201
|
Meller S, Al Khatri MSA, Alhammadi HK, Álvarez G, Alvergnat G, Alves LC, Callewaert C, Caraguel CGB, Carancci P, Chaber AL, Charalambous M, Desquilbet L, Ebbers H, Ebbers J, Grandjean D, Guest C, Guyot H, Hielm-Björkman A, Hopkins A, Kreienbrock L, Logan JG, Lorenzo H, Maia RDCC, Mancilla-Tapia JM, Mardones FO, Mutesa L, Nsanzimana S, Otto CM, Salgado-Caxito M, de los Santos F, da Silva JES, Schalke E, Schoneberg C, Soares AF, Twele F, Vidal-Martínez VM, Zapata A, Zimin-Veselkoff N, Volk HA. Expert considerations and consensus for using dogs to detect human SARS-CoV-2-infections. Front Med (Lausanne) 2022; 9:1015620. [PMID: 36569156 PMCID: PMC9773891 DOI: 10.3389/fmed.2022.1015620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/17/2022] [Indexed: 12/13/2022] Open
Affiliation(s)
- Sebastian Meller
- Department of Small Animal Medicine & Surgery, University of Veterinary Medicine Hannover, Hanover, Germany
| | | | - Hamad Khatir Alhammadi
- International Operations Department, Ministry of Interior of the United Arab Emirates, Abu Dhabi, United Arab Emirates
| | - Guadalupe Álvarez
- Faculty of Veterinary Science, University of Buenos Aires, Buenos Aires, Argentina
| | - Guillaume Alvergnat
- International Operations Department, Ministry of Interior of the United Arab Emirates, Abu Dhabi, United Arab Emirates
| | - Lêucio Câmara Alves
- Department of Veterinary Medicine, Federal Rural University of Pernambuco, Recife, Brazil
| | - Chris Callewaert
- Center for Microbial Ecology and Technology, Department of Biotechnology, Ghent University, Ghent, Belgium
| | - Charles G. B. Caraguel
- School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - Paula Carancci
- Faculty of Veterinary Science, University of Buenos Aires, Buenos Aires, Argentina
| | - Anne-Lise Chaber
- School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - Marios Charalambous
- Department of Small Animal Medicine & Surgery, University of Veterinary Medicine Hannover, Hanover, Germany
| | - Loïc Desquilbet
- École Nationale Vétérinaire d’Alfort, IMRB, Université Paris Est, Maisons-Alfort, France
| | | | | | - Dominique Grandjean
- École Nationale Vétérinaire d’Alfort, Université Paris-Est, Maisons-Alfort, France
| | - Claire Guest
- Medical Detection Dogs, Milton Keynes, United Kingdom
| | - Hugues Guyot
- Clinical Department of Production Animals, Fundamental and Applied Research for Animals & Health Research Unit, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Anna Hielm-Björkman
- Department of Equine and Small Animal Medicine, University of Helsinki, Helsinki, Finland
| | - Amy Hopkins
- Medical Detection Dogs, Milton Keynes, United Kingdom
| | - Lothar Kreienbrock
- Department of Biometry, Epidemiology and Information Processing, University of Veterinary Medicine Hannover, Hanover, Germany
| | - James G. Logan
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Arctech Innovation, The Cube, Dagenham, United Kingdom
| | - Hector Lorenzo
- Faculty of Veterinary Science, University of Buenos Aires, Buenos Aires, Argentina
| | | | | | - Fernando O. Mardones
- Escuela de Medicina Veterinaria, Facultad de Agronomía e Ingeniería Forestal and Facultad de Ciencias Biológicas y Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Leon Mutesa
- Center for Human Genetics, College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda
- Rwanda National Joint Task Force COVID-19, Kigali, Rwanda
| | | | - Cynthia M. Otto
- Penn Vet Working Dog Center, Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Marília Salgado-Caxito
- Escuela de Medicina Veterinaria, Facultad de Agronomía e Ingeniería Forestal and Facultad de Ciencias Biológicas y Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | | - Esther Schalke
- Bundeswehr Medical Service Headquarters, Koblenz, Germany
| | - Clara Schoneberg
- Department of Biometry, Epidemiology and Information Processing, University of Veterinary Medicine Hannover, Hanover, Germany
| | - Anísio Francisco Soares
- Department of Animal Morphology and Physiology, Federal Rural University of Pernambuco, Recife, Brazil
| | - Friederike Twele
- Department of Small Animal Medicine & Surgery, University of Veterinary Medicine Hannover, Hanover, Germany
| | - Victor Manuel Vidal-Martínez
- Laboratorio de Parasitología y Patología Acuática, Departamento de Recursos del Mar, Centro de Investigación y de Estudios Avanzados del IPN Unidad Mérida, Mérida, Yucatán, Mexico
| | - Ariel Zapata
- Faculty of Veterinary Science, University of Buenos Aires, Buenos Aires, Argentina
| | - Natalia Zimin-Veselkoff
- Escuela de Medicina Veterinaria, Facultad de Agronomía e Ingeniería Forestal and Facultad de Ciencias Biológicas y Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Holger A. Volk
- Department of Small Animal Medicine & Surgery, University of Veterinary Medicine Hannover, Hanover, Germany
- Center for Systems Neuroscience Hannover, Hanover, Germany
| |
Collapse
|
202
|
Oh SM, Lee JS, Jo HJ, Kim D, Park D, Hwang YH, Choi Y, Lee CM, Lee S, Chang E, Lee E, Kim TS, Seong MW, Choe PG, Kim NJ. Clinical application of the Panbio™ COVID-19 Ag rapid test device and SSf-COVID19 kit for the detection of SARS-CoV-2 infection. BMC Res Notes 2022; 15:357. [PMID: 36471453 PMCID: PMC9720920 DOI: 10.1186/s13104-022-06226-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 10/06/2022] [Accepted: 10/10/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE We evaluated the sensitivity and specificity of the Panbio™ COVID-19 Ag rapid test device using nasal swabs and those of the SSf-COVID19 kit, one of RT-PCR tests, using saliva specimens. These tests were compared with RT-PCR tests using nasopharyngeal swabs for the diagnosis of SARS-CoV-2 infection. The three diagnostic tests were simultaneously conducted for patients aged ≥ 18 years, who were about to be hospitalized or had been admitted for COVID-19 confirmed by RT-PCR in two research hospitals from August 20 to October 29, 2021. Nasal swabs were tested using the Panbio™ COVID-19 Ag rapid test device. More than 1 mL of saliva was self-collected and tested using the SSf-COVID19 kit. RESULTS In total, 157 patients were investigated; 124 patients who were about to be hospitalized and 33 patients already admitted for COVID-19. The overall sensitivity and specificity of the Panbio™ COVID-19 Ag rapid test device with nasal swabs were 64.7% (95% confidence interval [CI] 47.9-78.5%) and 100.0% (95% CI 97.0-100.0%), respectively. The median time to confirm a positive result was 180 s (interquartile range 60-255 s). The overall sensitivity and specificity of the SSf-COVID19 kit with saliva specimens were 94.1% (95% CI 80.9-98.4%) and 100.0% (95% CI 97.0-100.0%), respectively.
Collapse
Affiliation(s)
- Sang-Min Oh
- grid.31501.360000 0004 0470 5905Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehakro, Jongno-gu, 03080 Seoul, Republic of Korea ,grid.411545.00000 0004 0470 4320Present Address: Department of Internal Medicine, Jeonbuk National University Medical School and Hospital, 20 Geonjiro, Deokjin-gu, 54907 Jeonju, Jeollabuk-do Republic of Korea
| | - Jee-Soo Lee
- grid.31501.360000 0004 0470 5905Department of Laboratory Medicine, Seoul National University College of Medicine, 101 Daehakro, Jongno-gu, 03080 Seoul, Republic of Korea
| | - Hyeon Jae Jo
- grid.31501.360000 0004 0470 5905Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehakro, Jongno-gu, 03080 Seoul, Republic of Korea
| | - Donghwan Kim
- grid.31501.360000 0004 0470 5905Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehakro, Jongno-gu, 03080 Seoul, Republic of Korea
| | - Dohyeon Park
- grid.31501.360000 0004 0470 5905Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehakro, Jongno-gu, 03080 Seoul, Republic of Korea
| | - Young Hoon Hwang
- grid.31501.360000 0004 0470 5905Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehakro, Jongno-gu, 03080 Seoul, Republic of Korea
| | - Yunsang Choi
- grid.31501.360000 0004 0470 5905Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehakro, Jongno-gu, 03080 Seoul, Republic of Korea
| | - Chan Mi Lee
- grid.31501.360000 0004 0470 5905Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehakro, Jongno-gu, 03080 Seoul, Republic of Korea
| | - Seungjae Lee
- grid.31501.360000 0004 0470 5905Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehakro, Jongno-gu, 03080 Seoul, Republic of Korea
| | - Euijin Chang
- grid.31501.360000 0004 0470 5905Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehakro, Jongno-gu, 03080 Seoul, Republic of Korea
| | - Eunyoung Lee
- grid.31501.360000 0004 0470 5905Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehakro, Jongno-gu, 03080 Seoul, Republic of Korea ,grid.412479.dDivision of Infectious Diseases, Seoul Metropolitan Government – Seoul National University Boramae Medical Center, 20 Boramae-ro 5-gil, Dongjak-gu, 07061 Seoul, Republic of Korea
| | - Taek Soo Kim
- grid.31501.360000 0004 0470 5905Department of Laboratory Medicine, Seoul National University College of Medicine, 101 Daehakro, Jongno-gu, 03080 Seoul, Republic of Korea
| | - Moon-Woo Seong
- grid.31501.360000 0004 0470 5905Department of Laboratory Medicine, Seoul National University College of Medicine, 101 Daehakro, Jongno-gu, 03080 Seoul, Republic of Korea
| | - Pyoeng Gyun Choe
- grid.31501.360000 0004 0470 5905Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehakro, Jongno-gu, 03080 Seoul, Republic of Korea
| | - Nam Joong Kim
- grid.31501.360000 0004 0470 5905Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehakro, Jongno-gu, 03080 Seoul, Republic of Korea
| |
Collapse
|
203
|
Venturini E, Giometto S, Tamborino A, Becciolini L, Bosis S, Corsello G, Del Barba P, Garazzino S, Lo Vecchio A, Pugi A, Signa S, Stera G, Trapani S, Castelli Gattinara G, Lucenteforte E, Galli L. Sensitivity of three commercial tests for SARS-CoV-2 serology in children: an Italian multicentre prospective study. Ital J Pediatr 2022; 48:192. [PMID: 36461032 PMCID: PMC9716520 DOI: 10.1186/s13052-022-01381-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 11/09/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND US Food and Drug Administration has issued Emergency Use Authorizations for hundreds of serological assays to support Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) diagnosis. The aim of this study is to evaluate, for the first time in children, the performance of three widely utilized SARS-CoV-2 serology commercial assays, Diesse Diagnostics (IgG, IgA, IgM) and Roche Diagnostics, both Roche Nucleocapsid (N) IgG and Roche Spike (S) IgG assays. METHODS Sensitivity and 95% confidence intervals (CIs) were estimated for each of the three different serological tests and mixed and direct comparison were performed. Univariate and multivariate Poisson regression models were fitted to calculate incidence rate ratios and 95% CIs as estimate of the effects of age, gender, time on the serology title. A p-value < 0.05 indicated statistical significance. RESULTS Overall, 149 children were enrolled in the study. A low sensitivity was found for Diesse IgA, IgM and IgG. Compare to Diesse, Roche S had a higher sensitivity at 15-28 days from infection (0.94, 95%CI: 0.73-1.0) and Roche N at 28-84 days (0.78, 95%CI: 0.58-0.91). When a direct comparison of IgG tests sensitivity was feasible for patients with pairwise information, Roche S and Roche N showed a statistically significant higher sensitivity compared to Diesse in all the study periods, whereas there was no difference between the two Roche tests. CONCLUSION Roche S and Roche N serology tests seem to better perform in children. Large prospective studies are needed to better define the characteristics of those tests.
