable protection against emerging variants. METHODS To address this, here we developed two vaccine candidates, namely (i) DNA-CoV2-TMEP, expressing the multiepitopic CoV2-TMEP protein containing immunodominant and conserved T cell regions from SARS-CoV-2 structural proteins, and (ii) MVA-CoV2-B2AT, encoding a bi-cistronic multiepitopic construct that combines conserved B and T cell overlapping regions from SARS-CoV-2 structural proteins. RESULTS Both candidates were assessed in vitro and in vivo demonstrating their ability to induce robust immune responses. In C57BL/6 mice, DNA-CoV2-TMEP enhanced the recruitment of innate immune cells and stimulated SARS-CoV-2-specific polyfunctional T cells targeting multiple viral proteins. MVA-CoV2-B2AT elicited NAbs against various SARS-CoV-2 variants of concern (VoCs) and reduced viral replication and viral yields against the Beta variant in susceptible K18-hACE2 mice. The combination of MVA-CoV2-B2AT with a mutated ISG15 form as an adjuvant further increased the magnitude, breadth and polyfunctional profile of the response. CONCLUSION These findings underscore the potential of these multiepitopic proteins when expressed from DNA or MVA vectors to provide protection against SARS-CoV-2 and its variants, supporting their further development as next-generation COVID-19 vaccines.
Collapse
Affiliation(s)
- Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.M.-V.); (M.E.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
| | - Enrique Álvarez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.M.-V.); (M.E.)
| | - Laura Marcos-Villar
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.M.-V.); (M.E.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
| | - Laura Sin
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
| | - María López-Bravo
- Department of Microbial Biotechnology, CNB-CSIC, 28049 Madrid, Spain;
| | | | | | - Michela Falqui
- Department of Preventive Medicine, Public Health and Microbiology, Faculty of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (M.F.); (R.C.); (S.G.)
| | - Rocío Coloma
- Department of Preventive Medicine, Public Health and Microbiology, Faculty of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (M.F.); (R.C.); (S.G.)
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.M.-V.); (M.E.)
| | - Susana Guerra
- Department of Preventive Medicine, Public Health and Microbiology, Faculty of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (M.F.); (R.C.); (S.G.)
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.M.-V.); (M.E.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
| |
Collapse
|
121
|
Catala A, Davenport BJ, Morrison TE, Catalano CE. Second-Generation Phage Lambda Platform Employing SARS-CoV-2 Fusion Proteins as a Vaccine Candidate. Vaccines (Basel) 2024; 12:1201. [PMID: 39591104 PMCID: PMC11598875 DOI: 10.3390/vaccines12111201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/12/2024] [Accepted: 10/18/2024] [Indexed: 11/28/2024] Open
Abstract
The recent SARS-CoV-2 (COVID-19) pandemic exemplifies how newly emerging and reemerging viruses can quickly overwhelm and cripple global infrastructures. Coupled with synergistic factors such as increasing population densities, the constant and massive mobility of people across geographical areas and substantial changes to ecosystems worldwide, these pathogens pose serious health concerns on a global scale. Vaccines form an indispensable defense, serving to control and mitigate the impact of devastating outbreaks and pandemics. Towards these efforts, we developed a tunable vaccine platform that can be engineered to simultaneously display multiple viral antigens. Here, we describe a second-generation version wherein chimeric proteins derived from SARS-CoV-2 and bacteriophage lambda are engineered and used to decorate phage-like particles with defined surface densities and retention of antigenicity. This streamlines the engineering of particle decoration, thus improving the overall manufacturing potential of the system. In a prime-boost regimen, mice immunized with particles containing as little as 42 copies of the chimeric protein on their surface develop potent neutralizing antibody responses, and immunization protects mice against virulent SARS-CoV-2 challenge. The platform is highly versatile, making it a promising strategy to rapidly develop vaccines against a potentially broad range of infectious diseases.
Collapse
Affiliation(s)
- Alexis Catala
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Bennett J. Davenport
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (B.J.D.); (T.E.M.)
| | - Thomas E. Morrison
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (B.J.D.); (T.E.M.)
| | - Carlos E. Catalano
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| |
Collapse
|
122
|
Paul D, Verma J, Kumar S, Talukdar D, Jana P, Narendrakumar L, Kumar R, Tanwar S, Gosain M, Porey Karmakar S, Pareek M, Mani S, Chaudhuri S, Kshetrapal P, Wadhwa N, Bhatnagar S, Garg PK, Das B. A rapid point-of-care population-scale dipstick assay to identify and differentiate SARS-CoV-2 variants in COVID-19-positive patients. Front Microbiol 2024; 15:1459644. [PMID: 39498137 PMCID: PMC11532176 DOI: 10.3389/fmicb.2024.1459644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 10/09/2024] [Indexed: 11/07/2024] Open
Abstract
Delta and Omicron variants of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) are remarkably contagious, and have been recognized as variants of concern (VOC). The acquisition of spontaneous substitutions or insertion-deletion mutations (indels) in the spike protein-encoding gene substantially increases the binding affinity of the receptor binding domain (RBD)-hACE2 complex and upsurges the transmission of both variants. In this study, we analyzed thousands of genome sequences from 30 distinct SARS-CoV-2 variants, focusing on the unique nucleic acid signatures in the spike gene specific to the Delta and Omicron variants. Using these variant-specific sequences, we synthesized a range of oligonucleotides and optimized a multiplex PCR (mPCR) assay capable of accurately identifying and differentiating between the Delta and Omicron variants. Building on this mPCR assay, we developed a dipstick format by incorporating a tag linker sequence at the 5' end of the forward primer and adding biotin to the 3' end of the oligonucleotides, enhancing the assay's usability and accessibility. Streptavidin-coated latex beads and the dipstick imprinted with a probe for the tag linker sequence in the test strips were used for the detection assay. Our dipstick-based assay, developed as a rapid point-of-care test for identifying and differentiating SARS-CoV-2 variants has the potential to be used in low-resource settings and scaled up to the population level.
Collapse
Affiliation(s)
- Deepjyoti Paul
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Jyoti Verma
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Shakti Kumar
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Daizee Talukdar
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Pradipta Jana
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Lekshmi Narendrakumar
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Roshan Kumar
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Subhash Tanwar
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Mudita Gosain
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| | - Sonali Porey Karmakar
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Madhu Pareek
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Shailendra Mani
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Susmita Chaudhuri
- Multidisciplinary Clinical and Translational Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Pallavi Kshetrapal
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| | - Nitya Wadhwa
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| | - Shinjini Bhatnagar
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| | - Pramod Kumar Garg
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, India
| | - Bhabatosh Das
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
123
|
Jernbom AF, Skoglund L, Pin E, Sjöberg R, Tegel H, Hober S, Rostami E, Rasmusson A, Cunningham JL, Havervall S, Thålin C, Månberg A, Nilsson P. Prevalent and persistent new-onset autoantibodies in mild to severe COVID-19. Nat Commun 2024; 15:8941. [PMID: 39414823 PMCID: PMC11484904 DOI: 10.1038/s41467-024-53356-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024] Open
Abstract
Autoantibodies have been shown to be implied in COVID-19 but the emerging autoantibody repertoire remains largely unexplored. We investigated the new-onset autoantibody repertoire in 525 healthcare workers and hospitalized COVID-19 patients at five time points over a 16-month period in 2020 and 2021 using proteome-wide and targeted protein and peptide arrays. Our results show that prevalent new-onset autoantibodies against a wide range of antigens emerged following SARS-CoV-2 infection in relation to pre-infectious baseline samples and remained elevated for at least 12 months. We found an increased prevalence of new-onset autoantibodies after severe COVID-19 and demonstrated associations between distinct new-onset autoantibodies and neuropsychiatric symptoms post-COVID-19. Using epitope mapping, we determined the main epitopes of selected new-onset autoantibodies, validated them in independent cohorts of neuro-COVID and pre-pandemic healthy controls, and identified sequence similarities suggestive of molecular mimicry between main epitopes and the conserved fusion peptide of the SARS-CoV-2 Spike glycoprotein. Our work describes the complexity and dynamics of the autoantibody repertoire emerging with COVID-19 and supports the need for continued analysis of the new-onset autoantibody repertoire to elucidate the mechanisms of the post-COVID-19 condition.
Collapse
Affiliation(s)
- August F Jernbom
- Division of Affinity Proteomics, Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, Stockholm, Sweden.
| | - Lovisa Skoglund
- Division of Affinity Proteomics, Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Elisa Pin
- Division of Affinity Proteomics, Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Ronald Sjöberg
- Division of Affinity Proteomics, Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Hanna Tegel
- Division of Protein Technology, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Sophia Hober
- Division of Protein Technology, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Elham Rostami
- Section of Neurosurgery, Department of Medical Sciences, Uppsala University Hospital, Uppsala, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Annica Rasmusson
- Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
| | - Janet L Cunningham
- Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
| | - Sebastian Havervall
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm, Sweden
| | - Charlotte Thålin
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm, Sweden
| | - Anna Månberg
- Division of Affinity Proteomics, Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Peter Nilsson
- Division of Affinity Proteomics, Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
124
|
Reguzova A, Müller M, Pagallies F, Burri D, Salomon F, Rziha HJ, Bittner-Schrader Z, Verstrepen BE, Böszörményi KP, Verschoor EJ, Gerhauser I, Elbers K, Esen M, Manenti A, Monti M, Rammensee HG, Derouazi M, Löffler MW, Amann R. A multiantigenic Orf virus-based vaccine efficiently protects hamsters and nonhuman primates against SARS-CoV-2. NPJ Vaccines 2024; 9:191. [PMID: 39414789 PMCID: PMC11484955 DOI: 10.1038/s41541-024-00981-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 09/25/2024] [Indexed: 10/18/2024] Open
Abstract
Among the common strategies to design next-generation COVID-19 vaccines is broadening the antigenic repertoire thereby aiming to increase efficacy against emerging variants of concern (VoC). This study describes a new Orf virus-based vector (ORFV) platform to design a multiantigenic vaccine targeting SARS-CoV-2 spike and nucleocapsid antigens. Vaccine candidates were engineered, either expressing spike protein (ORFV-S) alone or co-expressing nucleocapsid protein (ORFV-S/N). Mono- and multiantigenic vaccines elicited comparable levels of spike-specific antibodies and virus neutralization in mice. Results from a SARS-CoV-2 challenge model in hamsters suggest cross-protective properties of the multiantigenic vaccine against VoC, indicating improved viral clearance with ORFV-S/N, as compared to equal doses of ORFV-S. In a nonhuman primate challenge model, vaccination with the ORFV-S/N vaccine resulted in long-term protection against SARS-CoV-2 infection. These results demonstrate the potential of the ORFV platform for prophylactic vaccination and represent a preclinical development program supporting first-in-man studies with the multiantigenic ORFV vaccine.
Collapse
Affiliation(s)
- Alena Reguzova
- Institute of Immunology, University Hospital Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Melanie Müller
- Institute of Immunology, University Hospital Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Felix Pagallies
- Institute of Immunology, University Hospital Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Dominique Burri
- Speransa Therapeutics, Bethmannstrasse 8, 60311, Frankfurt am Main, Germany
| | - Ferdinand Salomon
- Institute of Immunology, University Hospital Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Hanns-Joachim Rziha
- Institute of Immunology, University Hospital Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Zsofia Bittner-Schrader
- Institute of Immunology, University Hospital Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Babs E Verstrepen
- Department of Virology, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288GJ, Rijswijk, The Netherlands
- Department of Viroscience, Erasmus University Medical Center, Dr. Molewaterplein 50, 3015 GE, Rotterdam, The Netherlands
| | - Kinga P Böszörményi
- Department of Virology, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288GJ, Rijswijk, The Netherlands
| | - Ernst J Verschoor
- Department of Virology, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288GJ, Rijswijk, The Netherlands
| | - Ingo Gerhauser
- Department of Pathology, University of Veterinary Medicine Hannover Foundation, Bünteweg 17, 30559, Hannover, Germany
| | - Knut Elbers
- Boehringer Ingelheim International GmbH, Binger Strasse 173, 55216, Ingelheim am Rhein, Germany
- ViraTherapeutics GmbH, Bundesstraße 27, 6063, Rum, Austria
| | - Meral Esen
- Institute of Tropical Medicine, University of Tübingen, Wilhelmstraße 27, 72074, Tübingen, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen; Cluster of Excellence (EXC2124) "Controlling Microbes to Fight Infection", Tübingen, Germany
| | - Alessandro Manenti
- VisMederi Srl., Strada del Petriccio e Belriguardo 35, 53100, Siena, Italy
| | - Martina Monti
- VisMederi Srl., Strada del Petriccio e Belriguardo 35, 53100, Siena, Italy
| | - Hans-Georg Rammensee
- Institute of Immunology, University Hospital Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Madiha Derouazi
- Speransa Therapeutics, Bethmannstrasse 8, 60311, Frankfurt am Main, Germany
| | - Markus W Löffler
- Institute of Immunology, University Hospital Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany.
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty of Tübingen, University Hospital Tübingen, Otfried-Müller-Str. 4/1, 72076, Tübingen, Germany.
- Centre for Clinical Transfusion Medicine, Otfried-Müller-Str. 4/1, 72076, Tübingen, Germany.
| | - Ralf Amann
- Institute of Immunology, University Hospital Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany.
| |
Collapse
|
125
|
Arrousse N, Berdimurodov E, Bogacheva M, Zakham F, Esslali S, EL Kadiri S, Taleb M, Vapalahti O. N-alkylation of amines for the synthesis of potential antiviral agents: A structural modification approach. Heliyon 2024; 10:e38587. [PMID: 39397970 PMCID: PMC11470537 DOI: 10.1016/j.heliyon.2024.e38587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/15/2024] Open
Abstract
The threat of emerging viral outbreaks has increased the need for fast and effective development of therapeutics against emerging pathogens. One approach is to modify the structure of existing therapeutic agents to achieve the desired antiviral properties. Here, we attempted to synthesize a new antiviral compound by modifying the structure of chloroquine using the N-alkylation of the primary amine (N1,N1-diethylpentane-1,4-diamine) that is used in chloroquine synthesis. Chloroquine is commonly used to treat malaria. Like chloroquine, chloroquine is used for treating conditions such as rheumatoid arthritis, lupus, and malaria. For instance, in malaria treatment, it targets and inhibits the growth of the malaria parasite, aiding in its elimination from the body. The synthesized compounds MP1, C1, and TT1 were further tested in vitro against the B.1 lineage of SARS-CoV-2. One of the compounds, MP1, demonstrated minor effectiveness, with an IC50 of XX at only a high concentration (at a concentration of 60 μM) and decreased both the number of SARS-CoV-2 copies and the amount of infectious virus. Although the synthesized compounds failed to markedly inhibit SARS-CoV-2, this could be a pontial mechanism for manipulating the drug structure against other pathogens. MP1, TT1, C1, and chloroquine diphosphate were used as ligands for molecular docking to determine the principal interactions between these compounds and the active site of the protein downloaded from the Protein Data Bank (PDB ID: 6lzg). Finally, ADMET assays were performed on the synthesized compounds to determine their pharmacokinetics and bioavailability.
Collapse
Affiliation(s)
- Nadia Arrousse
- School of Science and Engineering, Al Akhawayn University in Ifrane, Hassan II avenue, 53000 Ifrane, Morocco
- Laboratory of Engineering, Electrochemistry, Modelling and Environment (LIEME), Faculty of Sciences, University Sidi Mohamed Ben Abdellah, Fez, Morocco
| | - Elyor Berdimurodov
- Faculty of Chemistry, National University of Uzbekistan, Tashkent, 100034, Uzbekistan
- Physics and Chemistry, Western Caspian University, AZ-1001, Baku, Azerbaijan
- University of Tashkent for Applied Sciences, Str. Gavhar 1, 100149, Tashkent, Uzbekistan
- Physics and Chemistry, Tashkent Institute of Irrigation and Agricultural Mechanization Engineers” National Research University, 100000, Tashkent, Uzbekistan
| | - Mariia Bogacheva
- Viral Zone Research Unit, Faculty of Medicine, University of Helsinki, Finland
| | - Fathiah Zakham
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Soukaina Esslali
- Laboratory of Chemistry-Biology Applied to the Environment, Chemistry Department, Faculty of Sciences, Moulay-Ismail University, B.P. 11201, Zitoune, Meknes, Morocco
| | - Sghir EL Kadiri
- Laboratory of Applied Chemistry & Environment (LCAE), Faculty of Science, Mohammed First University, Oujda, Morocco
| | - Mustapha Taleb
- Laboratory of Engineering, Electrochemistry, Modelling and Environment (LIEME), Faculty of Sciences, University Sidi Mohamed Ben Abdellah, Fez, Morocco
| | - Olli Vapalahti
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
126
|
Arevalo-Rodriguez I, Mateos-Haro M, Dinnes J, Ciapponi A, Davenport C, Buitrago-Garcia D, Bennouna-Dalero T, Roqué-Figuls M, Van den Bruel A, von Eije KJ, Emperador D, Hooft L, Spijker R, Leeflang MM, Takwoingi Y, Deeks JJ. Laboratory-based molecular test alternatives to RT-PCR for the diagnosis of SARS-CoV-2 infection. Cochrane Database Syst Rev 2024; 10:CD015618. [PMID: 39400904 PMCID: PMC11472845 DOI: 10.1002/14651858.cd015618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
BACKGROUND Diagnosing people with a SARS-CoV-2 infection played a critical role in managing the COVID-19 pandemic and remains a priority for the transition to long-term management of COVID-19. Initial shortages of extraction and reverse transcription polymerase chain reaction (RT-PCR) reagents impaired the desired upscaling of testing in many countries, which led to the search for alternatives to RNA extraction/purification and RT-PCR testing. Reference standard methods for diagnosing the presence of SARS-CoV-2 infection rely primarily on real-time reverse transcription-polymerase chain reaction (RT-PCR). Alternatives to RT-PCR could, if sufficiently accurate, have a positive impact by expanding the range of diagnostic tools available for the timely identification of people infected by SARS-CoV-2, access to testing and the use of resources. OBJECTIVES To assess the diagnostic accuracy of alternative (to RT-PCR assays) laboratory-based molecular tests for diagnosing SARS-CoV-2 infection. SEARCH METHODS We searched the COVID-19 Open Access Project living evidence database from the University of Bern until 30 September 2020 and the WHO COVID-19 Research Database until 31 October 2022. We did not apply language restrictions. SELECTION CRITERIA We included studies of people with suspected or known SARS-CoV-2 infection, or where tests were used to screen for infection, and studies evaluating commercially developed laboratory-based molecular tests for the diagnosis of SARS-CoV-2 infection considered as alternatives to RT-PCR testing. We also included all reference standards to define the presence or absence of SARS-CoV-2, including RT-PCR tests and established clinical diagnostic criteria. DATA COLLECTION AND ANALYSIS Two authors independently screened studies and resolved disagreements by discussing them with a third author. Two authors independently extracted data and assessed the risk of bias and applicability of the studies using the QUADAS-2 tool. We presented sensitivity and specificity, with 95% confidence intervals (CIs), for each test using paired forest plots and summarised results using average sensitivity and specificity using a bivariate random-effects meta-analysis. We illustrated the findings per index test category and assay brand compared to the WHO's acceptable sensitivity and specificity threshold for diagnosing SARS-CoV-2 infection using nucleic acid tests. MAIN RESULTS We included data from 64 studies reporting 94 cohorts of participants and 105 index test evaluations, with 74,753 samples and 7517 confirmed SARS-CoV-2 cases. We did not identify any published or preprint reports of accuracy for a considerable number of commercially produced NAAT assays. Most cohorts were judged at unclear or high risk of bias in more than three QUADAS-2 domains. Around half of the cohorts were considered at high risk of selection bias because of recruitment based on COVID status. Three quarters of 94 cohorts were at high risk of bias in the reference standard domain because of reliance on a single RT-PCR result to determine the absence of SARS-CoV-2 infection or were at unclear risk of bias due to a lack of clarity about the time interval between the index test assessment and the reference standard, the number of missing results, or the absence of a participant flow diagram. For index tests categories with four or more evaluations and when summary estimations were possible, we found that: a) For RT-PCR assays designed to omit/adapt RNA extraction/purification, the average sensitivity was 95.1% (95% CI 91.1% to 97.3%), and the average specificity was 99.7% (95% CI 98.5% to 99.9%; based on 27 evaluations, 2834 samples and 1178 SARS-CoV-2 cases); b) For RT-LAMP assays, the average sensitivity was 88.4% (95% CI 83.1% to 92.2%), and the average specificity was 99.7% (95% CI 98.7% to 99.9%; 24 evaluations, 29,496 samples and 2255 SARS-CoV-2 cases); c) for TMA assays, the average sensitivity was 97.6% (95% CI 95.2% to 98.8%), and the average specificity was 99.4% (95% CI 94.9% to 99.9%; 14 evaluations, 2196 samples and 942 SARS-CoV-2 cases); d) for digital PCR assays, the average sensitivity was 98.5% (95% CI 95.2% to 99.5%), and the average specificity was 91.4% (95% CI 60.4% to 98.7%; five evaluations, 703 samples and 354 SARS-CoV-2 cases); e) for RT-LAMP assays omitting/adapting RNA extraction, the average sensitivity was 73.1% (95% CI 58.4% to 84%), and the average specificity was 100% (95% CI 98% to 100%; 24 evaluations, 14,342 samples and 1502 SARS-CoV-2 cases). Only two index test categories fulfil the WHO-acceptable sensitivity and specificity requirements for SARS-CoV-2 nucleic acid tests: RT-PCR assays designed to omit/adapt RNA extraction/purification and TMA assays. In addition, WHO-acceptable performance criteria were met for two assays out of 35 when tests were used according to manufacturer instructions. At 5% prevalence using a cohort of 1000 people suspected of SARS-CoV-2 infection, the positive predictive value of RT-PCR assays omitting/adapting RNA extraction/purification will be 94%, with three in 51 positive results being false positives, and around two missed cases. For TMA assays, the positive predictive value of RT-PCR assays will be 89%, with 6 in 55 positive results being false positives, and around one missed case. AUTHORS' CONCLUSIONS Alternative laboratory-based molecular tests aim to enhance testing capacity in different ways, such as reducing the time, steps and resources needed to obtain valid results. Several index test technologies with these potential advantages have not been evaluated or have been assessed by only a few studies of limited methodological quality, so the performance of these kits was undetermined. Only two index test categories with enough evaluations for meta-analysis fulfil the WHO set of acceptable accuracy standards for SARS-CoV-2 nucleic acid tests: RT-PCR assays designed to omit/adapt RNA extraction/purification and TMA assays. These assays might prove to be suitable alternatives to RT-PCR for identifying people infected by SARS-CoV-2, especially when the alternative would be not having access to testing. However, these findings need to be interpreted and used with caution because of several limitations in the evidence, including reliance on retrospective samples without information about the symptom status of participants and the timing of assessment. No extrapolation of found accuracy data for these two alternatives to any test brands using the same techniques can be made as, for both groups, one test brand with high accuracy was overrepresented with 21/26 and 12/14 included studies, respectively. Although we used a comprehensive search and had broad eligibility criteria to include a wide range of tests that could be alternatives to RT-PCR methods, further research is needed to assess the performance of alternative COVID-19 tests and their role in pandemic management.
Collapse
Affiliation(s)
- Ingrid Arevalo-Rodriguez
- Clinical Biostatistics Unit, Hospital Universitario Ramón y Cajal (IRYCIS). CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Evidence Production & Methods Directorate, Cochrane, London, UK
| | - Miriam Mateos-Haro
- Clinical Biostatistics Unit, Hospital Universitario Ramón y Cajal (IRYCIS), Madrid, Spain
- Doctoral programme in Clinical Medicine and Public Health, Universidad de Granada, Granada, Spain
| | - Jacqueline Dinnes
- Department of Applied Health Sciences, School of Health Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Agustín Ciapponi
- Argentine Cochrane Centre, Institute for Clinical Effectiveness and Health Policy (IECS-CONICET), Buenos Aires, Argentina
| | - Clare Davenport
- Department of Applied Health Sciences, School of Health Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Diana Buitrago-Garcia
- Institute for Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Hospital Universitario Mayor - Méderi. Universidad del Rosario, Bogotá, Colombia
| | - Tayeb Bennouna-Dalero
- Preventive Medicine and Public Health Department, Hospital Universitario Ramón y Cajal (IRYCIS), Madrid, Spain
| | - Marta Roqué-Figuls
- Iberoamerican Cochrane Centre, Institut de Recerca Sant Pau (IR SANT PAU), CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | | | - Karin J von Eije
- Department of Viroscience, ErasmusMC, University Medical Center, Rotterdam, Netherlands
| | | | - Lotty Hooft
- Cochrane Netherlands, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - René Spijker
- Cochrane Netherlands, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Mariska Mg Leeflang
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Yemisi Takwoingi
- Department of Applied Health Sciences, School of Health Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Jonathan J Deeks
- Department of Applied Health Sciences, School of Health Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| |
Collapse
|
127
|
Akaberi D, Pourghasemi Lati M, Krambrich J, Berger J, Neilsen G, Strandback E, Turunen SP, Wannberg J, Gullberg H, Moche M, Chinthakindi PK, Nyman T, Sarafianos SG, Sandström A, Järhult JD, Sandberg K, Lundkvist Å, Verho O, Lennerstrand J. Identification of novel and potent inhibitors of SARS-CoV-2 main protease from DNA-encoded chemical libraries. Antimicrob Agents Chemother 2024; 68:e0090924. [PMID: 39194208 PMCID: PMC11459923 DOI: 10.1128/aac.00909-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/03/2024] [Indexed: 08/29/2024] Open
Abstract
In vitro screening of large compound libraries with automated high-throughput screening is expensive and time-consuming and requires dedicated infrastructures. Conversely, the selection of DNA-encoded chemical libraries (DECLs) can be rapidly performed with routine equipment available in most laboratories. In this study, we identified novel inhibitors of SARS-CoV-2 main protease (Mpro) through the affinity-based selection of the DELopen library (open access for academics), containing 4.2 billion compounds. The identified inhibitors were peptide-like compounds containing an N-terminal electrophilic group able to form a covalent bond with the nucleophilic Cys145 of Mpro, as confirmed by x-ray crystallography. This DECL selection campaign enabled the discovery of the unoptimized compound SLL11 (IC50 = 30 nM), proving that the rapid exploration of large chemical spaces enabled by DECL technology allows for the direct identification of potent inhibitors avoiding several rounds of iterative medicinal chemistry. As demonstrated further by x-ray crystallography, SLL11 was found to adopt a highly unique U-shaped binding conformation, which allows the N-terminal electrophilic group to loop back to the S1' subsite while the C-terminal amino acid sits in the S1 subsite. MP1, a close analog of SLL11, showed antiviral activity against SARS-CoV-2 in the low micromolar range when tested in Caco-2 and Calu-3 (EC50 = 2.3 µM) cell lines. As peptide-like compounds can suffer from low cell permeability and metabolic stability, the cyclization of the compounds will be explored in the future to improve their antiviral activity.