Collapse
Affiliation(s)
- Elisabetta Venturini
- grid.413181.e0000 0004 1757 8562Infectious Disease Unit, Meyer Children’s Hospital, Florence, Italy
| | - Sabrina Giometto
- grid.5395.a0000 0004 1757 3729Unit of Medical Statistics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Agnese Tamborino
- grid.413181.e0000 0004 1757 8562Infectious Disease Unit, Meyer Children’s Hospital, Florence, Italy
| | - Laura Becciolini
- grid.413181.e0000 0004 1757 8562Clinical Chemistry and Microbiology Laboratory, Meyer Children’s Hospital, Florence, Italy
| | - Samantha Bosis
- grid.414818.00000 0004 1757 8749Pediatric Highly Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giovanni Corsello
- grid.10776.370000 0004 1762 5517Unit of Pediatrics and Neonatal Intensive Therapy, Department of Promotion of Maternal and Infantile and Internal Medicine Health, and Specialist Excellence “G. D’Alessandro”, University of Palermo, Palermo, Italy
| | - Paolo Del Barba
- grid.18887.3e0000000417581884Department of Pediatrics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Garazzino
- grid.7605.40000 0001 2336 6580Paediatric Infectious Diseases Unit, Regina Margherita Children’s Hospital, University of Turin, Turin, Italy
| | - Andrea Lo Vecchio
- grid.4691.a0000 0001 0790 385XSection of Paediatrics, Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Alessandra Pugi
- grid.413181.e0000 0004 1757 8562Clinical Trial Office, Meyer Children’s Hospital, Florence, Italy
| | - Sara Signa
- grid.419504.d0000 0004 1760 0109Infectious Diseases Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Giacomo Stera
- grid.6292.f0000 0004 1757 1758Postgraduate School of Pediatrics, University of Bologna, Bologna, Italy
| | - Sandra Trapani
- grid.8404.80000 0004 1757 2304Department of Health Sciences, University of Florence, Florence, Italy
| | - Guido Castelli Gattinara
- grid.414603.4Vaccination Unit, University Hospital Paediatric Department, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Ersilia Lucenteforte
- grid.5395.a0000 0004 1757 3729Unit of Medical Statistics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Luisa Galli
- grid.413181.e0000 0004 1757 8562Infectious Disease Unit, Meyer Children’s Hospital, Florence, Italy ,grid.8404.80000 0004 1757 2304Department of Health Sciences, University of Florence, Florence, Italy
| |
Collapse
|
204
|
Zahran AM, Abdel-Rahim MH, Nasif KA, Hussein S, Hafez R, Ahmad AB, Saad K, Elhoufey A, Hussein HAM, Thabet AA, El-Badawy O. Association of follicular helper T and follicular regulatory T cells with severity and hyperglycemia in hospitalized COVID-19 patients. Virulence 2022; 13:569-577. [PMID: 35286241 PMCID: PMC8928811 DOI: 10.1080/21505594.2022.2047506] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 01/23/2022] [Accepted: 02/25/2022] [Indexed: 02/08/2023] Open
Abstract
We aimed to determine the levels of follicular helper T (Tfh) and follicular regulatory T (Tfr) cells in COVID-19 patients and determine whether their levels correlated with disease severity and presence of hyperglycemia. This study was carried out in 34 hospitalized COVID-19 patients and 20 healthy controls. Levels of total circulating Tfh, inducible T-cell costimulator (ICOS)+ activated Tfh, and Tfr cells were assessed in all participants by flow cytometry. Total CD4+CXCR5+ Tfh cells and ICOS+Foxp3-activated Tfh cells increased and ICOS+Foxp3+ Tfr cells decreased in COVID-19 patients, especially in diabetic patients and those with severe disease. Activated ICOS+ Tfh cells were directly correlated with lactate dehydrogenase, D-dimer, ferritin, and respiratory rate and inversely correlated with the partial pressure of carbon dioxide. COVID-19 is associated with marked activation of Tfh cells and a profound drop in Tfr cells, especially in severe and diabetic patients. Future studies on expanded cohorts of patients are needed to clarify the relationship between SARS-CoV-2 and acute-onset diabetes.
Collapse
Affiliation(s)
- Asmaa M. Zahran
- Department of Clinical Pathology, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Mona H. Abdel-Rahim
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Khalid A. Nasif
- Department of Medical Biochemistry, Faculty of Medicine, Minia University, Minia, Egypt
- Department Clinical Biochemistry, College of Medicine, King Khalid University,Abha, Saudi Arabia
| | - Safinaz Hussein
- Department of Internal Medicine, Clinical Hematology Unit, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Rania Hafez
- Department of Internal Medicine, Clinical Hematology Unit, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ahmad Bahieldeen Ahmad
- Department of Internal Medicine, Critical Care Unit, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Khaled Saad
- Department of Pediatrics, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Amira Elhoufey
- Department of Community Health Nursing, Faculty of Nursing, Assiut University, Assiut, Egypt
- Department of Community Health Nursing, Alddrab University College, Jazan University, Jazan, Saudi Arabia
| | - Hosni A. M. Hussein
- Department of Microbiology, Faculty of Science, Al Azhar University, Assiut 71524, Egypt
| | - Ali A. Thabet
- Department of Zoology, Faculty of Science, Al Azhar University, Assiut 71524, Egypt
| | - Omnia El-Badawy
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
205
|
Essa RZ, Wu YS, Batumalaie K, Sekar M, Poh CL. Antiviral peptides against SARS-CoV-2: therapeutic targets, mechanistic antiviral activity, and efficient delivery. Pharmacol Rep 2022; 74:1166-1181. [PMID: 36401119 PMCID: PMC9676828 DOI: 10.1007/s43440-022-00432-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/28/2022] [Accepted: 10/31/2022] [Indexed: 11/19/2022]
Abstract
The global pandemic of COVID-19 is a serious public health concern. Over 625 million confirmed cases and more than 6 million deaths have been recorded worldwide. Although several vaccines and antiviral medications have been developed, their efficacy is limited by the emerging new SARS-CoV-2 strains. Peptide-based therapeutics is a fast-growing class of new drugs and have unique advantages over large proteins and small molecules. Antiviral peptides (AVPs) are short polycationic antivirals with broad-spectrum effects, which have been shown to exert both prophylactic and therapeutic actions against reported coronaviruses. The potential therapeutic targets of AVPs are located either on the virus (e.g., E-protein and S-protein) to prohibit viral binding or host cells, particularly, those present on the cell surface (e.g., ACE2 and TMPRSS2). Despite AVPs having promising antiviral effects, their efficacy is limited by low bioavailability. Thus, nanoformulation is a prerequisite for prolonged bioavailability and efficient delivery. This review aimed to present an insight into the therapeutic AVP targets on both virus and host cells by discussing their antiviral activities and associated molecular mechanisms. Besides, it described the technique for discovering and developing possible AVPs based on their targets, as well as the significance of using nanotechnology for their efficient delivery against SARS-CoV-2.
Collapse
Affiliation(s)
- Raahilah Zahir Essa
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, 47500 Selangor, Malaysia
| | - Yuan-seng Wu
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, 47500 Selangor, Malaysia
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, 47500 Selangor, Malaysia
| | - Kalaivani Batumalaie
- Department of Biomedical Sciences, Faculty of Health Sciences, Asia Metropolitan University, 81750 Johor, Malaysia
| | - Mahendran Sekar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur, 30450 Ipoh, Perak Malaysia
| | - Chit-laa Poh
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, 47500 Selangor, Malaysia
| |
Collapse
|
206
|
Zhao H, Lu L, Peng Z, Chen LL, Meng X, Zhang C, Ip JD, Chan WM, Chu AWH, Chan KH, Jin DY, Chen H, Yuen KY, To KKW. SARS-CoV-2 Omicron variant shows less efficient replication and fusion activity when compared with Delta variant in TMPRSS2-expressed cells. Emerg Microbes Infect 2022; 11:277-283. [PMID: 34951565 PMCID: PMC8774049 DOI: 10.1080/22221751.2021.2023329] [Citation(s) in RCA: 284] [Impact Index Per Article: 94.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 12/22/2021] [Indexed: 02/07/2023]
Abstract
The novel SARS-CoV-2 Omicron variant (B.1.1.529), first found in early November 2021, has sparked considerable global concern and it has >50 mutations, many of which are known to affect transmissibility or cause immune escape. In this study, we sought to investigate the virological characteristics of the Omicron variant and compared it with the Delta variant which has dominated the world since mid-2021. Omicron variant replicated more slowly than the Delta variant in transmembrane serine protease 2 (TMPRSS2)-overexpressing VeroE6 (VeroE6/TMPRSS2) cells. Notably, the Delta variant replicated well in Calu3 cell line which has robust TMPRSS2 expression, while the Omicron variant replicated poorly in this cell line. Competition assay showed that Delta variant outcompeted Omicron variant in VeroE6/TMPRSS2 and Calu3 cells. To confirm the difference in entry pathway between the Omicron and Delta variants, we assessed the antiviral effect of bafilomycin A1, chloroquine (inhibiting endocytic pathway), and camostat (inhibiting TMPRSS2 pathway). Camostat potently inhibited the Delta variant but not the Omicron variant, while bafilomycin A1 and chloroquine could inhibit both Omicron and Delta variants. Moreover, the Omicron variant also showed weaker cell-cell fusion activity when compared with Delta variant in VeroE6/TMPRSS2 cells. Collectively, our results suggest that Omicron variant infection is not enhanced by TMPRSS2 but is largely mediated via the endocytic pathway. The difference in entry pathway between Omicron and Delta variants may have an implication on the clinical manifestations or disease severity.