Collapse
Affiliation(s)
- Dario Akaberi
- Department of Medical Biochemistry and Microbiology, Zoonosis Science Center, Uppsala University, Uppsala, Sweden
| | | | - Janina Krambrich
- Department of Medical Biochemistry and Microbiology, Zoonosis Science Center, Uppsala University, Uppsala, Sweden
| | - Julia Berger
- Department of Medical Sciences, Clinical Microbiology, Uppsala University, Uppsala, Sweden
| | - Grace Neilsen
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Emilia Strandback
- Department of Medical Biochemistry and Biophysics, Protein Science Facility, Karolinska Institutet, Stockholm, Sweden
| | - S. Pauliina Turunen
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Solna, Sweden
- Drug Discovery and Development, Science for Life Laboratory, Solna, Sweden
| | - Johan Wannberg
- Department of Medicinal Chemistry, Science for Life Laboratory, BMC, Uppsala University, Uppsala, Sweden
| | - Hjalmar Gullberg
- Science for Life Laboratory, Biochemical and Cellular Assay Facility, Drug Discovery and Development Platform, Department of Biochemistry and Biophysics, Stockholm University, Solna, Stockholm, Sweden
| | - Martin Moche
- Department of Medical Biochemistry and Biophysics, Protein Science Facility, Karolinska Institutet, Stockholm, Sweden
| | - Praveen Kumar Chinthakindi
- The Beijer Laboratory, Department of Medicinal Chemistry, Drug Design and Discovery, Uppsala University, Uppsala, Sweden
| | - Tomas Nyman
- Department of Medical Biochemistry and Biophysics, Protein Science Facility, Karolinska Institutet, Stockholm, Sweden
| | - Stefan G. Sarafianos
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Anja Sandström
- The Beijer Laboratory, Department of Medicinal Chemistry, Drug Design and Discovery, Uppsala University, Uppsala, Sweden
| | - Josef D. Järhult
- Department of Medical Sciences, Zoonosis Science Center, Uppsala University, Uppsala, Sweden
| | - Kristian Sandberg
- Science for Life Laboratory, Drug Discovery & Development Platform, Uppsala University, Uppsala, Sweden
| | - Åke Lundkvist
- Department of Medical Biochemistry and Microbiology, Zoonosis Science Center, Uppsala University, Uppsala, Sweden
| | - Oscar Verho
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Johan Lennerstrand
- Department of Medical Sciences, Clinical Microbiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
128
|
Tang Y, Tang K, Hu Y, Ye ZW, Luo W, Luo C, Cao H, Wang R, Yue X, Liu D, Liu C, Ge X, Liu T, Chen Y, Yuan S, Deng L. M protein ectodomain-specific immunity restrains SARS-CoV-2 variants replication. Front Immunol 2024; 15:1450114. [PMID: 39416782 PMCID: PMC11480003 DOI: 10.3389/fimmu.2024.1450114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 09/05/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction The frequent occurrence of mutations in the SARS-CoV-2 Spike (S) protein, with up to dozens of mutations, poses a severe threat to the current efficacy of authorized COVID-19 vaccines. Membrane (M) protein, which is the most abundant viral structural protein, exhibits a high level of amino acid sequence conservation. M protein ectodomain could be recognized by specific antibodies; however, the extent to which it is immunogenic and provides protection remains unclear. Methods We designed and synthesized multiple peptides derived from coronavirus M protein ectodomains, and determined the secondary structure of specific peptides using circular dichroism (CD) spectroscopy. Enzyme-linked immunosorbent assay (ELISA) was utilized to detect IgG responses against the synthesized peptides in clinical samples. To evaluate the immunogenicity of peptide vaccines, BALB/c mice were intraperitoneally immunized with peptide-keyhole limpet hemocyanin (KLH) conjugates adjuvanted with incomplete Freund's adjuvant (IFA). The humoral and T-cell immune responses induced by peptide-KLH conjugates were assessed using ELISA and ELISpot assays, respectively. The efficacy of the S2M2-30-KLH vaccine against SARS-CoV-2 variants was evaluated in vivo using the K18-hACE2 transgenic mouse model. The inhibitory effect of mouse immune serum on SARS-CoV-2 virus replication in vitro was evaluated using microneutralization assays. The subcellular localization of the M protein was evaluated using an immunofluorescent staining method, and the Fc-mediated antibody-dependent cellular cytotoxicity (ADCC) activity of the S2M2-30-specific monoclonal antibody (mAb) was measured using an ADCC reporter assay. Results Seroconversion rates for ectodomain-specific IgG were observed to be high in both SARS-CoV-2 convalescent patients and individuals immunized with inactivated vaccines. To assess the protective efficacy of the M protein ectodomain-based vaccine, we initially identified a highly immunogenic peptide derived from this ectodomain, named S2M2-30. The mouse serum specific to S2M2-30 showed inhibitory effects on the replication of SARS-CoV-2 variants in vitro. Immunizations of K18-hACE2-transgenic mice with the S2M2-30-keyhole limpet hemocyanin (KLH) vaccine significantly reduced the lung viral load caused by B.1.1.7/Alpha (UK) infection. Further mechanism investigations reveal that serum neutralizing activity, specific T-cell response and Fc-mediated antibody-dependent cellular cytotoxicity (ADCC) correlate with the specific immuno-protection conferred by S2M2-30. Discussion The findings of this study suggest that the antibody responses against M protein ectodomain in the population most likely exert a beneficial effect on preventing various SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Yibo Tang
- Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha, China
| | - Kaiming Tang
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yunqi Hu
- School of Public Health, Sun Yat-sen University, Shenzhen, China
| | - Zi-Wei Ye
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Wanyu Luo
- School of Public Health, Sun Yat-sen University, Shenzhen, China
| | - Cuiting Luo
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Hehe Cao
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ran Wang
- Laboratory of Infection and Virology, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Xinyu Yue
- School of Public Health, Sun Yat-sen University, Shenzhen, China
| | - Dejian Liu
- Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha, China
| | - Cuicui Liu
- Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha, China
| | - Xingyi Ge
- Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha, China
| | - Tianlong Liu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yaoqing Chen
- School of Public Health, Sun Yat-sen University, Shenzhen, China
- National Medical Products Administration Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Sun Yat-sen University, Guangzhou, China
| | - Shuofeng Yuan
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Lei Deng
- Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha, China
- Research and Development Department, Beijing Weimiao Biotechnology Co. Ltd., Beijing, China
| |
Collapse
|
129
|
Vránová L, Poláková I, Vaníková Š, Saláková M, Musil J, Vaníčková M, Vencálek O, Holub M, Bohoněk M, Řezáč D, Dresler J, Tachezy R, Šmahel M. Multiparametric analysis of the specific immune response against SARS-CoV-2. Infect Dis (Lond) 2024; 56:851-869. [PMID: 38805304 DOI: 10.1080/23744235.2024.2358379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/24/2024] [Accepted: 05/17/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND SARS-CoV-2, which causes COVID-19, has killed more than 7 million people worldwide. Understanding the development of postinfectious and postvaccination immune responses is necessary for effective treatment and the introduction of appropriate antipandemic measures. OBJECTIVES We analysed humoral and cell-mediated anti-SARS-CoV-2 immune responses to spike (S), nucleocapsid (N), membrane (M), and open reading frame (O) proteins in individuals collected up to 1.5 years after COVID-19 onset and evaluated immune memory. METHODS Peripheral blood mononuclear cells and serum were collected from patients after COVID-19. Sampling was performed in two rounds: 3-6 months after infection and after another year. Most of the patients were vaccinated between samplings. SARS-CoV-2-seronegative donors served as controls. ELISpot assays were used to detect SARS-CoV-2-specific T and B cells using peptide pools (S, NMO) or recombinant proteins (rS, rN), respectively. A CEF peptide pool consisting of selected viral epitopes was applied to assess the antiviral T-cell response. SARS-CoV-2-specific antibodies were detected via ELISA and a surrogate virus neutralisation assay. RESULTS We confirmed that SARS-CoV-2 infection induces the establishment of long-term memory IgG+ B cells and memory T cells. We also found that vaccination enhanced the levels of anti-S memory B and T cells. Multivariate comparison also revealed the benefit of repeated vaccination. Interestingly, the T-cell response to CEF was lower in patients than in controls. CONCLUSION This study supports the importance of repeated vaccination for enhancing immunity and suggests a possible long-term perturbation of the overall antiviral immune response caused by SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Lucie Vránová
- Department of Genetics and Microbiology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Ingrid Poláková
- Department of Genetics and Microbiology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Šárka Vaníková
- Department of Immunomonitoring and Flow Cytometry, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Martina Saláková
- Department of Genetics and Microbiology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Jan Musil
- Department of Immunomonitoring and Flow Cytometry, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Marie Vaníčková
- Department of Genetics and Microbiology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Ondřej Vencálek
- Department of Mathematical Analysis and Applications of Mathematics, Faculty of Science, Palacky University in Olomouc, Olomouc, Czech Republic
| | - Michal Holub
- Department of Infectious Diseases, First Faculty of Medicine, Military University Hospital Prague and Charles University, Prague, Czech Republic
| | - Miloš Bohoněk
- Department of Hematology and Blood Transfusion, Military University Hospital Prague, Prague, Czech Republic
- Faculty of Biomedical Engineering, Czech Technical University, Prague, Czech Republic
| | - David Řezáč
- Department of Infectious Diseases, First Faculty of Medicine, Military University Hospital Prague and Charles University, Prague, Czech Republic
| | - Jiří Dresler
- Military Health Institute, Military Medical Agency, Prague, Czech Republic
| | - Ruth Tachezy
- Department of Genetics and Microbiology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Michal Šmahel
- Department of Genetics and Microbiology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| |
Collapse
|
130
|
de Abreu Góes Pereira VM, Gularte JS, Demoliner M, da Silva MS, Girardi V, Filippi M, Frohlich J, Fink P, Hansen AW, Ferreira HL, Afrough B, Kritz-Wilson A, Spilki FR. Influenza outbreak during the surge of SARS-CoV-2 omicron in a metropolitan area from southern Brazil: genomic surveillance. J Med Virol 2024; 96:e29944. [PMID: 39360646 DOI: 10.1002/jmv.29944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 08/08/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024]
Abstract
Influenza circulation was significantly affected in 2020-21 by the COVID-19 pandemic. During this time, few influenza cases were recorded. However, in the summer of 2021-22, an increase in atypical influenza cases was observed, leading to the resurgence of influenza in the southernmost state of Brazil, Rio Grande do Sul (RS). The present study aimed to identify the circulation of FLUAV, FLUBV and SARS-CoV-2 and characterize the influenza genomes in respiratory samples using high-throughput sequencing technology (HTS). Respiratory samples (n = 694) from patients in RS were selected between July 2021 and August 2022. The samples were typed using reverse transcriptase real-time PCR (RT-qPCR) and showed 32% (223/694) of the samples to be positive for SARS-CoV-2, 7% for FLUAV (H3) (49/694). FLUBV was not detected. RT-qPCR data also resulted in FLUAV and SARS-CoV-2 co-infections in 1.7% (4/223) of samples tested. Whole genome sequencing of FLUAV produced 15 complete genomes of the H3N2 subtype, phylogenetically classified in the 3C.2a1b.2a.2a.3 subclade and revealing the dominance of viruses in the southern region of Brazil. Mutation analysis identified 72 amino acid substitutions in all genes, highlighting ongoing genetic evolution with potential implications for vaccine effectiveness, viral fitness, and pathogenicity. This study underscores limitations in current surveillance systems, advocating for comprehensive data inclusion to enhance understanding of influenza epidemiology in southern Brazil. These findings contribute valuable insights to inform more effective public health responses and underscore the critical need for continuous genomic surveillance.