Collapse
Affiliation(s)
- Hanjun Zhao
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Lu Lu
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Zheng Peng
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Lin-Lei Chen
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Xinjin Meng
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Chuyuan Zhang
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Jonathan Daniel Ip
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Wan-Mui Chan
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Allen Wing-Ho Chu
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Kwok-Hung Chan
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Dong-Yan Jin
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Honglin Chen
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Kwok-Yung Yuen
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Queen Mary Hospital, Hong Kong Special Administrative Region, People’s Republic of China
| | - Kelvin Kai-Wang To
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Queen Mary Hospital, Hong Kong Special Administrative Region, People’s Republic of China
| |
Collapse
|
207
|
Wang Z, Liang Z, Wei R, Wang H, Cheng F, Liu Y, Meng S. Quantitative determination of the electron beam radiation dose for SARS-CoV-2 inactivation to decontaminate frozen food packaging. Virol Sin 2022; 37:823-830. [PMID: 36309306 PMCID: PMC9605788 DOI: 10.1016/j.virs.2022.10.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/21/2022] [Indexed: 11/05/2022] Open
Abstract
The spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) from cold-chain foods to frontline workers poses a serious public health threat during the current global pandemic. There is an urgent need to design concise approaches for effective virus inactivation under different physicochemical conditions to reduce the risk of contagion through viral contaminated surfaces of cold-chain foods. By employing a time course of electron beam exposure to a high titer of SARS-CoV-2 at cold-chain temperatures, a radiation dose of 2 kGy was demonstrated to reduce the viral titer from 104.5 to 0 median tissue culture infectious dose (TCID50)/mL. Next, using human coronavirus OC43 (HCoV-OC43) as a suitable SARS-CoV-2 surrogate, 3 kGy of high-energy electron radiation was defined as the inactivation dose for a titer reduction of more than 4 log units on tested packaging materials. Furthermore, quantitative reverse transcription PCR (RT-qPCR) was used to test three viral genes, namely, E, N, and ORF1ab. There was a strong correlation between TCID50 and RT-qPCR for SARS-CoV-2 detection. However, RT-qPCR could not differentiate between the infectivity of the radiation-inactivated and nonirradiated control viruses. As the defined radiation dose for effective viral inactivation fell far below the upper safe dose limit for food processing, our results provide a basis for designing radiation-based approaches for the decontamination of SARS-CoV-2 in frozen food products. We further demonstrate that cell-based virus assays are essential to evaluate the SARS-CoV-2 inactivation efficiency for the decontaminating strategies.
Collapse
Affiliation(s)
- Zihao Wang
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhentao Liang
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Rongguo Wei
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China,University of Chinese Academy of Sciences, Beijing, 100049, China,Department of Clinical Laboratory, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, 530022, China
| | - Hongwei Wang
- China Isotope and Radiaton Corporation, Beijing, 100089, China
| | - Fang Cheng
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yang Liu
- Changchun CNNC CIRC Radiation Technology Co., LTD, Changchun, 130022, China
| | - Songdong Meng
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China,Corresponding author
| |
Collapse
|
208
|
Cardoso AR, Alves JF, Frasco MF, Piloto AM, Serrano V, Mateus D, Sebastião AI, Matos AM, Carmo A, Cruz T, Fortunato E, Sales MGF. An ultra-sensitive electrochemical biosensor using the Spike protein for capturing antibodies against SARS-CoV-2 in point-of-care. Mater Today Bio 2022; 16:100354. [PMID: 35847374 PMCID: PMC9270181 DOI: 10.1016/j.mtbio.2022.100354] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/02/2022] [Accepted: 07/05/2022] [Indexed: 11/30/2022] Open
Abstract
This work presents an innovative ultra-sensitive biosensor having the Spike protein on carbon-based screen-printed electrodes (SPEs), for monitoring in point-of-care antibodies against SARS-CoV-2, a very important tool for epidemiological monitoring of COVID-19 infection and establishing vaccination schemes. In an innovative and simple approach, a highly conductive support is combined with the direct adsorption of Spike protein to enable an extensive antibody capture. The high conductivity was ensured by using carboxylated carbon nanotubes on the carbon electrode, by means of a simple and quick approach, which also increased the surface area. These were then modified with EDC/NHS chemistry to produce an amine layer and undergo Spike protein adsorption, to generate a stable layer capable of capturing the antibodies against SARS-CoV-2 in serum with great sensitivity. Electrochemical impedance spectroscopy was used to evaluate the analytical performance of this biosensor in serum. It displayed a linear response between 1.0 pg/mL and 10 ng/mL, with a detection limit of ∼0.7 pg/mL. The analysis of human positive sera containing antibody in a wide range of concentrations yielded accurate data, correlating well with the reference method. It also offered the unique ability of discriminating antibody concentrations in sera below 2.3 μg/mL, the lowest value detected by the commercial method. In addition, a proof-of-concept study was performed by labelling anti-IgG antibodies with quantum dots to explore a new electrochemical readout based on the signal generated upon binding to the anti-S protein antibodies recognised on the surface of the biosensor. Overall, the alternative serologic assay presented is a promising tool for assessing protective immunity to SARS-CoV-2 and a potential guide for revaccination. An ultra-sensitive biosensor for detection of low levels of antibodies against SARS-CoV-2. Highly conductive substrate with adsorbed protein S and point-of-care capability. Application to human sera samples and good correlation with commercial method. Electrochemical impedance readings with an iron-based redox probe. Alternative electrochemical impedance readings with anti-IgG labelled with quantum dots.
Collapse
Affiliation(s)
- Ana R Cardoso
- BioMark@UC/CEB - LABBELS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal.,BioMark@ISEP/ CEB - LABBELS, School of Engineering, Polytechnic Institute of Porto, Porto, Portugal.,CENIMAT
- i3N, Department of Materials Science, School of Science and Technology, NOVA University of Lisbon and CEMOP/UNINOVA, Campus de Caparica, 2829-516, Caparica, Portugal
| | - João Frederico Alves
- BioMark@UC/CEB - LABBELS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - Manuela F Frasco
- BioMark@UC/CEB - LABBELS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - Ana Margarida Piloto
- BioMark@ISEP/ CEB - LABBELS, School of Engineering, Polytechnic Institute of Porto, Porto, Portugal
| | - Verónica Serrano
- BioMark@UC/CEB - LABBELS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - Daniela Mateus
- BioMark@UC/CEB - LABBELS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Ana Isabel Sebastião
- Center for Neuroscience and Cell Biology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Ana Miguel Matos
- Chemical Engineering Processes and Forest Products Research Center, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Anália Carmo
- Clinical Pathology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Teresa Cruz
- Center for Neuroscience and Cell Biology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Elvira Fortunato
- CENIMAT
- i3N, Department of Materials Science, School of Science and Technology, NOVA University of Lisbon and CEMOP/UNINOVA, Campus de Caparica, 2829-516, Caparica, Portugal
| | - M Goreti F Sales
- BioMark@UC/CEB - LABBELS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal.,BioMark@ISEP/ CEB - LABBELS, School of Engineering, Polytechnic Institute of Porto, Porto, Portugal
| |
Collapse
|
209
|
Moffat MA, Dessie AS, O’Leary K, Lumba R, Rhee DS. Short-term outcomes of infants born to mothers with SARS-CoV-2 infection. J Matern Fetal Neonatal Med 2022; 35:8192-8198. [PMID: 34470140 PMCID: PMC8425428 DOI: 10.1080/14767058.2021.1966412] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 08/06/2021] [Indexed: 12/23/2022]
Abstract
OBJECTIVE The rate of transmission of SARS-CoV-2 from mothers to infants in the peri- and post-natal period remains an area of ongoing investigation. This study aims to determine rates of development of clinically significant COVID-19 disease within 1 month among infants born to symptomatic and asymptomatic SARS-CoV-2 positive mothers. MATERIALS AND METHODS This was a single-center, retrospective cohort study of all infants born to SARS-CoV-2 positive mothers who were admitted to the Well Baby Nursery (WBN) at New York University Langone Hospital-Brooklyn from 23 March-23 September 2020. Infants born to asymptomatic mothers were allowed to room-in, while infants born to mothers with symptoms of SARS-CoV-2 were isolated and discharged home to an alternate primary caregiver. A phone follow-up program contacted mothers at 2 weeks and 1 month post discharge to inquire about newborn symptoms, maternal symptoms, personal protective equipment (PPE) usage, and any presentations to care. Medical records were also reviewed for clinic and hospital visits to determine if exposed infants developed any symptoms following discharge. RESULTS Of 1903 deliveries during the study period, 131 mothers (21 symptomatic, 110 asymptomatic) tested positive for SARS-CoV-2 and had infants admitted to the WBN. 57 infants (21 born to symptomatic mothers, 36 born to asymptomatic mothers) were tested prior to discharge, and none were positive. 121 of 133 infants had at least 1 follow up call in the study period. Of these, 31 had symptoms potentially concerning for SARS-CoV-2 infection or Multisystem Inflammatory Syndrome in Children, and 19 presented to medical care for these symptoms. 4 infants had SARS- CoV-2 testing after discharge, and none were positive. 2 infants were admitted to the hospital for fever but neither had a positive SARS-CoV-2 result. 65% of mothers reported always adhering to PPE recommendations. CONCLUSION Our results suggest that infants born both to symptomatic and asymptomatic mothers are unlikely to develop clinically significant COVID-19 disease in the peri- and post-natal periods.
Collapse
Affiliation(s)
- Michael A. Moffat
- Department of Pediatrics, New York University Grossman School of Medicine, New York, NY, USA
| | - Almaz S. Dessie
- Department of Emergency Medicine, Division of Pediatric Emergency Medicine, Columbia University Vagelos College of Physicians & Surgeons, New York, NY, USA
| | - Kathryn O’Leary
- Department of Pediatrics, New York University Grossman School of Medicine, New York, NY, USA
| | - Rishi Lumba
- Department of Pediatrics, New York University Grossman School of Medicine, New York, NY, USA
| | - David S. Rhee
- Department of Pediatrics, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
210
|
Johnston M, Ceren Ates H, Glatz RT, Mohsenin H, Schmachtenberg R, Göppert N, Huzly D, Urban GA, Weber W, Dincer C. Multiplexed biosensor for point-of-care COVID-19 monitoring: CRISPR-powered unamplified RNA diagnostics and protein-based therapeutic drug management. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2022; 61:129-138. [PMID: 36405570 PMCID: PMC9643339 DOI: 10.1016/j.mattod.2022.11.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 10/24/2022] [Accepted: 11/02/2022] [Indexed: 06/16/2023]
Abstract
In late 2019 SARS-CoV-2 rapidly spread to become a global pandemic, therefore, measures to attenuate chains of infection, such as high-throughput screenings and isolation of carriers were taken. Prerequisite for a reasonable and democratic implementation of such measures, however, is the availability of sufficient testing opportunities (beyond reverse transcription PCR, the current gold standard). We, therefore, propose an electrochemical, microfluidic multiplexed polymer-based biosensor in combination with CRISPR/Cas-powered assays for low-cost and accessible point-of-care nucleic acid testing. In this study, we simultaneously screen for and identify SARS-CoV-2 infections (Omicron-variant) in clinical specimens (Sample-to-result time: ∼30 min), employing LbuCas13a, whilst bypassing reverse transcription as well as target amplification of the viral RNA (LODs of 2,000 and 7,520 copies/µl for the E and RdRP genes, respectively, and 50 copies/ml for combined targets), both of which are necessary for detection via PCR and other isothermal methods. In addition, we demonstrate the feasibility of combining synthetic biology-driven assays based on different classes of biomolecules, in this case protein-based ß-lactam antibiotic detection, on the same device. The programmability of the effector and multiplexing capacity (up to six analytes) of our platform, in combination with a miniaturized measurement setup, including a credit card sized near field communication (NFC) potentiostat and a microperistaltic pump, provide a promising on-site tool for identifying individuals infected with variants of concern and monitoring their disease progression alongside other potential biomarkers or medication clearance.
Collapse
Affiliation(s)
- Midori Johnston
- Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg, Germany
- FIT Freiburg Center for Interactive Materials and Bioinspired Technologies, University of Freiburg, Freiburg, Germany
| | - H Ceren Ates
- Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg, Germany
- FIT Freiburg Center for Interactive Materials and Bioinspired Technologies, University of Freiburg, Freiburg, Germany
| | - Regina T Glatz
- Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg, Germany
- FIT Freiburg Center for Interactive Materials and Bioinspired Technologies, University of Freiburg, Freiburg, Germany
| | - Hasti Mohsenin
- Faculty of Biology and Signalling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Rosanne Schmachtenberg
- Faculty of Biology and Signalling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Nathalie Göppert
- Institute of Virology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Daniela Huzly
- Institute of Virology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gerald A Urban
- Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg, Germany
- Freiburg Materials Research Center, University of Freiburg, Freiburg, Germany
| | - Wilfried Weber
- Faculty of Biology and Signalling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Can Dincer
- Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg, Germany
- FIT Freiburg Center for Interactive Materials and Bioinspired Technologies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
211
|
Khongthaw B, Dulta K, Chauhan PK, Kumar V, Ighalo JO. Lycopene: a therapeutic strategy against coronavirus disease 19 (COVID- 19). Inflammopharmacology 2022; 30:1955-1976. [PMID: 36050507 PMCID: PMC9436159 DOI: 10.1007/s10787-022-01061-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/18/2022] [Indexed: 02/07/2023]
Abstract
Lycopene is a group of phytochemicals found in nature, primarily in fruits and vegetables. Lycopene is thought to protect against a variety of diseases attributed to its antioxidant capabilities. Lycopene has anti-inflammatory, anti-cancer, and immunity-boosting qualities, among other biological and pharmacological benefits. COVID-19 (coronavirus disease 19) is an infectious disease caused by the SARS-CoV-2 virus, which has recently emerged as one of the world's leading causes of death. Patients may be asymptomatic or show signs of respiratory, cytokine release syndrome, gastrointestinal, or even multiple organ failure, all of which can lead to death. In COVID-19, inflammation, and cytokine storm are the key pathogenic mechanisms, according to SARS-CoV-2 infection symptoms. ARDS develops in some vulnerable hosts, which is accompanied by an inflammatory "cytokine syndrome" that causes lung damage. Immunological and inflammatory markers were linked to disease severity in mild and severe COVID-19 cases, implying that inflammatory markers, including IL-6, CRP, ESR, and PCT were significantly linked with COVID-19 severity. Patients with severe illness have reduced levels of several immune subsets, including CD4 + T, NK, and CD8 + cells. As a result, lycopene can be commended for bolstering physiological defenses against COVID-19 infections.
Collapse
Affiliation(s)
- Banlambhabok Khongthaw
- Shoolini University of Biotechnology and Management Sciences, Bajhol, Solan, Himachal Pradesh, 173229, India
| | - Kanika Dulta
- Shoolini University of Biotechnology and Management Sciences, Bajhol, Solan, Himachal Pradesh, 173229, India
| | - Pankaj Kumar Chauhan
- Shoolini University of Biotechnology and Management Sciences, Bajhol, Solan, Himachal Pradesh, 173229, India.
| | - Vinod Kumar
- Department of Life Sciences, Graphic Era (Deemed to be University), Dehradun, Uttarakhand, 248002, India
| | - Joshua O Ighalo
- Department of Chemical Engineering, Nnamdi Azikiwe University, P. M. B. 5025, Awka, Nigeria.
- Department of Chemical Engineering, Kansas State University, Manhattan, KS, USA.
| |
Collapse
|
212
|
Serna-Muñoz R, Hernández-Terán A, Soto-Nava M, Tapia-Trejo D, Ávila-Ríos S, Mejía-Nepomuceno F, García E, Castillejos-López M, Higuera-Iglesias AL, Aquino-Gálvez A, Thirion-Romero I, Pérez-Padilla R, Aguilar-Faisal JL, Vázquez-Pérez JA. Longitudinal Characterization of a Neutralizing and Total Antibody Response in Patients with Severe COVID-19 and Fatal Outcomes. Vaccines (Basel) 2022; 10:2063. [PMID: 36560474 PMCID: PMC9785259 DOI: 10.3390/vaccines10122063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/23/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
The host immune response to SARS-CoV-2 appears to play a critical role in disease pathogenesis and clinical manifestations in severe COVID-19 cases. Until now, the importance of developing a neutralizing antibody response in the acute phase and its relationship with progression to severe disease or fatal outcome among hospitalized patients remains unclear. In this study, we aim to characterize and compare longitudinally the primary humoral immune host response in the early stages of the disease, looking for an association between neutralization, antibody titers, infective viral lineage, and the clinical outcome in hospitalized and non-hospitalized patients. A total of 111 patients admitted at INER from November 2021 to June 2022 were included. We found that patients with negative or low neutralization showed a significant reduction in survival probability compared to patients with medium or high neutralization. We observed a significant decrease in the median of neutralization in patients infected with viral variants with changes in RBD of the spike protein. Our results suggest that developing an early and robust neutralizing response against SARS-CoV-2 may increase survival probability in critical patients.
Collapse
Affiliation(s)
- Ricardo Serna-Muñoz
- Departamento de Investigación en Tabaquismo y EPOC, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas INER, Mexico City 14080, Mexico
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Alejandra Hernández-Terán
- Departamento de Investigación en Tabaquismo y EPOC, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas INER, Mexico City 14080, Mexico
| | - Maribel Soto-Nava
- CIENI Centre for Research in Infectious Diseases, National Institute of Respiratory Diseases, Mexico City 14080, Mexico
| | - Daniela Tapia-Trejo
- CIENI Centre for Research in Infectious Diseases, National Institute of Respiratory Diseases, Mexico City 14080, Mexico
| | - Santiago Ávila-Ríos
- CIENI Centre for Research in Infectious Diseases, National Institute of Respiratory Diseases, Mexico City 14080, Mexico
| | - Fidencio Mejía-Nepomuceno
- Departamento de Investigación en Tabaquismo y EPOC, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas INER, Mexico City 14080, Mexico
| | - Emma García
- Departamento de Unidad de Epidemiología Hospitalaria e Infectología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas INER, Mexico City 14080, Mexico
| | - Manuel Castillejos-López
- Departamento de Unidad de Epidemiología Hospitalaria e Infectología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas INER, Mexico City 14080, Mexico
| | - Anjarath Lorena Higuera-Iglesias
- Departamento de Unidad de Epidemiología Hospitalaria e Infectología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas INER, Mexico City 14080, Mexico
| | - Arnoldo Aquino-Gálvez
- Laboratorio de Biología Molecular, Departamento de Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas INER, Mexico City 14080, Mexico
| | - Ireri Thirion-Romero
- Departamento de Investigación en Tabaquismo y EPOC, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas INER, Mexico City 14080, Mexico
| | - Rogelio Pérez-Padilla
- Departamento de Investigación en Tabaquismo y EPOC, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas INER, Mexico City 14080, Mexico
| | | | - Joel Armando Vázquez-Pérez
- Departamento de Investigación en Tabaquismo y EPOC, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas INER, Mexico City 14080, Mexico
| |
Collapse
|
213
|
Santos HM, Carvalho LB, Lodeiro C, Martins G, Gomes IL, D. T. Antunes W, Correia V, Almeida-Santos MM, Rebelo-de-Andrade H, Matos AP, Capelo J. “How to dissect viral infections and their interplay with the host-proteome by immunoaffinity and mass spectrometry: A tutorial.”. Microchem J 2022. [DOI: 10.1016/j.microc.2022.108323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
214
|
Turkkan A, Saglik I, Turan C, Sahin A, Akalin H, Ener B, Kara A, Celebi S, Sahin E, Hacimustafaoglu M. Nine-month course of SARS-CoV-2 antibodies in individuals with COVID-19 infection. Ir J Med Sci 2022; 191:2803-2811. [PMID: 35048229 PMCID: PMC8769943 DOI: 10.1007/s11845-021-02716-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/07/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND The continual course of the pandemic points to the importance of studies on the rate and durability of protective immunity after infection or vaccination. AIMS In this study, we aimed to monitor anti-nucleocapsid (N) and anti-spike (S) antibodies against SARS-CoV-2 nearly 9 months duration after infection. METHODS Anti-nucleocapsid (N) (at 11-15-20-29-38 weeks) and anti-spike antibodies (at 11 and 38 weeks) against SARS-CoV-2 were monitored during 38 weeks after the initial symptoms of COVID-19. RESULTS Of 37 cases between 18 and 57 years old, 54% were women. The findings showed that anti-N antibodies decreased significantly after the 15th week (between 15 and 20 weeks, p = 0.016; 20-29 weeks, p = 0.0009; and 29-38 weeks, p = 0.049). At the 38th week, mean antibody levels decreased 35% compared to the 11th week, and 8% of the cases turned negative results. Anti-N antibody average level was 56.48 on the 11th week (the cut-off index threshold ≥ 1). It was estimated statistically that it would decrease to an average of 20.48 in weeks 53-62. In females, average antibody levels of all measurements were lower than males (p > 0.05). Anti-S antibody levels 14% increased at 38th week compared to 11th week (quantitative positivity threshold ≥ 0.8 U/ml), and no cases were negative at 38th week. CONCLUSIONS Patients had ≥ 90% positivity after at least 9 months of symptoms, both anti-N and anti-S antibodies. In all samples, both anti-N and anti-S antibody levels were lower in females. The findings suggest that the quantitative values of anti-S antibodies remained high for at least 9 months and could provide protection.
Collapse
Affiliation(s)
- Alpaslan Turkkan
- Department of Public Health, Bursa Uludag University Medical Faculty, Bursa, Turkey
| | - Imran Saglik
- Department of Medical Microbiology, Bursa Uludag University Medical Faculty, Bursa, Turkey
| | - Cansu Turan
- Department of Pediatric Infectious Diseases, Bursa Uludag University Medical Faculty, Bursa, Turkey
| | - Ahmet Sahin
- Biochemistry and Clinical Biochemistry, Guven Tip Laboratuarı, Bursa, Turkey
| | - Halis Akalin
- Department of Infectious Diseases and Clinical Microbiology, Bursa Uludag University Medical Faculty, Bursa, Turkey
| | - Beyza Ener
- Department of Medical Microbiology, Bursa Uludag University Medical Faculty, Bursa, Turkey
| | - Ates Kara
- Department of Pediatric Infectious Diseases, Hacettepe University Medical Faculty, Ankara, Turkey
| | - Solmaz Celebi
- Department of Pediatric Infectious Diseases, Bursa Uludag University Medical Faculty, Bursa, Turkey
| | - Emre Sahin
- Department of Pediatric Infectious Diseases, Bursa Uludag University Medical Faculty, Bursa, Turkey
| | - Mustafa Hacimustafaoglu
- Department of Pediatric Infectious Diseases, Bursa Uludag University Medical Faculty, Bursa, Turkey.
| |
Collapse
|
215
|
Chen J, Li K, Zhang Z, Li K, Yu PS. A Survey on Applications of Artificial Intelligence in Fighting Against COVID-19. ACM COMPUTING SURVEYS 2022; 54:1-32. [DOI: 10.1145/3465398] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 05/01/2021] [Indexed: 01/05/2025]
Abstract
The COVID-19 pandemic caused by the SARS-CoV-2 virus has spread rapidly worldwide, leading to a global outbreak. Most governments, enterprises, and scientific research institutions are participating in the COVID-19 struggle to curb the spread of the pandemic. As a powerful tool against COVID-19, artificial intelligence (AI) technologies are widely used in combating this pandemic. In this survey, we investigate the main scope and contributions of AI in combating COVID-19 from the aspects of disease detection and diagnosis, virology and pathogenesis, drug and vaccine development, and epidemic and transmission prediction. In addition, we summarize the available data and resources that can be used for AI-based COVID-19 research. Finally, the main challenges and potential directions of AI in fighting against COVID-19 are discussed. Currently, AI mainly focuses on medical image inspection, genomics, drug development, and transmission prediction, and thus AI still has great potential in this field. This survey presents medical and AI researchers with a comprehensive view of the existing and potential applications of AI technology in combating COVID-19 with the goal of inspiring researchers to continue to maximize the advantages of AI and big data to fight COVID-19.