Collapse
Affiliation(s)
| | - Juliana Schons Gularte
- Departamento de Virologia, Laboratório de Microbiologia Molecular, Universidade Feevale, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Meriane Demoliner
- Departamento de Virologia, Laboratório de Microbiologia Molecular, Universidade Feevale, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Mariana Soares da Silva
- Departamento de Virologia, Laboratório de Microbiologia Molecular, Universidade Feevale, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Viviane Girardi
- Departamento de Virologia, Laboratório de Microbiologia Molecular, Universidade Feevale, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Micheli Filippi
- Departamento de Virologia, Laboratório de Microbiologia Molecular, Universidade Feevale, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Julia Frohlich
- Departamento de Virologia, Laboratório de Microbiologia Molecular, Universidade Feevale, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Pietra Fink
- Departamento de Virologia, Laboratório de Microbiologia Molecular, Universidade Feevale, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Alana Witt Hansen
- Departamento de Virologia, Laboratório de Microbiologia Molecular, Universidade Feevale, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Helena Lage Ferreira
- Department of Veterinary Medicine, FZEA- USP, University of Sao Paulo, Pirassununga, São Paulo, Brazil
| | - Babak Afrough
- Health Security Agency, New Variant Assessment Platform (NVAP), UK
| | | | - Fernando Rosado Spilki
- Departamento de Virologia, Laboratório de Microbiologia Molecular, Universidade Feevale, Novo Hamburgo, Rio Grande do Sul, Brazil
| |
Collapse
|
131
|
Ben Said S, Jaballah R, Yaakoubi H, Ben Salah H, Youssef R, Mzid N, Kacemi M, Trabelsi I, Ben Ayed A, Ben Ayed S, Boukadida L, Zorgati A, Boukef R. Canine olfactory detection and its relevance for the medical identification of patients with COVID-19. Infect Dis (Lond) 2024; 56:880-886. [PMID: 38889329 DOI: 10.1080/23744235.2024.2363887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/20/2024] Open
Abstract
INTRODUCTION The assessment of Volatile Organic Compounds (VOCs) in exhaled breath or sweat represents a potential non-invasive and rapid diagnostic tool for respiratory diseases. OBJECTIVE To determine if trained dogs can reliably identify the odour associated with COVID19. METHODS This is a monocentric prospective study carried out in the Emergency Department (ED) of a university hospital fromJulyto November 2021.Axillary sweat samples from all patients were collected bytwo trained health care professionals. The samples were collected in the form of sterile gauze swabs placed under the armpits for at least 4 h for each patient.Then, Tubes wereshiftedto the double-blind dog training centre for VOC detection by two individuals. RESULTS Dogs were tested using a total of 129 axillary sweat samples; 69 of the 107 patients who tested positive for COVID-19 based on their odours had a positive PCR/Antigen test and 19 of the 22 patients who were tested negative for COVID-19 by the dogs had a negative PCR test. The sniffer dog infection detection method had a sensitivity of 95.83% and a specificity of 33.33%. The PPV was 64.49% and the NPVwas 86.36%. The measurement of the intensity of the connection between the two variables (disease/sign) was very strong (Q = 0.84). This link is statistically significant (X2 = 19.13) with a probability p ≤ 0.001. CONCLUSION Overall, the use of trained detection dogs as a screening method for SARS-CoV-2 is an interesting avenue of research that warrants further exploration and validation.
Collapse
Affiliation(s)
- Salma Ben Said
- Emergency Department, Sahloul University Hospital, Sousse, Tunisia
| | - Rahma Jaballah
- Emergency Department, Sahloul University Hospital, Sousse, Tunisia
| | - Hajer Yaakoubi
- Emergency Department, Sahloul University Hospital, Sousse, Tunisia
| | - Houda Ben Salah
- Emergency Department, Sahloul University Hospital, Sousse, Tunisia
| | - Rym Youssef
- Emergency Department, Sahloul University Hospital, Sousse, Tunisia
| | - Nouhel Mzid
- Emergency Department, Sahloul University Hospital, Sousse, Tunisia
| | - Marouen Kacemi
- Emergency Department, Sahloul University Hospital, Sousse, Tunisia
| | - Imen Trabelsi
- Emergency Department, Sahloul University Hospital, Sousse, Tunisia
| | - Ali Ben Ayed
- K9 Dog Center Security And Training, sousse, Tunisia
| | - Saed Ben Ayed
- K9 Dog Center Security And Training, sousse, Tunisia
| | - Lotfi Boukadida
- Emergency Department, Sahloul University Hospital, Sousse, Tunisia
| | - Asma Zorgati
- Emergency Department, Sahloul University Hospital, Sousse, Tunisia
| | - Riadh Boukef
- Emergency Department, Sahloul University Hospital, Sousse, Tunisia
| |
Collapse
|
132
|
Lee H, Liao JD, Tsai HP, Wang H, Sitjar J. Focused ion beam-fabricated nanorod substrate for label-free surface-enhanced Raman spectroscopy and enabling dual virus detection. Talanta 2024; 278:126466. [PMID: 38944940 DOI: 10.1016/j.talanta.2024.126466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/27/2024] [Accepted: 06/22/2024] [Indexed: 07/02/2024]
Abstract
The COVID-19 pandemic presents global challenges, notably with co-infections in respiratory tract involving SARS-CoV-2 variants and influenza strains. Detecting multiple viruses simultaneously is crucial for accurate diagnosis, effective tracking infectious sources, and containment of the epidemic. This study uses a label-free surface-enhanced Raman spectroscopy (SERS) method using Au NPs/pZrO2 (250) and FIB-made Au NRs (100) to detect dual viruses, including SARS-CoV-2 Delta variant (D) and influenza A (A) or B (B) virus. Results demonstrate distinct peaks facilitating virus differentiation, especially between D and A or B, with clear disparities between substrates; specific peaks at 950 and 1337 cm-1 are pivotal for discerning viruses using Au NPs/pZrO2 (250), while those at 1050, 1394, and 1450 cm-1 and 1033, 1165, 1337, and 1378 cm-1 are key for validation using Au NRs (100). Differences in substrate surface morphology and spatial disposition of accommodating viruses significantly influence hotspot formation and Raman signal amplification efficiency, thereby affecting the ability to distinguish various viruses. Furthermore, both substrates offer insights, even in the presence of oxymetazoline hydrochloride (an interfering substance), with practical implications in viral diagnosis. The customized design and reproducibility underscore efficient Raman signal amplification, even in challenging environments, highlighting potential for widespread virus detection.
Collapse
Affiliation(s)
- Han Lee
- Laboratory of Engineered Materials for Biomedical Applications, Department of Materials Science and Engineering, National Cheng Kung University, 1 University Road, Tainan, 701, Taiwan.
| | - Jiunn-Der Liao
- Laboratory of Engineered Materials for Biomedical Applications, Department of Materials Science and Engineering, National Cheng Kung University, 1 University Road, Tainan, 701, Taiwan.
| | - Huey-Pin Tsai
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 704, Taiwan; Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan.
| | - Hao Wang
- Laboratory of Engineered Materials for Biomedical Applications, Department of Materials Science and Engineering, National Cheng Kung University, 1 University Road, Tainan, 701, Taiwan.
| | - Jaya Sitjar
- Laboratory of Engineered Materials for Biomedical Applications, Department of Materials Science and Engineering, National Cheng Kung University, 1 University Road, Tainan, 701, Taiwan.
| |
Collapse
|
133
|
Stojčević-Maletić J, Barjaktarović I, Đerić M, Čabarkapa V, Petrović V, Patić A, Bogdanović-Vasić S. COMPARISON OF ABBOTT REAL TIME SARS-COV-2 ASSAY AND LIFERIVER NOVEL CORONAVIRUS REAL TIME MULTIPLEX KIT FOR THE RT-PCR BASED DETECTION OF SARS-COV-2 FROM NASOPHARYNGEAL SWABS. Acta Clin Croat 2024; 63:275-282. [PMID: 40104229 PMCID: PMC11912847 DOI: 10.20471/acc.2024.63.02.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 02/15/2022] [Indexed: 03/20/2025] Open
Abstract
The objective of this study was to compare performance of two authorized tests, the Abbott Real Time SARS-CoV-2 (ACOV) assay (Abbott Molecular Inc., North Chicago, IL, USA) and Liferiver Novel Coronavirus Real Time Multiplex RT-PCR (Liferiver 2019-nCoV) kit (BioVendor Group, Brno, Czech Republic), and to determine whether the selection of targeted genes has an impact on test specificity. We included 105 nasopharyngeal swabs from adult individuals with symptoms or suspected of coronavirus disease 2019 (COVID-19), aged from 26 to 91 years, previously tested by the ACOV and subsequently tested by the Liferiver 2019-nCoV. In this comparative analysis, we found that the ACOV assay detected more cases of COVID-19 infection than the Liferiver 2019-nCoV assay. The Liferiver 2019-nCoV kit showed a high level of agreement with the ACOV assay. The positive percent agreement was 88.89% (95% confidence intervals (95% CI): 77.42%-100.0%), and the kappa coefficient (kappa) was 0.901 (95% CI: 0.817-0.985). The negative percent agreement was 94.12% (95% CI: 89.74%-98.50%), while 4.76% of SARS-CoV-2 cases were false-negative using the Liferiver test. However, due to the possible false-negative results using the Liferiver 2019-nCOV test, we recommend complete testing with the ACOV assay.
Collapse
Affiliation(s)
- Jelena Stojčević-Maletić
- University of Novi Sad, Faculty of Medicine, Novi Sad, Serbia
- University Clinical Center of Vojvodina, Center for Laboratory Diagnostics, Novi Sad, Serbia
| | - Iva Barjaktarović
- University of Novi Sad, Faculty of Medicine, Novi Sad, Serbia
- University Clinical Center of Vojvodina, Center for Laboratory Diagnostics, Novi Sad, Serbia
| | - Mirjana Đerić
- University of Novi Sad, Faculty of Medicine, Novi Sad, Serbia
- University Clinical Center of Vojvodina, Center for Laboratory Diagnostics, Novi Sad, Serbia
| | - Velibor Čabarkapa
- University of Novi Sad, Faculty of Medicine, Novi Sad, Serbia
- University Clinical Center of Vojvodina, Center for Laboratory Diagnostics, Novi Sad, Serbia
| | - Vladimir Petrović
- University of Novi Sad, Faculty of Medicine, Novi Sad, Serbia
- Institute of Public Health of Vojvodina, Novi Sad, Serbia
| | - Aleksandra Patić
- University of Novi Sad, Faculty of Medicine, Novi Sad, Serbia
- Institute of Public Health of Vojvodina, Novi Sad, Serbia
| | | |
Collapse
|
134
|
Verma SK, Ana-Sosa-Batiz F, Timis J, Shafee N, Maule E, Pinto PBA, Conner C, Valentine KM, Cowley DO, Miller R, Elong Ngono A, Tran L, Varghese K, Dos Santos Alves RP, Hastie KM, Saphire EO, Webb DR, Jarnagin K, Kim K, Shresta S. Influence of Th1 versus Th2 immune bias on viral, pathological, and immunological dynamics in SARS-CoV-2 variant-infected human ACE2 knock-in mice. EBioMedicine 2024; 108:105361. [PMID: 39353281 PMCID: PMC11472634 DOI: 10.1016/j.ebiom.2024.105361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/30/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Mouse models that recapitulate key features of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection are important tools for understanding complex interactions between host genetics, immune responses, and SARS-CoV-2 pathogenesis. Little is known about how predominantly cellular (Th1 type) versus humoral (Th2 type) immune responses influence SARS-CoV-2 dynamics, including infectivity and disease course. METHODS We generated knock-in (KI) mice expressing human ACE2 (hACE2) and/or human TMPRSS2 (hTMPRSS2) on Th1-biased (C57BL/6; B6) and Th2-biased (BALB/c) genetic backgrounds. Mice were infected intranasally with SARS-CoV-2 Delta (B.1.617.2) or Omicron BA.1 (B.1.1.529) variants, followed by assessment of disease course, respiratory tract infection, lung histopathology, and humoral and cellular immune responses. FINDINGS In both B6 and BALB/c mice, hACE2 expression was required for infection of the lungs with Delta, but not Omicron BA.1. Disease severity was greater in Omicron BA.1-infected hTMPRSS2-KI and double-KI BALB/c mice compared with B6 mice, and in Delta-infected double-KI B6 and BALB/c mice compared with hACE2-KI mice. hACE2-KI B6 mice developed more severe lung pathology and more robust SARS-CoV-2-specific splenic CD8 T cell responses compared with hACE2-KI BALB/c mice. There were no notable differences between the two genetic backgrounds in plasma cell, germinal center B cell, or antibody responses to SARS-CoV-2. INTERPRETATION SARS-CoV-2 Delta and Omicron BA.1 infection, disease course, and CD8 T cell response are influenced by the host genetic background. These humanized mice hold promise as important tools for investigating the mechanisms underlying the heterogeneity of SARS-CoV-2-induced pathogenesis and immune response. FUNDING This work was funded by NIH U19 AI142790-02S1, the GHR Foundation, the Arvin Gottleib Foundation, and the Overton family (to SS and EOS); Prebys Foundation (to SS); NIH R44 AI157900 (to KJ); and by an American Association of Immunologists Career Reentry Fellowship (FASB).