Collapse
Affiliation(s)
- Jianguo Chen
- Hunan University, China and University of Toronto, Toronto, ON, Canada
| | - Kenli Li
- Hunan University, Changsha, Hunan, China
| | | | - Keqin Li
- State University of New York, USA and Hunan University, Changsha, Hunan, China
| | - Philip S. Yu
- University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
216
|
SHRIVASTAVA ABHINAV, PADHIARI SATYAJIT, SINGH RAJNISH. Recurrence of disease in a patient with Covid-19: Re-activation or re-infection? THE NATIONAL MEDICAL JOURNAL OF INDIA 2022; 35:165-167. [DOI: 10.25259/nmji-35-3-165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARSCoV-2) was expected to induce a monophasic disease with subsequent immunity. However, case reports have since emerged which have found patients with either re-infection or re-activation of the virus. We describe a 44-year-old man with severe Covid-19-induced pneumonia who had recurrence of the disease after testing Covid-19-negative on three consecutive reverse transcriptase-polymerase chain reaction (RT-PCR) tests. Our patient underlines that caution should be exercised while planning for discharge of a patient irrespective of his previous negative test, especially in vulnerable patients and those who had moderate-to-severe disease requiring the use of immunosuppressive therapy. The fact that such patients could experience a re-activation or re-infection, requires monitoring and vigilance in the management of the pandemic at individual and collective levels.
Collapse
Affiliation(s)
- ABHINAV SHRIVASTAVA
- Department of Cardiology, Atal Bihari Vajpayee Institute of Medical Sciences and Dr Ram Manohar Lohia Hospital, New Delhi 110001, India
| | - SATYAJIT PADHIARI
- Department of Medicine, Atal Bihari Vajpayee Institute of Medical Sciences and Dr Ram Manohar Lohia Hospital, New Delhi 110001, India
| | - RAJNISH SINGH
- Department of Medicine, Atal Bihari Vajpayee Institute of Medical Sciences and Dr Ram Manohar Lohia Hospital, New Delhi 110001, India
| |
Collapse
|
217
|
Ghasemi D, Araeynejad F, Maghsoud O, Gerami N, Keihan AH, Rezaie E, Mehdizadeh S, Hosseinzadeh R, Mohammadi R, Bahardoust M, Heiat M. The Trend of IgG and IgM Antibodies During 6-Month Period After the Disease Episode in COVID-19 Patients. IRANIAN JOURNAL OF SCIENCE AND TECHNOLOGY, TRANSACTIONS A: SCIENCE 2022; 46:1555-1562. [PMCID: PMC9702912 DOI: 10.1007/s40995-022-01382-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 11/01/2022] [Indexed: 11/29/2022]
|
218
|
Strickler SS, Esper A, Wells L, Wood A, Frediani JK, Nehl E, Waggoner JJ, Rebolledo PA, Levy JM, Figueroa J, Ramachandra T, Lam W, Martin GS. Severe acute respiratory syndrome coronavirus 2 vaccine breakthrough infections: A single metro-based testing network experience. Front Med (Lausanne) 2022; 9:1031083. [PMID: 36507539 PMCID: PMC9732086 DOI: 10.3389/fmed.2022.1031083] [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: 08/29/2022] [Accepted: 11/10/2022] [Indexed: 11/26/2022] Open
Abstract
Objectives Understanding the incidence and characteristics that influence severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine breakthrough infections (VBIs) is imperative for developing public health policies to mitigate the coronavirus disease of 2019 (COVID-19) pandemic. We examined these factors and post-vaccination mitigation practices in individuals partially and fully vaccinated against SARS-CoV-2. Materials and methods Adults >18 years old were voluntarily enrolled from a single metro-based SARS-CoV-2 testing network from January to July 2021. Participants were categorized as asymptomatic or symptomatic, and as unvaccinated, partially vaccinated, or fully vaccinated. All participants had confirmed SARS-CoV-2 infection based on standard of care (SOC) testing with nasopharyngeal swabs. Variant analysis by rRT-PCR was performed in a subset of time-matched vaccinated and unvaccinated individuals. A subgroup of partially and fully vaccinated individuals with a positive SARS-CoV-2 rRT-PCR was contacted to assess disease severity and post-vaccination mitigation practices. Results Participants (n = 1,317) voluntarily underwent testing for SARS-CoV-2 during the enrollment period. A total of 29.5% of the population received at least one SARS-CoV-2 vaccine (n = 389), 12.8% partially vaccinated (n = 169); 16.1% fully vaccinated (n = 213). A total of 21.3% of partially vaccinated individuals tested positive (n = 36) and 9.4% of fully vaccinated individuals tested positive (n = 20) for SARS-CoV-2. Pfizer/BioNTech mRNA-1273 was the predominant vaccine received (1st dose = 66.8%, 2nd dose = 67.9%). Chronic liver disease and immunosuppression were more prevalent in the vaccinated (partially/fully) group compared to the unvaccinated group (p = 0.003, p = 0.021, respectively). There were more asymptomatic individuals in the vaccinated group compared to the unvaccinated group [n = 6 (10.7%), n = 16 (4.1%), p = 0.045]. CT values were lower for the unvaccinated group (median 24.3, IQR 19.1-30.5) compared to the vaccinated group (29.4, 22.0-33.7, p = 0.004). In the vaccinated group (n = 56), 18 participants were successfully contacted, 7 were lost to follow-up, and 2 were deceased. A total of 50% (n = 9) required hospitalization due to COVID-19 illness. Adherence to nationally endorsed mitigation strategies varied post-vaccination. Conclusion The incidence of SARS-CoV-2 infection at this center was 21.3% in the partially vaccinated group and 9.4% in the fully vaccinated group. Chronic liver disease and immunosuppression were more prevalent in the vaccinated SARS-CoV-2 positive group, suggesting that these may be risk factors for VBIs. Partially and fully vaccinated individuals had a higher incidence of asymptomatic SARS-CoV-2 and higher CT values compared to unvaccinated SARS-CoV-2 positive individuals.
Collapse
Affiliation(s)
- Samantha S. Strickler
- School of Medicine, Emory University, Atlanta, GA, United States,*Correspondence: Samantha S. Strickler,
| | - Annette Esper
- School of Medicine, Emory University, Atlanta, GA, United States
| | - Leona Wells
- School of Medicine, Emory University, Atlanta, GA, United States
| | - Anna Wood
- School of Medicine, Emory University, Atlanta, GA, United States
| | - Jennifer K. Frediani
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, United States
| | - Eric Nehl
- Rollins School of Public Health, Atlanta, GA, United States
| | | | - Paulina A. Rebolledo
- School of Medicine, Emory University, Atlanta, GA, United States,Hubert Department of Global Health, Rollins School of Public Health, Atlanta, GA, United States
| | - Joshua M. Levy
- School of Medicine, Emory University, Atlanta, GA, United States
| | - Janet Figueroa
- School of Medicine, Emory University, Atlanta, GA, United States
| | | | - Wilbur Lam
- School of Medicine, Emory University, Atlanta, GA, United States,Georgia Institute of Technology, Atlanta, GA, United States
| | | |
Collapse
|
219
|
Dong T, Han C, Jiang M, Zhang T, Kang Q, Wang P, Zhou F. A Four-Channel Surface Plasmon Resonance Sensor Functionalized Online for Simultaneous Detections of Anti-SARS-CoV-2 Antibody, Free Viral Particles, and Neutralized Viral Particles. ACS Sens 2022; 7:3560-3570. [PMID: 36382569 DOI: 10.1021/acssensors.2c02067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Current tests for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) detect either the constituent nucleic acids/proteins of the viral particles or antibodies specific to the virus, but cannot provide information about viral neutralization by an antibody and the efficacy of an antibody. Such information is important about individuals' vulnerability to severe symptoms or their likelihood of showing no symptoms. We immobilized online SARS-CoV-2 spike (S1) protein and angiotensin-converting enzyme 2 (ACE2) into separate surface plasmon resonance (SPR) channels of a tris-nitrilotriacetic acid (tris-NTA) chip to simultaneously detect the anti-S1 antibody and viral particles in serum samples. In addition, with a high-molecular-weight-cutoff filter, we separated the neutralized viral particles from the free antibody molecules and used a sensing channel immobilized with Protein G to determine antibody-neutralized viral particles. The optimal density of probe molecules in each fluidic channel can be precisely controlled through the closure and opening of the specific ports. By utilizing the high surface density of ACE2, multiple assays can be carried out without regenerations. These three species can be determined with a short analysis time (<12 min per assay) and excellent sensor-to-sensor/cycle-to-cycle reproducibility (RSD < 5%). When coupled with an autosampler, continuous assays can be performed in an unattended manner at a single chip for up to 6 days. Such a sensor capable of assaying serum samples containing the three species at different levels provides additional insights into the disease status and immunity of persons being tested, which should be helpful for containing the SARS-CoV-2 spread during the era of incessant viral mutations.
Collapse
Affiliation(s)
- Tianbao Dong
- Institute of Surface Analysis and Chemical Biology, University of Jinan, Jinan, Shandong, P. R. China, 250022
| | - Chaowei Han
- Institute of Surface Analysis and Chemical Biology, University of Jinan, Jinan, Shandong, P. R. China, 250022
| | - Meng Jiang
- Institute of Surface Analysis and Chemical Biology, University of Jinan, Jinan, Shandong, P. R. China, 250022
| | - Tiantian Zhang
- University Hospital, University of Jinan, Jinan, Shandong, P. R. China, 250022
| | - Qing Kang
- Institute of Surface Analysis and Chemical Biology, University of Jinan, Jinan, Shandong, P. R. China, 250022
| | - Pengcheng Wang
- Institute of Surface Analysis and Chemical Biology, University of Jinan, Jinan, Shandong, P. R. China, 250022
| | - Feimeng Zhou
- Institute of Surface Analysis and Chemical Biology, University of Jinan, Jinan, Shandong, P. R. China, 250022
| |
Collapse
|
220
|
Evaluation of compressed sodium chloride on the inactivation of SARS-CoV-2 and surrogates. PLoS One 2022; 17:e0277881. [PMID: 36409696 PMCID: PMC9678310 DOI: 10.1371/journal.pone.0277881] [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: 02/17/2022] [Accepted: 11/05/2022] [Indexed: 11/22/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) causes the global COVID-19 pandemic. Limited studies have been performed on various types of disinfectants utilized to control the spread of this highly contagious virus. This study aimed to investigate the inactivation of SARS-CoV-2 using compressed sodium chloride (CSC) surface. A real-time reverse transcriptase quantitative PCR (RT-qPCR) assay was used to evaluate the effectiveness of CSC on the disintegration of viral RNA in a time dependent manner. The effects of CSC on viral infectivity were determined using a TCID50 assay of a surrogate virus, hCoV-229E, in MRC-5 cell culture. The results demonstrated that CSC achieved a 2 to 3- log10 reduction of viral genomic RNA for a laboratory strain of hCoV-229E, and clinical samples of hCoV-229E and hCoV-OC43. A 3 to 4-log10 reduction was observed for SARS-CoV-2 (RdRp and E gene) suggesting that a CSC surface could effectively disintegrate the SARS-CoV-2 RNA genome. CSC was observed to have a 6 log10 inactivation of infectious hCoV-229E using cell culture after 5 minutes of exposure compared to the control, indicating good disinfection efficacy of a CSC surface against virus.
Collapse
|
221
|
Ang GY, Chan KG, Yean CY, Yu CY. Lateral Flow Immunoassays for SARS-CoV-2. Diagnostics (Basel) 2022; 12:2854. [PMID: 36428918 PMCID: PMC9689684 DOI: 10.3390/diagnostics12112854] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/09/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022] Open
Abstract
The continued circulation of SARS-CoV-2 virus in different parts of the world opens up the possibility for more virulent variants to evolve even as the coronavirus disease 2019 transitions from pandemic to endemic. Highly transmissible and virulent variants may seed new disruptive epidemic waves that can easily put the healthcare system under tremendous pressure. Despite various nucleic acid-based diagnostic tests that are now commercially available, the wide applications of these tests are largely hampered by specialized equipment requirements that may not be readily available, accessible and affordable in less developed countries or in low resource settings. Hence, the availability of lateral flow immunoassays (LFIs), which can serve as a diagnostic tool by detecting SARS-CoV-2 antigen or as a serological tool by measuring host immune response, is highly appealing. LFI is rapid, low cost, equipment-free, scalable for mass production and ideal for point-of-care settings. In this review, we first summarize the principle and assay format of these LFIs with emphasis on those that were granted emergency use authorization by the US Food and Drug Administration followed by discussion on the specimen type, marker selection and assay performance. We conclude with an overview of challenges and future perspective of LFI applications.
Collapse
Affiliation(s)
- Geik Yong Ang
- Faculty of Sports Science and Recreation, Universiti Teknologi MARA, Shah Alam 40450, Malaysia
| | - Kok Gan Chan
- Division of Genetics and Molecular Biology, Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia
- International Genome Centre, Jiangsu University, Zhenjiang 212013, China
| | - Chan Yean Yean
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Malaysia
| | - Choo Yee Yu
- Laboratory of Vaccine and Biomolecules, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
| |
Collapse
|
222
|
Fox T, Geppert J, Dinnes J, Scandrett K, Bigio J, Sulis G, Hettiarachchi D, Mathangasinghe Y, Weeratunga P, Wickramasinghe D, Bergman H, Buckley BS, Probyn K, Sguassero Y, Davenport C, Cunningham J, Dittrich S, Emperador D, Hooft L, Leeflang MM, McInnes MD, Spijker R, Struyf T, Van den Bruel A, Verbakel JY, Takwoingi Y, Taylor-Phillips S, Deeks JJ. Antibody tests for identification of current and past infection with SARS-CoV-2. Cochrane Database Syst Rev 2022; 11:CD013652. [PMID: 36394900 PMCID: PMC9671206 DOI: 10.1002/14651858.cd013652.pub2] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND The diagnostic challenges associated with the COVID-19 pandemic resulted in rapid development of diagnostic test methods for detecting SARS-CoV-2 infection. Serology tests to detect the presence of antibodies to SARS-CoV-2 enable detection of past infection and may detect cases of SARS-CoV-2 infection that were missed by earlier diagnostic tests. Understanding the diagnostic accuracy of serology tests for SARS-CoV-2 infection may enable development of effective diagnostic and management pathways, inform public health management decisions and understanding of SARS-CoV-2 epidemiology. OBJECTIVES To assess the accuracy of antibody tests, firstly, to determine if a person presenting in the community, or in primary or secondary care has current SARS-CoV-2 infection according to time after onset of infection and, secondly, to determine if a person has previously been infected with SARS-CoV-2. Sources of heterogeneity investigated included: timing of test, test method, SARS-CoV-2 antigen used, test brand, and reference standard for non-SARS-CoV-2 cases. SEARCH METHODS The COVID-19 Open Access Project living evidence database from the University of Bern (which includes daily updates from PubMed and Embase and preprints from medRxiv and bioRxiv) was searched on 30 September 2020. We included additional publications from the Evidence for Policy and Practice Information and Co-ordinating Centre (EPPI-Centre) 'COVID-19: Living map of the evidence' and the Norwegian Institute of Public Health 'NIPH systematic and living map on COVID-19 evidence'. We did not apply language restrictions. SELECTION CRITERIA We included test accuracy studies of any design that evaluated commercially produced serology tests, targeting IgG, IgM, IgA alone, or in combination. Studies must have provided data for sensitivity, that could be allocated to a predefined time period after onset of symptoms, or after a positive RT-PCR test. Small studies with fewer than 25 SARS-CoV-2 infection cases were excluded. We included any reference standard to define the presence or absence of SARS-CoV-2 (including reverse transcription polymerase chain reaction tests (RT-PCR), clinical diagnostic criteria, and pre-pandemic samples). DATA COLLECTION AND ANALYSIS We use standard screening procedures with three reviewers. Quality assessment (using the QUADAS-2 tool) and numeric study results were extracted independently by two people. Other study characteristics were extracted by one reviewer and checked by a second. We present sensitivity and specificity with 95% confidence intervals (CIs) for each test and, for meta-analysis, we fitted univariate random-effects logistic regression models for sensitivity by eligible time period and for specificity by reference standard group. Heterogeneity was investigated by including indicator variables in the random-effects logistic regression models. We tabulated results by test manufacturer and summarised results for tests that were evaluated in 200 or more samples and that met a modification of UK Medicines and Healthcare products Regulatory Agency (MHRA) target performance criteria. MAIN RESULTS We included 178 separate studies (described in 177 study reports, with 45 as pre-prints) providing 527 test evaluations. The studies included 64,688 samples including 25,724 from people with confirmed SARS-CoV-2; most compared the accuracy of two or more assays (102/178, 57%). Participants with confirmed SARS-CoV-2 infection were most commonly hospital inpatients (78/178, 44%), and pre-pandemic samples were used by 45% (81/178) to estimate specificity. Over two-thirds of studies recruited participants based on known SARS-CoV-2 infection status (123/178, 69%). All studies were conducted prior to the introduction of SARS-CoV-2 vaccines and present data for naturally acquired antibody responses. Seventy-nine percent (141/178) of studies reported sensitivity by week after symptom onset and 66% (117/178) for convalescent phase infection. Studies evaluated enzyme-linked immunosorbent assays (ELISA) (165/527; 31%), chemiluminescent assays (CLIA) (167/527; 32%) or lateral flow assays (LFA) (188/527; 36%). Risk of bias was high because of participant selection (172, 97%); application and interpretation of the index test (35, 20%); weaknesses in the reference standard (38, 21%); and issues related to participant flow and timing (148, 82%). We judged that there were high concerns about the applicability of the evidence related to participants in 170 (96%) studies, and about the applicability of the reference standard in 162 (91%) studies. Average sensitivities for current SARS-CoV-2 infection increased by week after onset for all target antibodies. Average sensitivity for the combination of either IgG or IgM was 41.1% in week one (95% CI 38.1 to 44.2; 103 evaluations; 3881 samples, 1593 cases), 74.9% in week two (95% CI 72.4 to 77.3; 96 evaluations, 3948 samples, 2904 cases) and 88.0% by week three after onset of symptoms (95% CI 86.3 to 89.5; 103 evaluations, 2929 samples, 2571 cases). Average sensitivity during the convalescent phase of infection (up to a maximum of 100 days since onset of symptoms, where reported) was 89.8% for IgG (95% CI 88.5 to 90.9; 253 evaluations, 16,846 samples, 14,183 cases), 92.9% for IgG or IgM combined (95% CI 91.0 to 94.4; 108 evaluations, 3571 samples, 3206 cases) and 94.3% for total antibodies (95% CI 92.8 to 95.5; 58 evaluations, 7063 samples, 6652 cases). Average sensitivities for IgM alone followed a similar pattern but were of a lower test accuracy in every time slot. Average specificities were consistently high and precise, particularly for pre-pandemic samples which provide the least biased estimates of specificity (ranging from 98.6% for IgM to 99.8% for total antibodies). Subgroup analyses suggested small differences in sensitivity and specificity by test technology however heterogeneity in study results, timing of sample collection, and smaller sample numbers in some groups made comparisons difficult. For IgG, CLIAs were the most sensitive (convalescent-phase infection) and specific (pre-pandemic samples) compared to both ELISAs and LFAs (P < 0.001 for differences across test methods). The antigen(s) used (whether from the Spike-protein or nucleocapsid) appeared to have some effect on average sensitivity in the first weeks after onset but there was no clear evidence of an effect during convalescent-phase infection. Investigations of test performance by brand showed considerable variation in sensitivity between tests, and in results between studies evaluating the same test. For tests that were evaluated in 200 or more samples, the lower bound of the 95% CI for sensitivity was 90% or more for only a small number of tests (IgG, n = 5; IgG or IgM, n = 1; total antibodies, n = 4). More test brands met the MHRA minimum criteria for specificity of 98% or above (IgG, n = 16; IgG or IgM, n = 5; total antibodies, n = 7). Seven assays met the specified criteria for both sensitivity and specificity. In a low-prevalence (2%) setting, where antibody testing is used to diagnose COVID-19 in people with symptoms but who have had a negative PCR test, we would anticipate that 1 (1 to 2) case would be missed and 8 (5 to 15) would be falsely positive in 1000 people undergoing IgG or IgM testing in week three after onset of SARS-CoV-2 infection. In a seroprevalence survey, where prevalence of prior infection is 50%, we would anticipate that 51 (46 to 58) cases would be missed and 6 (5 to 7) would be falsely positive in 1000 people having IgG tests during the convalescent phase (21 to 100 days post-symptom onset or post-positive PCR) of SARS-CoV-2 infection. AUTHORS' CONCLUSIONS Some antibody tests could be a useful diagnostic tool for those in whom molecular- or antigen-based tests have failed to detect the SARS-CoV-2 virus, including in those with ongoing symptoms of acute infection (from week three onwards) or those presenting with post-acute sequelae of COVID-19. However, antibody tests have an increasing likelihood of detecting an immune response to infection as time since onset of infection progresses and have demonstrated adequate performance for detection of prior infection for sero-epidemiological purposes. The applicability of results for detection of vaccination-induced antibodies is uncertain.