Collapse
Affiliation(s)
- Shailendra Kumar Verma
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | | | - Julia Timis
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | | | - Erin Maule
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | | | - Chris Conner
- Synbal Inc., 1759 Yorktown Rd., San Mateo, CA, 94402, USA
| | - Kristen M Valentine
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Dale O Cowley
- TransViragen Inc., 109 Mason Farm Road, Chapel Hill, NC, 27599, USA
| | - Robyn Miller
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Annie Elong Ngono
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Linda Tran
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Krithik Varghese
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | | | - Kathryn M Hastie
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Erica Ollmann Saphire
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - David R Webb
- Synbal Inc., 1759 Yorktown Rd., San Mateo, CA, 94402, USA
| | - Kurt Jarnagin
- Synbal Inc., 1759 Yorktown Rd., San Mateo, CA, 94402, USA
| | - Kenneth Kim
- Histopathology Core Facility, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA.
| | - Sujan Shresta
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA; Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, UC San Diego School of Medicine, La Jolla, CA, 92037, USA.
| |
Collapse
|
135
|
Panei CJ, Fuentealba NA, Bravi ME, Moré G, Brasso N. Nested PCR effective to detect low viral loads of SARS-CoV-2 in animal samples. Prev Vet Med 2024; 231:106303. [PMID: 39128181 DOI: 10.1016/j.prevetmed.2024.106303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/17/2024] [Accepted: 07/30/2024] [Indexed: 08/13/2024]
Abstract
SARS-CoV-2 emerged from an animal source and was then transmitted to humans, causing the COVID-19 pandemic. Since a wide range of animals are susceptible to SARS-CoV-2 infection, the zoonotic potential of SARS-CoV-2 increases with every new animal infected. The molecular gold standard assay for SARS-CoV-2 detection is real-time RT-PCR, where the Ct obtained is proportional to the amount of nucleic acid and can be a semi-quantitative measure of the viral load. However, since the use of real-time RT-PCR assays in animal samples is low due to the high costs, the use of validated nested PCR assays will help to monitor large-scale animal samplings, by reducing the costs of detection. In the present study, 140 samples from dogs and cats (15 SARS-CoV-2-positive samples with Ct values from 27 to 33, and 125 negative samples), previously analyzed by real-time RT-PCR, were analyzed by nested PCR. To increase the number of positive samples to determine the sensitivity of the assay, 40 human samples obtained during COVID-19 diagnosis in 2020 were included. The specificity of the primers was analyzed against samples positive to canine coronavirus (CCV) and feline infectious peritonitis virus (FIPV). To calculate the limit of detection (LoD) of the nested PCR, the viral load was estimated extrapolating the Ct value obtained by real-time RT-PCR. The Ct values obtained were considered as semi-quantitative and were able to distinguish between high, moderate and low viral loads. The Kappa value or "agreement" between assays and reliability of the nested PCR were also determined. Eleven of the animal samples analyzed by nested PCR targeting the N gene were detected as positive, while 129 were detected as negative to the virus, with Ct values ranging between17 and 31.5. All the samples from humans analyzed by nested PCR were positive. These results indicate that the assay has a sensitivity of near 95 % and a specificity of 100 %. No unspecific reactions analyzed by nested PCR were observed with the samples positive to CCV and FIPV. The samples detected as positive to SARS-CoV-2 by nested PCR were those that presented a Ct between17 and 31.5. The LoD of the nested PCR was estimated close to 50 copies/µL of viral load, corresponding with a Ct of 31.5. The Kappa value between assays was excellent (k = 0.829). The results obtained demonstrate that nested PCR is useful to detect SARS-CoV-2 low viral loads at a lower cost than with real-time RT-PCR.
Collapse
Affiliation(s)
- Carlos Javier Panei
- Laboratorio de Virología, Facultad de Ciencias Veterinarias (FCV), Universidad Nacional de La Plata (UNLP), 60 & 118, La Plata, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), (C1425FQB), Godoy Cruz 2290, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Nadia Analía Fuentealba
- Laboratorio de Virología, Facultad de Ciencias Veterinarias (FCV), Universidad Nacional de La Plata (UNLP), 60 & 118, La Plata, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), (C1425FQB), Godoy Cruz 2290, Ciudad Autónoma de Buenos Aires, Argentina
| | - María Emilia Bravi
- Laboratorio de Virología, Facultad de Ciencias Veterinarias (FCV), Universidad Nacional de La Plata (UNLP), 60 & 118, La Plata, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), (C1425FQB), Godoy Cruz 2290, Ciudad Autónoma de Buenos Aires, Argentina
| | - Gastón Moré
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty University of Bern, Länggassstrasse 122, Bern 3012, Switzerland; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), (C1425FQB), Godoy Cruz 2290, Ciudad Autónoma de Buenos Aires, Argentina
| | - Natalia Brasso
- Laboratorio de Virología, Facultad de Ciencias Veterinarias (FCV), Universidad Nacional de La Plata (UNLP), 60 & 118, La Plata, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), (C1425FQB), Godoy Cruz 2290, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
136
|
Schoefbaenker M, Günther T, Lorentzen EU, Romberg ML, Hennies MT, Neddermeyer R, Müller MM, Mellmann A, Bojarzyn CR, Lenz G, Stelljes M, Hrincius ER, Vollenberg R, Ludwig S, Tepasse PR, Kühn JE. Characterisation of the antibody-mediated selective pressure driving intra-host evolution of SARS-CoV-2 in prolonged infection. PLoS Pathog 2024; 20:e1012624. [PMID: 39405332 PMCID: PMC11508484 DOI: 10.1371/journal.ppat.1012624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 10/25/2024] [Accepted: 09/28/2024] [Indexed: 10/26/2024] Open
Abstract
Neutralising antibodies against the SARS-CoV-2 spike (S) protein are major determinants of protective immunity, though insufficient antibody responses may cause the emergence of escape mutants. We studied the humoral immune response causing intra-host evolution in a B-cell depleted, haemato-oncologic patient experiencing clinically severe, prolonged SARS-CoV-2 infection with a virus of lineage B.1.177.81. Following bamlanivimab treatment at an early stage of infection, the patient developed a bamlanivimab-resistant mutation, S:S494P. After five weeks of apparent genetic stability, the emergence of additional substitutions and deletions within the N-terminal domain (NTD) and the receptor binding domain (RBD) of S was observed. Notably, the composition and frequency of escape mutations changed in a short period with an unprecedented dynamic. The triple mutant S:Delta141-4 E484K S494P became dominant until virus elimination. Routine serology revealed no evidence of an antibody response in the patient. A detailed analysis of the variant-specific immune response by pseudotyped virus neutralisation test, surrogate virus neutralisation test, and immunoglobulin-capture enzyme immunoassay showed that the onset of an IgM-dominated antibody response coincided with the appearance of escape mutations. The formation of neutralising antibodies against S:Delta141-4 E484K S494P correlated with virus elimination. One year later, the patient experienced clinically mild re-infection with Omicron BA.1.18, which was treated with sotrovimab and resulted in an increase in Omicron-reactive antibodies. In conclusion, the onset of an IgM-dominated endogenous immune response in an immunocompromised patient coincided with the appearance of additional mutations in the NTD and RBD of S in a bamlanivimab-resistant virus. Although virus elimination was ultimately achieved, this humoral immune response escaped detection by routine diagnosis and created a situation temporarily favouring the rapid emergence of various antibody escape mutants with known epidemiological relevance.
Collapse
Affiliation(s)
| | - Theresa Günther
- Institute of Virology Muenster, University of Muenster, Muenster, Germany
| | - Eva Ulla Lorentzen
- Institute of Virology Muenster, University of Muenster, Muenster, Germany
| | | | - Marc Tim Hennies
- Institute of Virology Muenster, University of Muenster, Muenster, Germany
| | - Rieke Neddermeyer
- Institute of Virology Muenster, University of Muenster, Muenster, Germany
| | | | - Alexander Mellmann
- Institute of Hygiene, University Hospital Muenster, University of Muenster, Muenster, Germany
| | | | - Georg Lenz
- Department of Medicine A, Haematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
| | - Matthias Stelljes
- Department of Medicine A, Haematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
| | | | - Richard Vollenberg
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clinical Infectiology, University Hospital Muenster, Muenster, Germany
| | - Stephan Ludwig
- Institute of Virology Muenster, University of Muenster, Muenster, Germany
| | - Phil-Robin Tepasse
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clinical Infectiology, University Hospital Muenster, Muenster, Germany
| | - Joachim Ewald Kühn
- Institute of Virology Muenster, University of Muenster, Muenster, Germany
| |
Collapse
|
137
|
He J, Zhong R, Xue L, Wang Y, Chen Y, Xiong Z, Yang Z, Chen S, Liang W, He J. Exhaled Volatile Organic Compounds Detection in Pneumonia Screening: A Comprehensive Meta-analysis. Lung 2024; 202:501-511. [PMID: 39180684 PMCID: PMC11427597 DOI: 10.1007/s00408-024-00737-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/01/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND Pneumonia is a common lower respiratory tract infection, and early diagnosis is crucial for timely treatment and improved prognosis. Traditional diagnostic methods for pneumonia, such as chest imaging and microbiological examinations, have certain limitations. Exhaled volatile organic compounds (VOCs) detection, as an emerging non-invasive diagnostic technique, has shown potential application value in pneumonia screening. METHOD A systematic search was conducted on PubMed, Embase, Cochrane Library, and Web of Science, with the retrieval time up to March 2024. The inclusion criteria were diagnostic studies evaluating exhaled VOCs detection for the diagnosis of pneumonia, regardless of the trial design type. A meta-analysis was performed using a bivariate model for sensitivity and specificity. RESULTS A total of 14 diagnostic studies were included in this meta-analysis. The pooled results demonstrated that exhaled VOCs detection had a combined sensitivity of 0.94 (95% CI: 0.92-0.95) and a combined specificity of 0.83 (95% CI: 0.81-0.84) in pneumonia screening, with an area under the summary receiver operating characteristic (SROC) curve (AUC) of 0.96. The pooled diagnostic odds ratio (DOR) was 104.37 (95% CI: 27.93-390.03), and the pooled positive and negative likelihood ratios (LR) were 8.98 (95% CI: 3.88-20.80) and 0.11 (95% CI: 0.05-0.22), indicating a high diagnostic performance. CONCLUSION This study highlights the potential of exhaled VOCs detection as an effective, non-invasive screening method for pneumonia, which could facilitate future diagnosis in pneumonia. Further high-quality, large-scale studies are required to confirm the clinical utility of exhaled VOCs detection in pneumonia screening. STUDY REGISTRATION PROSPERO, Review no. CRD42024520498.
Collapse
Affiliation(s)
- Juan He
- Nanshan School, Guangzhou Medical University, Jingxiu Road, Panyu District, Guangzhou, 511436, China.
| | - Ran Zhong
- Department of Thoracic Surgery and Oncology, National Center for Respiratory Health, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Nanshan School, Guangzhou Medical University, Jingxiu Road, Panyu District, Guangzhou, 511436, China
| | - Linlu Xue
- Guangzhou Yuexiu Huanghuagang Street Community Health Service Center, Guangzhou, 510075, China
| | - Yixuan Wang
- Nanshan School, Guangzhou Medical University, Jingxiu Road, Panyu District, Guangzhou, 511436, China
| | - Yang Chen
- Nanshan School, Guangzhou Medical University, Jingxiu Road, Panyu District, Guangzhou, 511436, China
| | - Zihui Xiong
- Nanshan School, Guangzhou Medical University, Jingxiu Road, Panyu District, Guangzhou, 511436, China
| | - Ziya Yang
- The First Clinical School, Guangzhou Medical University, Jingxiu Road, Panyu District, Guangzhou, 511436, China
| | - Sitong Chen
- ChromX Health Company Limited, Guangzhou, 510120, China
| | - Wenhua Liang
- Department of Thoracic Surgery and Oncology, National Center for Respiratory Health, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
| | - Jianxing He
- Department of Thoracic Surgery and Oncology, National Center for Respiratory Health, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
| |
Collapse
|
138
|
Long X, Gong Z, Gan Y, Yuan P, Tang Y, Yang Y, Zhong S. Sensitive Detection of Escherichia coli O157:H7 Using Allosteric Probe and Hairpin Switches-Based Isothermal Transcription Amplification. Anal Chem 2024; 96:15608-15613. [PMID: 39307963 DOI: 10.1021/acs.analchem.4c02413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2024]
Abstract
Pathogens pose a serious threat to public and population health, leading to serious outbreak and spread of diseases irrespective of the region. The capability to directly, sensitively, and specifically detect viable pathogens in low numbers in food and clinical samples is very desirable but remains a challenge. In this work, we present a novel assay of a combination of an aptamer-based allosteric probe and hairpin switch-controlled T7 RNA polymerase-based isothermal transcription amplification, which enables rapid, ultrasensitive, label-free detection of direct pathogens. It can detect Escherichia coli as low as 73.2 CFU/mL. Moreover, with the usage of the proposed assay, sensitive quantification of E. coliO157:H7 in milk samples has been achieved, showing significant potential as a simple and sensitive tool to quantify pathogens in milk and other foods.
Collapse
Affiliation(s)
- Xi Long
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Zan Gong
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Yuqing Gan
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Panpan Yuan
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Yalan Tang
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Yanjing Yang
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Shian Zhong
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, the "Double-First Class" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha Medical University, Changsha 410219, China
| |
Collapse
|
139
|
Hsu JL, Liu MC, Tsau PW, Chung FT, Lin SM, Chen ML, Ro LS. Clinical presentations, systemic inflammation response and ANDC scores in hospitalized patients with COVID-19. Sci Rep 2024; 14:22480. [PMID: 39341876 PMCID: PMC11438960 DOI: 10.1038/s41598-024-73001-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 09/12/2024] [Indexed: 10/01/2024] Open
Abstract
The association of anosmia/ageusia with a positive severe respiratory syndrome coronavirus 2 (SARS-CoV-2) test is well-established, suggesting these symptoms are reliable indicators of coronavirus disease 2019 (COVID-19) infection. This study investigates the clinical characteristics and systemic inflammatory markers in hospitalized COVID-19 patients in Taiwan, focusing on those with anosmia/ageusia. We conducted a retrospective observational study on 231 hospitalized COVID-19 patients (alpha variant) from April to July 2021. Clinical symptoms, dyspnea grading, and laboratory investigations, including neutrophil-lymphocyte ratios (NLRs), platelet-lymphocyte ratios (PLRs), and ANDC scores (an early warning score), were analyzed. Cough (64.1%), fever (58.9%), and dyspnea (56.3%) were the most common symptoms, while anosmia/ageusia affected 9% of patients. Those with anosmia/ageusia were younger, had lower BMI, lower systemic inflammatory markers, and better ANDC scores than those without these symptoms. Female patients exhibited lower NLR values and ANDC scores compared to male patients (all p < 0.05). Multivariable regression analysis demonstrated significant associations between NLR and CRP and ferritin levels (all p < 0.01), and between PLR and ESR and ferritin levels (p < 0.01). Categorized ANDC scores significantly correlated with the total hospital length of stay (all p < 0.05). Despite ethnic differences in the prevalence of anosmia/ageusia, our study highlights similar clinical presentations and inflammatory profiles to those observed in Western countries. The ANDC score effectively predicted hospital stay duration. These findings suggest that anosmia/ageusia may be associated with less severe disease and a lower inflammatory response, particularly in younger and female patients. The ANDC score can serve as a valuable prognostic tool in assessing the severity and expected hospital stay of COVID-19 patients.
Collapse
Affiliation(s)
- Jung Lung Hsu
- Department of Neurology, New Taipei Municipal TuCheng Hospital (Built and Operated by Chang Gung Medical Foundation, New Taipei City, Taiwan.
- Department of Neurology, Chang Gung Memorial Hospital at Linkou Medical Center and Chang Gung University College of Medicine, 5 Fu-Shin Street, Kwei-Shan, Taoyuan, 333, Taiwan.
- Graduate Institute of Mind, Brain, & Consciousness, Taipei Medical University, Taipei, Taiwan.
- Brain & Consciousness Research Center, Shuang Ho Hospital, New Taipei City, Taiwan.
| | - Mei-Chuen Liu
- Department of Nursing, New Taipei Municipal TuCheng Hospital (Built and Operated by Chang Gung Medical Foundation, New Taipei City, Taiwan
| | - Po-Wei Tsau
- Department of Neurology, New Taipei Municipal TuCheng Hospital (Built and Operated by Chang Gung Medical Foundation, New Taipei City, Taiwan
| | - Fu-Tsai Chung
- Department of Thoracic Medicine, New Taipei Municipal TuCheng Hospital (Built and Operated by Chang Gung Medical Foundation, New Taipei City, Taiwan
- Department of Respiratory Therapy, New Taipei Municipal TuCheng Hospital (Built and Operated by Chang Gung Medical Foundation, New Taipei City, Taiwan
- Department of Thoracic Medicine, Chang Gung Memorial Hospital at Linkou and School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shu-Min Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital at Linkou and School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Mei-Lan Chen
- School of Nursing, Byrdine F. Lewis College of Nursing and Health Professions, Georgia State University, Atlanta, USA
| | - Long-Sun Ro
- Department of Neurology, Chang Gung Memorial Hospital at Linkou Medical Center and Chang Gung University College of Medicine, 5 Fu-Shin Street, Kwei-Shan, Taoyuan, 333, Taiwan.
| |
Collapse
|
140
|
Chen C, Wang Y, Kaur G, Adiga A, Espinoza B, Venkatramanan S, Warren A, Lewis B, Crow J, Singh R, Lorentz A, Toney D, Marathe M. Wastewater-based Epidemiology for COVID-19 Surveillance and Beyond: A Survey. ARXIV 2024:arXiv:2403.15291v2. [PMID: 38562450 PMCID: PMC10984000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The pandemic of COVID-19 has imposed tremendous pressure on public health systems and social economic ecosystems over the past years. To alleviate its social impact, it is important to proactively track the prevalence of COVID-19 within communities. The traditional way to estimate the disease prevalence is to estimate from reported clinical test data or surveys. However, the coverage of clinical tests is often limited and the tests can be labor-intensive, requires reliable and timely results, and consistent diagnostic and reporting criteria. Recent studies revealed that patients who are diagnosed with COVID-19 often undergo fecal shedding of SARS-CoV-2 virus into wastewater, which makes wastewater-based epidemiology for COVID-19 surveillance a promising approach to complement traditional clinical testing. In this paper, we survey the existing literature regarding wastewater-based epidemiology for COVID-19 surveillance and summarize the current advances in the area. Specifically, we have covered the key aspects of wastewater sampling, sample testing, and presented a comprehensive and organized summary of wastewater data analytical methods. Finally, we provide the open challenges on current wastewater-based COVID-19 surveillance studies, aiming to encourage new ideas to advance the development of effective wastewater-based surveillance systems for general infectious diseases.
Collapse
Affiliation(s)
- Chen Chen
- Department of Computer Science, University of Virginia, Charlottesville, 22904, United States
| | - Yunfan Wang
- Department of Computer Science, University of Virginia, Charlottesville, 22904, United States
| | - Gursharn Kaur
- Biocomplexity Institute and Initiative, University of Virginia, Charlottesville, 22904, United States
| | - Aniruddha Adiga
- Biocomplexity Institute and Initiative, University of Virginia, Charlottesville, 22904, United States
| | - Baltazar Espinoza
- Biocomplexity Institute and Initiative, University of Virginia, Charlottesville, 22904, United States
| | - Srinivasan Venkatramanan
- Biocomplexity Institute and Initiative, University of Virginia, Charlottesville, 22904, United States
| | - Andrew Warren
- Biocomplexity Institute and Initiative, University of Virginia, Charlottesville, 22904, United States
| | - Bryan Lewis
- Biocomplexity Institute and Initiative, University of Virginia, Charlottesville, 22904, United States
| | - Justin Crow
- Virginia Department of Health, Richmond, 23219, United States
| | - Rekha Singh
- Virginia Department of Health, Richmond, 23219, United States
| | - Alexandra Lorentz
- Division of Consolidated Laboratory Services, Department of General Services, Richmond, 23219, United States
| | - Denise Toney
- Division of Consolidated Laboratory Services, Department of General Services, Richmond, 23219, United States
| | - Madhav Marathe
- Department of Computer Science, University of Virginia, Charlottesville, 22904, United States
- Biocomplexity Institute and Initiative, University of Virginia, Charlottesville, 22904, United States
| |
Collapse
|
141
|
Fischer RJ, Bushmaker T, Williamson BN, Pérez-Pérez L, Feldmann F, Lovaglio J, Scott D, Saturday G, Feldmann H, Munster VJ, de Wit E, van Doremalen N. Compartmentalized SARS-CoV-2 Replication in the Upper vs Lower Respiratory Tract After Intranasal Inoculation or Aerosol Exposure. J Infect Dis 2024; 230:657-661. [PMID: 38261786 PMCID: PMC11420804 DOI: 10.1093/infdis/jiae018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/21/2023] [Accepted: 01/19/2024] [Indexed: 01/25/2024] Open
Abstract
Nonhuman primate models are essential for the development of vaccines and antivirals against infectious diseases. Rhesus macaques are a widely utilized infection model for SARS-CoV-2. We compared cellular tropism and virus replication in rhesus macaques inoculated with SARS-CoV-2 via the intranasal route or via exposure to aerosols. Intranasal inoculation resulted in replication in the upper respiratory tract with limited involvement in the lower respiratory tract, whereas exposure to aerosols resulted in infection throughout the respiratory tract. In comparison with multiroute inoculation, intranasal and aerosol inoculation resulted in reduced SARS-CoV-2 replication in the respiratory tract.
Collapse
Affiliation(s)
| | | | | | | | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Jamie Lovaglio
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Dana Scott
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Greg Saturday
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | | | | | | | | |
Collapse
|
142
|
Rahimi S, Balusamy SR, Perumalsamy H, Ståhlberg A, Mijakovic I. CRISPR-Cas target recognition for sensing viral and cancer biomarkers. Nucleic Acids Res 2024; 52:10040-10067. [PMID: 39189452 PMCID: PMC11417378 DOI: 10.1093/nar/gkae736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/08/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024] Open
Abstract
Nucleic acid-based diagnostics is a promising venue for detection of pathogens causing infectious diseases and mutations related to cancer. However, this type of diagnostics still faces certain challenges, and there is a need for more robust, simple and cost-effective methods. Clustered regularly interspaced short palindromic repeats (CRISPRs), the adaptive immune systems present in the prokaryotes, has recently been developed for specific detection of nucleic acids. In this review, structural and functional differences of CRISPR-Cas proteins Cas9, Cas12 and Cas13 are outlined. Thereafter, recent reports about applications of these Cas proteins for detection of viral genomes and cancer biomarkers are discussed. Further, we highlight the challenges associated with using these technologies to replace the current diagnostic approaches and outline the points that need to be considered for designing an ideal Cas-based detection system for nucleic acids.