Collapse
Affiliation(s)
- Tilly Fox
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Julia Geppert
- Division of Health Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Jacqueline Dinnes
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Katie Scandrett
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Jacob Bigio
- Research Institute of the McGill University Health Centre, Montreal, Canada
- McGill International TB Centre, Montreal, Canada
| | - Giorgia Sulis
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada
| | - Dineshani Hettiarachchi
- Department of Anatomy Genetics and Biomedical Informatics, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Yasith Mathangasinghe
- Department of Anatomy Genetics and Biomedical Informatics, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
- Australian Regenerative Medicine Institute, Monash University, Clayton, Australia
| | - Praveen Weeratunga
- Department of Clinical Medicine, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | | | | | - Brian S Buckley
- Cochrane Response, Cochrane, London, UK
- Department of Surgery, University of the Philippines, Manila, Philippines
| | | | | | - Clare Davenport
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Jane Cunningham
- Global Malaria Programme, World Health Organization, Geneva, Switzerland
| | | | | | - Lotty Hooft
- Cochrane Netherlands, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht , Netherlands
| | - Mariska Mg Leeflang
- Epidemiology and Data Science, Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Public Health, Amsterdam, Netherlands
| | | | - René Spijker
- Medical Library, Amsterdam UMC, University of Amsterdam, Amsterdam Public Health, Amsterdam, Netherlands
- Cochrane Netherlands, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Thomas Struyf
- Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Ann Van den Bruel
- Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Jan Y Verbakel
- Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Yemisi Takwoingi
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Sian Taylor-Phillips
- Division of Health Sciences, Warwick Medical School, University of Warwick, Coventry, UK
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Jonathan J Deeks
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| |
Collapse
|
223
|
Wong SC, Au AKW, Lo JYC, Ho PL, Hung IFN, To KKW, Yuen KY, Cheng VCC. Evolution and Control of COVID-19 Epidemic in Hong Kong. Viruses 2022; 14:2519. [PMID: 36423128 PMCID: PMC9698160 DOI: 10.3390/v14112519] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Hong Kong SAR has adopted universal masking, social distancing, testing of all symptomatic and high-risk groups for isolation of confirmed cases in healthcare facilities, and quarantine of contacts as epidemiological control measures without city lockdown or border closure. These measures successfully suppressed the community transmission of pre-Omicron SARS-CoV-2 variants or lineages during the first to the fourth wave. No nosocomial SARS-CoV-2 infection was documented among healthcare workers in the first 300 days. The strategy of COVID-19 containment was adopted to provide additional time to achieve population immunity by vaccination. The near-zero COVID-19 situation for about 8 months in 2021 did not enable adequate immunization of the eligible population. A combination of factors was identified, especially population complacency associated with the low local COVID-19 activity, together with vaccine hesitancy. The importation of the highly transmissible Omicron variant kickstarted the fifth wave of COVID-19, which could no longer be controlled by our initial measures. The explosive fifth wave, which was partially contributed by vertical airborne transmission in high-rise residential buildings, resulted in over one million cases of infection. In this review, we summarize the epidemiology of COVID-19 and the infection control and public health measures against the importation and dissemination of SARS-CoV-2 until day 1000.
Collapse
Affiliation(s)
- Shuk-Ching Wong
- Infection Control Team, Queen Mary Hospital, Hong Kong West Cluster, Hong Kong SAR, China
| | - Albert Ka-Wing Au
- Centre for Health Protection, Department of Health, Hong Kong SAR, China
| | - Janice Yee-Chi Lo
- Centre for Health Protection, Department of Health, Hong Kong SAR, China
| | - Pak-Leung Ho
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Carol Yu Center for Infection, The University of Hong Kong, Hong Kong SAR, China
| | - Ivan Fan-Ngai Hung
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kelvin Kai-Wang To
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kwok-Yung Yuen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Vincent Chi-Chung Cheng
- Infection Control Team, Queen Mary Hospital, Hong Kong West Cluster, Hong Kong SAR, China
- Department of Microbiology, Queen Mary Hospital, Hong Kong SAR, China
| |
Collapse
|
224
|
Kuanr M, Mohapatra P, Mittal S, Maindarkar M, Fouda MM, Saba L, Saxena S, Suri JS. Recommender System for the Efficient Treatment of COVID-19 Using a Convolutional Neural Network Model and Image Similarity. Diagnostics (Basel) 2022; 12:2700. [PMID: 36359545 PMCID: PMC9689970 DOI: 10.3390/diagnostics12112700] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/30/2022] [Accepted: 11/03/2022] [Indexed: 09/09/2023] Open
Abstract
Background: Hospitals face a significant problem meeting patients' medical needs during epidemics, especially when the number of patients increases rapidly, as seen during the recent COVID-19 pandemic. This study designs a treatment recommender system (RS) for the efficient management of human capital and resources such as doctors, medicines, and resources in hospitals. We hypothesize that a deep learning framework, when combined with search paradigms in an image framework, can make the RS very efficient. Methodology: This study uses a Convolutional neural network (CNN) model for the feature extraction of the images and discovers the most similar patients. The input queries patients from the hospital database with similar chest X-ray images. It uses a similarity metric for the similarity computation of the images. Results: This methodology recommends the doctors, medicines, and resources associated with similar patients to a COVID-19 patients being admitted to the hospital. The performance of the proposed RS is verified with five different feature extraction CNN models and four similarity measures. The proposed RS with a ResNet-50 CNN feature extraction model and Maxwell-Boltzmann similarity is found to be a proper framework for treatment recommendation with a mean average precision of more than 0.90 for threshold similarities in the range of 0.7 to 0.9 and an average highest cosine similarity of more than 0.95. Conclusions: Overall, an RS with a CNN model and image similarity is proven as an efficient tool for the proper management of resources during the peak period of pandemics and can be adopted in clinical settings.
Collapse
Affiliation(s)
- Madhusree Kuanr
- Department of Computer Science and Engineering, IIIT, Bhubaneswar 751003, India
| | | | - Sanchi Mittal
- Department of Computer Science and Engineering, IIIT, Bhubaneswar 751003, India
| | - Mahesh Maindarkar
- Stroke Monitoring and Diagnostic Division, AtheroPointTM, Roseville, CA 95661, USA
| | - Mostafa M. Fouda
- Department of Electrical and Computer Engineering, Idaho State University, Pocatello, ID 83209, USA
| | - Luca Saba
- Department of Radiology, University of Cagliari, 09123 Cagliari, Italy
| | - Sanjay Saxena
- Department of Computer Science and Engineering, IIIT, Bhubaneswar 751003, India
| | - Jasjit S. Suri
- Stroke Monitoring and Diagnostic Division, AtheroPointTM, Roseville, CA 95661, USA
- Knowledge Engineering Center, Global Biomedical Technologies, Inc., Roseville, CA 95661, USA
| |
Collapse
|
225
|
Zarei Ghobadi M, Emamzadeh R, Teymoori-Rad M, Afsaneh E. Exploration of blood-derived coding and non-coding RNA diagnostic immunological panels for COVID-19 through a co-expressed-based machine learning procedure. Front Immunol 2022; 13:1001070. [PMID: 36405703 PMCID: PMC9670818 DOI: 10.3389/fimmu.2022.1001070] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/17/2022] [Indexed: 05/12/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS- CoV-2) is the causative virus of the pandemic coronavirus disease 2019 (COVID-19). Evaluating the immunological factors and other implicated processes underlying the progression of COVID-19 is essential for the recognition and then the design of efficacious therapies. Therefore, we analyzed RNAseq data obtained from PBMCs of the COVID-19 patients to explore coding and non-coding RNA diagnostic immunological panels. For this purpose, we integrated multiple RNAseq data and analyzed them overall as well as by considering the state of disease including severe and non-severe conditions. Afterward, we utilized a co-expressed-based machine learning procedure comprising weighted-gene co-expression analysis and differential expression gene as filter phase and recursive feature elimination-support vector machine as wrapper phase. This procedure led to the identification of two modules containing 5 and 84 genes which are mostly involved in cell dysregulation and innate immune suppression, respectively. Moreover, the role of vitamin D in regulating some classifiers was highlighted. Further analysis disclosed the role of discriminant miRNAs including miR-197-3p, miR-150-5p, miR-340-5p, miR-122-5p, miR-1307-3p, miR-34a-5p, miR-98-5p and their target genes comprising GAN, VWC2, TNFRSF6B, and CHST3 in the metabolic pathways. These classifiers differentiate the final fate of infection toward severe or non-severe COVID-19. The identified classifier genes and miRNAs may help in the proper design of therapeutic procedures considering their involvement in the immune and metabolic pathways.
Collapse
Affiliation(s)
- Mohadeseh Zarei Ghobadi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Rahman Emamzadeh
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Majid Teymoori-Rad
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Elaheh Afsaneh
- Department of Physics, University of Isfahan, Hezar Jarib, Isfahan, Iran
| |
Collapse
|
226
|
Tami A, van der Gun BTF, Wold KI, Vincenti-González MF, Veloo ACM, Knoester M, Harmsma VPR, de Boer GC, Huckriede ALW, Pantano D, Gard L, Rodenhuis-Zybert IA, Upasani V, Smit J, Dijkstra AE, de Haan JJ, van Elst JM, van den Boogaard J, O’ Boyle S, Nacul L, Niesters HGM, Friedrich AW. The COVID HOME study research protocol: Prospective cohort study of non-hospitalised COVID-19 patients. PLoS One 2022; 17:e0273599. [PMID: 36327223 PMCID: PMC9632784 DOI: 10.1371/journal.pone.0273599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/05/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Guidelines on COVID-19 management are developed as we learn from this pandemic. However, most research has been done on hospitalised patients and the impact of the disease on non-hospitalised and their role in transmission are not yet well understood. The COVID HOME study conducts research among COVID-19 patients and their family members who were not hospitalised during acute disease, to guide patient care and inform public health guidelines for infection prevention and control in the community and household. METHODS An ongoing prospective longitudinal observational study of COVID-19 outpatients was established in March 2020 at the beginning of the COVID-19 pandemic in the Netherlands. Laboratory confirmed SARS-CoV-2 infected individuals of all ages that did not merit hospitalisation, and their household (HH) members, were enrolled after written informed consent. Enrolled participants were visited at home within 48 hours after initial diagnosis, and then weekly on days 7, 14 and 21 to obtain clinical data, a blood sample for biochemical parameters/cytokines and serological determination; and a nasopharyngeal/throat swab plus urine, stool and sperm or vaginal secretion (if consenting) to test for SARS-CoV-2 by RT-PCR (viral shedding) and for viral culturing. Weekly nasopharyngeal/throat swabs and stool samples, plus a blood sample on days 0 and 21 were also taken from HH members to determine whether and when they became infected. All participants were invited to continue follow-up at 3-, 6-, 12- and 18-months post-infection to assess long-term sequelae and immunological status.