Collapse
Affiliation(s)
- Shadi Rahimi
- Division of Systems and Synthetic Biology, Department of Life Sciences, Chalmers University of Technology, SE-41296 Gothenburg, Sweden
| | - Sri Renukadevi Balusamy
- Department of Food Science and Biotechnology, Sejong University, Gwangjin-gu, Seoul, Republic of Korea
| | - Haribalan Perumalsamy
- Center for Creative Convergence Education, Hanyang University, Seoul 04763, Republic of Korea
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul 04763, South Korea
| | - Anders Ståhlberg
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ivan Mijakovic
- Division of Systems and Synthetic Biology, Department of Life Sciences, Chalmers University of Technology, SE-41296 Gothenburg, Sweden
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
143
|
Kittang BR, Øien AT, Engtrø E, Skjellanger M, Krüger K. Clinical characteristics, treatment, and outcomes for elderly patients in a dedicated Covid-19 ward at a primary health care facility in western Norway: a retrospective observational study. BMC Health Serv Res 2024; 24:1098. [PMID: 39300524 PMCID: PMC11414310 DOI: 10.1186/s12913-024-11539-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND The coronavirus pandemic has hit the oldest and frailest individuals hard, particularly patients and residents in nursing homes. In March 2020, we established a Covid-19 ward at a nursing home in Bergen, western Norway for elderly patients with Sars-CoV-2 infection and in the need of treatment and care in a primary health care facility. The aims of this study were to describe the organization of the ward, the clinical outcomes of infection, treatment, mortality rates in the population, the level of advanced care planning, and end-of-life care for those who died. METHODS We present patient characteristics, outcomes, vaccination status, treatment, decisions regarding treatment intensity upon clinical deterioration, and mortality for the patients in the ward. Clinical factors possibly related to a fatal outcome were analysed with chi square test (categorical variables) or t-test (continuous variables). RESULTS 257 patients were included from March 2020 to April 2022. Fifty-nine patients (23.0%) developed respiratory failure. Ten patients (3.9%) were admitted to hospital. Advance care planning was undertaken for 245 (95.3%) of the patients. 30-day mortality rate decreased from 42 to 4% during the study period. Of the 29 (11.3%) patients who died, all were well alleviated in the terminal phase, and 26 (89.7%) of them had a Clinical Frailty Scale (CFS) value ≥ 7. A high score for CFS, respiratory failure and respiratory co-infection were significantly associated with Covid-19 related death within 30 days. CONCLUSIONS Covid-19-related mortality markedly decreased during the study period, and a high score for CFS was related to a fatal outcome. Thorough planning of treatment intensity upon deterioration, low hospitalization rates, and good relief for those who died suggest that dedicated Covid-19 wards in nursing homes can provide good treatment for the patients and relieve other nursing homes and specialist health care services.
Collapse
Affiliation(s)
- Bård Reiakvam Kittang
- Department of Nursing Home Medicine, Municipality of Bergen, Bergen, Norway.
- Department of Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway.
- Department of Clinical Science, University of Bergen, Bergen, Norway.
| | - Ane Tveiten Øien
- Department of Nursing Home Medicine, Municipality of Bergen, Bergen, Norway
| | - Einar Engtrø
- Department of Nursing Home Medicine, Municipality of Bergen, Bergen, Norway
| | - Marian Skjellanger
- Department of Nursing Home Medicine, Municipality of Bergen, Bergen, Norway
| | - Kjell Krüger
- Department of Nursing Home Medicine, Municipality of Bergen, Bergen, Norway
| |
Collapse
|
144
|
Belete MA, Anley DT, Tsega SS, Moges N, Anteneh RM, Zemene MA, Gebeyehu AA, Dessie AM, Kebede N, Chanie ES, Alemayehu E. The potential of circulating microRNAs as novel diagnostic biomarkers of COVID-19: a systematic review and meta-analysis. BMC Infect Dis 2024; 24:1011. [PMID: 39300343 PMCID: PMC11414062 DOI: 10.1186/s12879-024-09915-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024] Open
Abstract
INTRODUCTION The COVID-19 pandemic has caused an unprecedented health threat globally, necessitating innovative and efficient diagnostic approaches for timely identification of infected individuals. Despite few emerging reports, the clinical utility of circulating microRNAs (miRNAs) in early and accurate diagnosis of COVID-19 is not well-evidenced. Hence, this meta-analysis aimed to explore the diagnostic potential of circulating miRNAs for COVID-19. The protocol for this study was officially recorded on PROSPERO under registration number CRD42023494959. METHODS Electronic databases including Embase, PubMed, Scopus, and other sources were exhaustively searched to recover studies published until 16th January, 2024. Pooled specificity, sensitivity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic ratio (DOR), positive predictive value (PPV), negative predictive value (NPV), and area under the curve (AUC) were computed from the metadata using Stata 14.0 software. Risk of bias appraisal of included articles was carried out using Review Manager (Rev-Man) 5.3 package through the modified QUADAS-2 tool. Subgroup, heterogeneity, meta-regression and sensitivity analyses were undertaken. Publication bias and clinical applicability were also evaluated via Deeks' funnel plot and Fagan nomogram (scattergram), respectively. RESULT A total of 43 studies from 13 eligible articles, involving 5175 participants (3281 COVID-19 patients and 1894 healthy controls), were analyzed. Our results depicted that miRNAs exhibit enhanced pooled specificity 0.91 (95% CI: 0.88-0.94), sensitivity 0.94 (95% CI: 0.91-0.96), DOR of 159 (95% CI: 87-288), and AUC values of 0.97 (95% CI: 0.95-0.98) with high pooled PPV 96% (95% CI: 94-97%) and NPV 88% (95% CI: 86-90%) values. Additionally, highest diagnostic capacity was observed in studies involving larger sample size (greater than 100) and those involving the African population, demonstrating consistent diagnostic effectiveness across various specimen types. Notably, a total of 12 distinct miRNAs were identified as suitable for both exclusion and confirmation of COVID-19 cases, denoting their potential clinical applicability. CONCLUSION Our study depicted that miRNAs show significantly high diagnostic accuracy in differentiating COVID-19 patients from healthy counterparts, suggesting their possible use as viable biomarkers. Nonetheless, thorough and wide-ranging longitudinal researches are necessary to confirm the clinical applicability of miRNAs in diagnosing COVID-19.
Collapse
Affiliation(s)
- Melaku Ashagrie Belete
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia.
| | - Denekew Tenaw Anley
- Department of Public Health, College of Health Science, Debre Tabor University, Debre Tabor, Ethiopia
| | - Sintayehu Simie Tsega
- Department of Medical Nursing, School of Nursing, College of Medicine and Health Science, University of Gondar, Gondar, Ethiopia
| | - Natnael Moges
- Department of Pediatrics and Child Health Nursing, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Rahel Mulatie Anteneh
- Department of Public Health, College of Health Science, Debre Tabor University, Debre Tabor, Ethiopia
| | - Melkamu Aderajew Zemene
- Department of Public Health, College of Health Science, Debre Tabor University, Debre Tabor, Ethiopia
| | - Asaye Alamneh Gebeyehu
- Department of Public Health, College of Health Science, Debre Tabor University, Debre Tabor, Ethiopia
| | - Anteneh Mengist Dessie
- Department of Public Health, College of Health Science, Debre Tabor University, Debre Tabor, Ethiopia
| | - Natnael Kebede
- Department of Health Promotion, School of Public Health College of Medicine Health Sciences, Wollo University, Dessie, Ethiopia
| | - Ermias Sisay Chanie
- Department of Pediatrics and Child Health Nursing, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Ermiyas Alemayehu
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| |
Collapse
|
145
|
Seaman WT, Keener O, Nanayakkara D, Mollan KR, Premkumar L, Cuadra EC, Jones CD, Pettifor A, Bowman NM, Wang F, Webster-Cyriaque J. Oral SARS-CoV-2 host responses predict the early COVID-19 disease course. Sci Rep 2024; 14:21788. [PMID: 39294156 PMCID: PMC11411107 DOI: 10.1038/s41598-024-67504-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 07/11/2024] [Indexed: 09/20/2024] Open
Abstract
Oral fluids provide ready detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and host responses. This study sought to evaluate relationships between oral virus, oral and systemic anti-SARS-CoV-2-specific antibodies, and symptoms. Oral fluids (saliva/throat wash (saliva/TW)) and serum were collected from asymptomatic and symptomatic, nasopharyngeal (NP) SARS-CoV-2 RT-qPCR+ human participants (n = 45). SARS-CoV-2 RT-qPCR and N-antigen detection by immunoblot and lateral flow assay (LFA) were performed. RT-qPCR for subgenomic RNA (sgRNA) was sequence confirmed. SARS-CoV-2-anti-S protein RBD LFA and ELISA assessed IgM and IgG responses. Structural analysis identified host salivary molecules analogous to SARS-CoV-2-N-antigen. At time of enrollment (baseline, BL), LFA-detected N-antigen in 86% of TW and was immunoblot-confirmed. However, only 3/17 were saliva/TW qPCR+ . Sixty percent of saliva and 83% of TW demonstrated persistent N-antigen at 4 weeks. N-antigen LFA signal in three anti-spike sero-negative participants suggested potential cross-detection of 4 structurally analogous salivary RNA binding proteins (alignment 19-29aa, RMSD 1-1.5 Angstroms). At enrollment, symptomatic participants demonstrated replication-associated sgRNA junctions, were IgG+ (94%/100% in saliva/TW), and IgM+ (63%/54%). At 4 weeks, SARS-CoV-2 IgG (100%/83%) and IgM (80%/67%) persisted. Oral and serum IgG correlated 100% with NP+ PCR status. Cough and fatigue severity (p = 0.010 and 0.018 respectively), and presence of weakness, nausea, and composite upper respiratory symptoms (p = 0.037, 0.005, and 0.017, respectively) were negatively associated with saliva IgM but not TW or serum IgM. Throat wash IgM levels were higher in women compared to men, although the association did not reach statistical significance (median: 290 (female) versus 0.697, p = 0.056). Important to transmission and disease course, oral viral replication and persistence showed clear relationships with select symptoms and early oral IgM responses during early infection. N-antigen cross-reactivity may reflect mimicry of structurally analogous host proteins.
Collapse
Affiliation(s)
- William T Seaman
- National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
- Division of Oral and Craniofacial Health Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Olive Keener
- Division of Oral and Craniofacial Health Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- North Carolina School of Math and Science, Durham, NC, USA
| | - Dinelka Nanayakkara
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Katie R Mollan
- UNC School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Rd, Medical Biomolecular Research Building, Room 2341b, Chapel Hill, NC, 27599, USA
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lakshmanane Premkumar
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Edwing Centeno Cuadra
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Corbin D Jones
- UNC School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Rd, Medical Biomolecular Research Building, Room 2341b, Chapel Hill, NC, 27599, USA
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Audrey Pettifor
- UNC School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Rd, Medical Biomolecular Research Building, Room 2341b, Chapel Hill, NC, 27599, USA
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Natalie M Bowman
- UNC School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Rd, Medical Biomolecular Research Building, Room 2341b, Chapel Hill, NC, 27599, USA
| | | | - Jennifer Webster-Cyriaque
- National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA.
- Division of Oral and Craniofacial Health Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- UNC School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Rd, Medical Biomolecular Research Building, Room 2341b, Chapel Hill, NC, 27599, USA.
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
146
|
Muraoka D, Moi ML, Muto O, Nakatsukasa T, Deng S, Takashima C, Yamaguchi R, Sawada SI, Hayakawa H, Nguyen TTN, Haseda Y, Soga T, Matsushita H, Ikeda H, Akiyoshi K, Harada N. Low-frequency CD8 + T cells induced by SIGN-R1 + macrophage-targeted vaccine confer SARS-CoV-2 clearance in mice. NPJ Vaccines 2024; 9:173. [PMID: 39294173 PMCID: PMC11411095 DOI: 10.1038/s41541-024-00961-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/01/2024] [Indexed: 09/20/2024] Open
Abstract
Vaccine-induced T cells and neutralizing antibodies are essential for protection against SARS-CoV-2. Previously, we demonstrated that an antigen delivery system, pullulan nanogel (PNG), delivers vaccine antigen to lymph node medullary macrophages and thereby enhances the induction of specific CD8+ T cells. In this study, we revealed that medullary macrophage-selective delivery by PNG depends on its binding to a C-type lectin SIGN-R1. In a K18-hACE2 mouse model of SARS-CoV-2 infection, vaccination with a PNG-encapsulated receptor-binding domain of spike protein decreased the viral load and prolonged the survival in the CD8+ T cell- and B cell-dependent manners. T cell receptor repertoire analysis revealed that although the vaccine induced T cells at various frequencies, low-frequency specific T cells mainly promoted virus clearance. Thus, the induction of specific CD8+ T cells that respond quickly to viral infection, even at low frequencies, is important for vaccine efficacy and can be achieved by SIGN-R1+ medullary macrophage-targeted antigen delivery.
Collapse
Affiliation(s)
- Daisuke Muraoka
- Department of Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan.
| | - Meng Ling Moi
- School of International Health, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan.
- Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan.
| | - Osamu Muto
- Division of Cancer Systems Biology, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Cancer Informatics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takaaki Nakatsukasa
- Department of Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Situo Deng
- Department of Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Chieko Takashima
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Rui Yamaguchi
- Division of Cancer Systems Biology, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Cancer Informatics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shin-Ichi Sawada
- Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba University, Chiba, Japan
| | - Haruka Hayakawa
- School of International Health, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | | | | | | | - Hirokazu Matsushita
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Hiroaki Ikeda
- Department of Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kazunari Akiyoshi
- Department of Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | |
Collapse
|
147
|
Park HS, Matsuoka Y, Santos C, Luongo C, Liu X, Yang L, Kaiser JA, Duncan EF, Johnson RF, Teng IT, Kwong PD, Buchholz UJ, Le Nouën C. Intranasal parainfluenza virus-vectored vaccine expressing SARS-CoV-2 spike protein of Delta or Omicron B.1.1.529 induces mucosal and systemic immunity and protects hamsters against homologous and heterologous challenge. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.612598. [PMID: 39372768 PMCID: PMC11451599 DOI: 10.1101/2024.09.12.612598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
The continuous emergence of new SARS-CoV-2 variants requires that COVID vaccines be updated to match circulating strains. We generated B/HPIV3-vectored vaccines expressing 6P-stabilized S protein of the ancestral, B.1.617.2/Delta, or B.1.1.529/Omicron variants as pediatric vaccines for intranasal immunization against HPIV3 and SARS-CoV-2 and characterized these in hamsters. Following intranasal immunization, these B/HPIV3 vectors replicated in the upper and lower respiratory tract and induced mucosal and serum anti-S IgA and IgG. B/HPIV3 expressing ancestral or B.1.617.2/Delta-derived S-6P induced serum antibodies that effectively neutralized SARS-CoV-2 of the ancestral and B.1.617.2/Delta lineages, while the cross-neutralizing potency of B.1.1.529/Omicron S-induced antibodies was lower. Despite the lower cross-neutralizing titers induced by B/HPIV3 expressing S-6P from B.1.1.529/Omicron, a single intranasal dose of all three versions of B/HPIV3 vectors was protective against matched or heterologous WA1/2020, B.1.617.2/Delta or BA.1 (B.1.1.529.1)/Omicron challenge; hamsters were protected from challenge virus replication in the lungs, while low levels of challenge virus were detectable in the upper respiratory tract of a small number of animals. Immunization also protected against lung inflammatory response after challenge, with mild inflammatory cytokine induction associated with the slightly lower level of cross-protection of WA1/2020 and B.1.617.2/Delta variants against the BA.1/Omicron variant. Serum antibodies elicited by all vaccine candidates were broadly reactive against 20 antigenic variants, but the antigenic breadth of antibodies elicited by B/HPIV3-expressed S-6P from the ancestral or B.1.617.2/Delta variant exceeded that of the S-6P B.1.1.529/Omicron expressing vector. These results will guide development of intranasal B/HPIV3 vectors with S antigens matching circulating SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Hong-Su Park
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yumiko Matsuoka
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Celia Santos
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cindy Luongo
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xueqiao Liu
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lijuan Yang
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jaclyn A. Kaiser
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eleanor F. Duncan
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Reed F. Johnson
- SARS-CoV-2 Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - I-Ting Teng
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Peter D. Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ursula J. Buchholz
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cyril Le Nouën
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
148
|
Chhetri SP, Bhandari VS, Maharjan R, Lamichhane TR. Identification of lead inhibitors for 3CLpro of SARS-CoV-2 target using machine learning based virtual screening, ADMET analysis, molecular docking and molecular dynamics simulations. RSC Adv 2024; 14:29683-29692. [PMID: 39297030 PMCID: PMC11408992 DOI: 10.1039/d4ra04502e] [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: 06/20/2024] [Accepted: 09/04/2024] [Indexed: 09/21/2024] Open
Abstract
The SARS-CoV-2 3CLpro is a critical target for COVID-19 therapeutics due to its role in viral replication. We employed a screening pipeline to identify novel inhibitors by combining machine learning classification with similarity checks of approved medications. A voting classifier, integrating three machine learning classifiers, was used to filter a large database (∼10 million compounds) for potential inhibitors. This ensemble-based machine learning technique enhances overall performance and robustness compared to individual classifiers. From the screening, three compounds M1, M2 and M3 were selected for further analysis. Absorption, distribution, metabolism, excretion, and toxicity (ADMET) analysis compared these candidates to nirmatrelvir and azvudine. Molecular docking followed by 200 ns MD simulations showed that only M1 (6-[2,4-bis(dimethylamino)-6,8-dihydro-5H-pyrido[3,4-d]pyrimidine-7-carbonyl]-1H-pyrimidine-2,4-dione) remained stable. For azvudine and M1, the estimated median lethal doses are 1000 and 550 mg kg-1, respectively, with maximum tolerated doses of 0.289 and 0.614 log mg per kg per day. The predicted inhibitory activity of M1 is 7.35, similar to that of nirmatrelvir. The binding free energy based on Molecular Mechanics Poisson-Boltzmann Surface Area (MM-PBSA) of M1 is -18.86 ± 4.38 kcal mol-1, indicating strong binding interactions. These findings suggest that M1 merits further investigation as a potential SARS-CoV-2 treatment.
Collapse
Affiliation(s)
| | | | - Rajesh Maharjan
- Central Department of Physics, Tribhuvan University Kathmandu 44600 Nepal
| | | |
Collapse
|
149
|
Kotov IA, Agletdinov MR, Roev GV, Pimkina EV, Nadtoka MI, Peresadina AV, Bukharina AY, Svetlichny DV, Goncharov SE, Vykhodtseva AV, Borisova NI, Lysenkov VG, Chanyshev MD, Agabalaev DNO, Saenko VV, Cherkashina AS, Semenenko TA, Dubodelov DV, Khafizov KF, Akimkin VG. Genomic surveillance of SARS-CoV-2 in Russia: insights from the VGARus platform. JOURNAL OF MICROBIOLOGY, EPIDEMIOLOGY AND IMMUNOBIOLOGY 2024; 101:435-447. [DOI: 10.36233/0372-9311-554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
Introduction. In response to the COVID-19 pandemic in the Russian Federation, comprehensive response measures were taken. One of these measures was the development of a viral genome aggregation platform (VGARus) to monitor virus variability.
The aim of this paper is to describe the role of the VGARus platform in tracking genetic variation in SARS-CoV-2.
Materials and methods. VGARus utilizes sequencing data and bioinformatics tools to monitor genetic variations in SARS-CoV-2. The viral genomes were aligned using NextClade, which also translated them into amino acids and identified mutations. The viral variability over time was analyzed by counting the number of amino acid changes compared to the reference sequence.
Results. The analysis of data within VGARus enabled the identification of new virus variants, contributing to improved diagnostic tests and vaccine development. The platform allowed for the prediction of epidemiologic trends, facilitating a rapid response to changes in the epidemiologic situation. For example, using VGARus, an increase in COVID-19 incidence was accurately predicted in the summer of 2022 and early 2023, which were associated with the emergence of Omicron subvariants BA.5 and XBB. Data from the platform helps validate the effectiveness of primers and DNA probes to ensure high diagnostic accuracy and reduce the risk of false negatives.
Conclusion. VGARus demonstrates the growing role of genomic surveillance in combating COVID-19 and improving preparedness for future infectious disease outbreaks. The platform is a powerful tool for generating evidence-based solutions to combat a pandemic and mitigate its health, economic and societal impacts. It provides the ability to promptly obtain information on the epidemiologic situation in a particular region of the Russian Federation, use genomic data for phylogenetic analysis, compare the mutational spectrum of SARS-CoV-2 sequences with foreign samples. VGARus data allow for both retrospective analysis and predictive hypotheses. For example, we can clearly see the dynamics of the change of different virus variants: sequences belonging to the Alpha, Beta, Delta, Omicron lineages and many less common ones, clearly form the upsurges of morbidity, the interaction of which is reflected in the epidemiological picture. It is also currently being expanded to monitor other pathogens, increasing its public health relevance.
Collapse
|
150
|
Veugen JMJ, Schoenmakers T, van Loo IHM, Haagmans BL, Leers MPG, Lamers MM, Lucchesi M, van Bussel BCT, van Mook WNKA, Nuijts RMMA, Savelkoul PHM, Dickman MM, Wolffs PFG. Advancing COVID-19 diagnostics: rapid detection of intact SARS-CoV-2 using viability RT-PCR assay. Microbiol Spectr 2024; 12:e0016024. [PMID: 39037224 PMCID: PMC11370235 DOI: 10.1128/spectrum.00160-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/10/2024] [Indexed: 07/23/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19). Commonly used methods for both clinical diagnosis of SARS-CoV-2 infection and management of infected patients involve the detection of viral RNA, but the presence of infectious virus particles is unknown. Viability PCR (v-PCR) uses a photoreactive dye to bind non-infectious RNA, ideally resulting in the detection of RNA only from intact virions. This study aimed to develop and validate a rapid v-PCR assay for distinguishing intact and compromised SARS-CoV-2. Propidium monoazide (PMAxx) was used as a photoreactive dye. Mixtures with decreasing percentages of intact SARS-CoV-2 (from 100% to 0%) were prepared from SARS-CoV-2 virus stock and a clinical sample. Each sample was divided into a PMAxx-treated part and a non-PMAxx-treated part. Reverse transcription-PCR (RT-PCR) using an in-house developed SARS-CoV-2 viability assay was then applied to both sample sets. The difference in intact SARS-CoV-2 was determined by subtracting the cycle threshold (Ct) value of the PMAxx-treated sample from the non-PMAxx-treated sample. Mixtures with decreasing concentrations of intact SARS-CoV-2 showed increasingly lower delta Ct values as the percentage of intact SARS-CoV-2 decreased, as expected. This relationship was observed in both high and low viral load samples prepared from cultured SARS-CoV-2 virus stock, as well as for a clinical sample prepared directly from a SARS-CoV-2 positive nasopharyngeal swab. In this study, a rapid v-PCR assay has been validated that can distinguish intact from compromised SARS-CoV-2. The presence of intact virus particles, as determined by v-PCR, may indicate SARS-CoV-2 infectiousness. IMPORTANCE This study developed a novel method that can help determine whether someone who has been diagnosed with coronavirus disease 2019 (COVID-19) is still capable of spreading the virus to others. Current tests only detect the presence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA, but cannot tell whether the particles are still intact and can thus infect cells. The researchers used a dye that selectively blocks the detection of damaged virions and free RNA. They showed that this viability PCR reliably distinguishes intact SARS-CoV-2 capable of infecting from damaged SARS-CoV-2 or free RNA in both cultured virus samples and a clinical sample. Being able to quickly assess contagiousness has important implications for contact tracing and safely ending isolation precautions. This viability PCR technique provides a simple way to obtain valuable information, beyond just positive or negative test results, about the actual risk someone poses of transmitting SARS-CoV-2 through the air or surfaces they come into contact with.
Collapse
Affiliation(s)
- Judith M. J. Veugen
- University Eye Clinic Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, the Netherlands
- Department of Medical Microbiology, Infectious Diseases & Infection Prevention, Maastricht University Medical Center, Maastricht, the Netherlands
- Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, the Netherlands
| | - Tom Schoenmakers
- Department of Clinical Chemistry and Hematology, Zuyderland Medical Center, Sittard-Geleen/Heerlen, the Netherlands
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
| | - Inge H. M. van Loo
- Department of Medical Microbiology, Infectious Diseases & Infection Prevention, Maastricht University Medical Center, Maastricht, the Netherlands
- Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, the Netherlands
| | - Bart L. Haagmans
- Viroscience Department, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Mathie P. G. Leers
- Department of Clinical Chemistry and Hematology, Zuyderland Medical Center, Sittard-Geleen/Heerlen, the Netherlands
- Faculty of Science, Environmental Sciences, Open Universiteit, Heerlen, the Netherlands
| | - Mart M. Lamers
- Viroscience Department, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Mayk Lucchesi
- Department of Medical Microbiology, Infectious Diseases & Infection Prevention, Maastricht University Medical Center, Maastricht, the Netherlands
- Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, the Netherlands
| | - Bas C. T. van Bussel
- Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, the Netherlands
- Department of Intensive Care Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Walther N. K. A. van Mook
- Department of Intensive Care Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School of Health Professions Education (SHE), Maastricht University, Maastricht, the Netherlands
| | - Rudy M. M. A. Nuijts
- University Eye Clinic Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, the Netherlands
- Department of Ophthalmology, Zuyderland Medical Center, Heerlen, the Netherlands
| | - Paul H. M. Savelkoul
- Department of Medical Microbiology, Infectious Diseases & Infection Prevention, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Mor M. Dickman
- University Eye Clinic Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, the Netherlands
| | - Petra F. G. Wolffs
- Department of Medical Microbiology, Infectious Diseases & Infection Prevention, Maastricht University Medical Center, Maastricht, the Netherlands
- Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, the Netherlands
| |
Collapse
|