Collapse
Affiliation(s)
- Adriana Tami
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Bernardina T. F. van der Gun
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Karin I. Wold
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - María F. Vincenti-González
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Alida C. M. Veloo
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marjolein Knoester
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Valerie P. R. Harmsma
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gerolf C. de Boer
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Anke L. W. Huckriede
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Daniele Pantano
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Lilli Gard
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Izabela A. Rodenhuis-Zybert
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Vinit Upasani
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jolanda Smit
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Akkelies E. Dijkstra
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jacco J. de Haan
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jip M. van Elst
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Shennae O’ Boyle
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Luis Nacul
- Department of Clinical Research, Faculty of Medicine and London School of Hygiene and Tropical Medicine, University of British Columbia, Vancouver, Canada
| | - Hubert G. M. Niesters
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Alex W. Friedrich
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
227
|
Shahbaz S, Oyegbami O, Saito S, Osman M, Sligl W, Elahi S. Differential effects of age, sex and dexamethasone therapy on ACE2/TMPRSS2 expression and susceptibility to SARS-CoV-2 infection. Front Immunol 2022; 13:1021928. [PMID: 36405732 PMCID: PMC9671168 DOI: 10.3389/fimmu.2022.1021928] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/21/2022] [Indexed: 08/30/2023] Open
Abstract
ACE2 and TMPRSS2 are crucial for SARS-CoV-2 entry into the cell. Although ACE2 facilitates viral entry, its loss leads to promoting the devastating clinical symptoms of COVID-19 disease. Thus, enhanced ACE2/TMPRSS2 expression is likely to increase predisposition of target cells to SARS-CoV-2 infection. However, little evidence existed about the biological kinetics of these two enzymes and whether dexamethasone treatment modulates their expression. Here, we show that the expression of ACE2 at the protein and mRNA levels was significantly higher in the lung and heart tissues of neonatal compared to adult mice. However, the expression of TMPRSS2 was developmentally regulated. Our results may introduce a novel concept for the reduced susceptibility of the young to SARS-CoV-2 infection. Moreover, ACE2 expression but not TMPRSS2 was upregulated in adult female lungs compared to their male counterparts. Interestingly, the ACE2 and TMPRSS2 expressions were upregulated by dexamethasone treatment in the lung and heart tissues in both neonatal and adult mice. Furthermore, our findings provide a novel mechanism for the observed differential therapeutic effects of dexamethasone in COVID-19 patients. As such, dexamethasone exhibits different therapeutic effects depending on the disease stage. This was supported by increased ACE2/TMPRSS2 expression and subsequently enhanced infection of normal human bronchial epithelial cells (NHBE) and Vero E6 cells with SARS-CoV-2 once pre-treated with dexamethasone. Therefore, our results suggest that individuals who take dexamethasone for other clinical conditions may become more prone to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Shima Shahbaz
- Department of Dentistry, Division of Foundational Sciences, Edmonton, AB, Canada
| | - Olaide Oyegbami
- Department of Dentistry, Division of Foundational Sciences, Edmonton, AB, Canada
| | - Suguru Saito
- Department of Dentistry, Division of Foundational Sciences, Edmonton, AB, Canada
| | - Mohammed Osman
- Department of Medicine, Division of Rheumatology, University of Alberta, Edmonton, AB, Canada
| | - Wendy Sligl
- Department of Critical Care Medicine, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, Division of Infectious Diseases, University of Alberta, Edmonton, AB, Canada
| | - Shokrollah Elahi
- Department of Dentistry, Division of Foundational Sciences, Edmonton, AB, Canada
- Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, Edmonton, AB, Canada
- Women and Children Health Research Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
228
|
Descriptive evaluation of antibody responses to severe acute respiratory coronavirus virus 2 (SARS-CoV-2) infection in plasma and gingival crevicular fluid in a nursing home cohort-Arkansas, June-August 2020. Infect Control Hosp Epidemiol 2022; 43:1610-1617. [PMID: 34802478 PMCID: PMC9379264 DOI: 10.1017/ice.2021.484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To characterize and compare severe acute respiratory coronavirus virus 2 (SARS-CoV-2)-specific immune responses in plasma and gingival crevicular fluid (GCF) from nursing home residents during and after natural infection. DESIGN Prospective cohort. SETTING Nursing home. PARTICIPANTS SARS-CoV-2-infected nursing home residents. METHODS A convenience sample of 14 SARS-CoV-2-infected nursing home residents, enrolled 4-13 days after real-time reverse transcription polymerase chain reaction diagnosis, were followed for 42 days. After diagnosis, plasma SARS-CoV-2-specific pan-Immunoglobulin (Ig), IgG, IgA, IgM, and neutralizing antibodies were measured at 5 time points, and GCF SARS-CoV-2-specific IgG and IgA were measured at 4 time points. RESULTS All participants demonstrated immune responses to SARS-CoV-2 infection. Among 12 phlebotomized participants, plasma was positive for pan-Ig and IgG in all 12 participants. Neutralizing antibodies were positive in 11 participants; IgM was positive in 10 participants, and IgA was positive in 9 participants. Among 14 participants with GCF specimens, GCF was positive for IgG in 13 participants and for IgA in 12 participants. Immunoglobulin responses in plasma and GCF had similar kinetics; median times to peak antibody response were similar across specimen types (4 weeks for IgG; 3 weeks for IgA). Participants with pan-Ig, IgG, and IgA detected in plasma and GCF IgG remained positive throughout this evaluation, 46-55 days after diagnosis. All participants were viral-culture negative by the first detection of antibodies. CONCLUSIONS Nursing home residents had detectable SARS-CoV-2 antibodies in plasma and GCF after infection. Kinetics of antibodies detected in GCF mirrored those from plasma. Noninvasive GCF may be useful for detecting and monitoring immunologic responses in populations unable or unwilling to be phlebotomized.
Collapse
|
229
|
Gu SH, Song DH, Yun H, Kim J, Lee S, Lee H, Lee TH, Kang SM, Jung YS, Hur G, Lee D. Molecular characterization of SARS-CoV-2 from the saliva of patients in the Republic of Korea in 2020. Health Sci Rep 2022; 5:e856. [PMID: 36210871 PMCID: PMC9528954 DOI: 10.1002/hsr2.856] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 09/07/2022] [Accepted: 09/13/2022] [Indexed: 11/06/2022] Open
Abstract
Background and aims Despite global vaccination efforts, the number of confirmed cases of coronavirus disease 2019 (COVID-19) remains high. To overcome the crisis precipitated by the ongoing pandemic, characteristic studies such as virus diagnosis, isolation, and genome analysis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are necessary. Herein, we report the isolation and molecular characterization of SARS-CoV-2 from the saliva of patients who had tested positive for COVID-19 at Proving Ground in Taean County, Republic of Korea, in 2020. Methods We analyzed the whole-genome sequence of SARS-CoV-2 isolated from the saliva samples of patients through next-generation sequencing. We also successfully isolated SARS-CoV-2 from the saliva samples of two patients by using cell culture, which was used to study the cytopathic effects and viral replication in Vero E6 cells. Results Whole-genome sequences of the isolates, SARS-CoV-2 ADD-2 and ADD-4, obtained from saliva were identical, and phylogenetic analysis using Bayesian inference methods showed SARS-CoV-2 GH clade (B.1.497) genome-specific clustering. Typical coronavirus-like particles, with diameters of 70-120 nm, were observed in the SARS-CoV-2 infected Vero E6 cells using transmission electron microscopy. Conclusion In conclusion, this report provides insights into the molecular diagnosis, isolation, genetic characteristics, and diversity of SARS-CoV-2 isolated from the saliva of patients. Further studies are needed to explore and monitor the evolution and characteristics of SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Se Hun Gu
- Chem‐Bio Technology CenterAgency for Defense DevelopmentDaejeonRepublic of Korea
| | - Dong Hyun Song
- Chem‐Bio Technology CenterAgency for Defense DevelopmentDaejeonRepublic of Korea
| | - Hyeongseok Yun
- Chem‐Bio Technology CenterAgency for Defense DevelopmentDaejeonRepublic of Korea
| | - Jung‐Eun Kim
- Chem‐Bio Technology CenterAgency for Defense DevelopmentDaejeonRepublic of Korea
| | - Seung‐Ho Lee
- Chem‐Bio Technology CenterAgency for Defense DevelopmentDaejeonRepublic of Korea
| | - Hyunjin Lee
- Chem‐Bio Technology CenterAgency for Defense DevelopmentDaejeonRepublic of Korea
| | - Tae Ho Lee
- Institute of Health and EnvironmentDaejeonRepublic of Korea
| | - Seol Muk Kang
- Defense Test and Evaluation Research InstituteAgency for Defense DevelopmentTaeanRepublic of Korea
| | - Yu Sub Jung
- Defense Test and Evaluation Research InstituteAgency for Defense DevelopmentTaeanRepublic of Korea
| | - Gyeunghaeng Hur
- Chem‐Bio Technology CenterAgency for Defense DevelopmentDaejeonRepublic of Korea
| | - Daesang Lee
- Chem‐Bio Technology CenterAgency for Defense DevelopmentDaejeonRepublic of Korea
| |
Collapse
|
230
|
Moraleda C, Domínguez-Rodríguez S, Mesa JM, García-Sánchez P, de la Serna M, Alonso-Cadenas JA, Bermejo A, Sabrido G, Martínez-Campos L, González-Posada AF, Illán-Ramos M, Cobos-Carrascosa E, Ballesteros Á, Galán JC, Llorente F, Aguilera-Alonso D, Jiménez AB, Catalán P, Dahdouh E, Navarro I, Fernández-Garoz B, Mendoza P, Pérez-Jorge C, Cabezas-Fernández T, Blázquez-Gamero D, Rivas G, Gonzalez-Donapetry P, Sáez E, Fernández-Pinero J, Lucas-Fernández J, Pérez-Ramírez E, Merino P, Miragaya S, Lorente J, Iglesias I, Tagarro A. Oral saliva swab reverse transcription PCR for Covid-19 in the paediatric population. Arch Dis Child 2022; 107:1051-1058. [PMID: 35688603 PMCID: PMC9240444 DOI: 10.1136/archdischild-2021-323712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/12/2022] [Indexed: 11/21/2022]
Abstract
OBJECTIVES To evaluate the performance of oral saliva swab (OSS) reverse transcription PCR (RT-PCR) compared with RT-PCR and antigen rapid diagnostic test (Ag-RDT) on nasopharyngeal swabs (NPS) for SARS-CoV-2 in children. DESIGN Cross-sectional multicentre diagnostic study. SETTING Study nested in a prospective, observational cohort (EPICO-AEP) performed between February and March 2021 including 10 hospitals in Spain. PATIENTS Children from 0 to 18 years with symptoms compatible with Covid-19 of ≤5 days of duration were included. Two NPS samples (Ag-RDT and RT-PCR) and one OSS sample for RT-PCR were collected. MAIN OUTCOME Performance of Ag-RDT and RT-PCR on NPS and RT-PCR on OSS sample for SARS-CoV-2. RESULTS 1174 children were included, aged 3.8 years (IQR 1.7-9.0); 73/1174 (6.2%) patients tested positive by at least one of the techniques. Sensitivity and specificity of OSS RT-PCR were 72.1% (95% CI 59.7 to 81.9) and 99.6% (95% CI 99 to 99.9), respectively, versus 61.8% (95% CI 49.1 to 73) and 99.9% (95% CI 99.4 to 100) for the Ag-RDT. Kappa index was 0.79 (95% CI 0.72 to 0.88) for OSS RT-PCR and 0.74 (95% CI 0.65 to 0.84) for Ag-RDT versus NPS RT-PCR. CONCLUSIONS RT-PCR on the OSS sample is an accurate option for SARS-CoV-2 testing in children. A less intrusive technique for younger patients, who usually are tested frequently, might increase the number of patients tested.
Collapse
Affiliation(s)
- Cinta Moraleda
- Pediatric Infectious Diseases Unit. Department of Pediatrics, Hospital Universitario 12 de Octubre. Pediatric Research and Clinical Trials Unit (UPIC). RITIP (Translational Research Network in Pediatric Infectious Diseases), Madrid, Spain
- Instituto de Investigación 12 de Octubre (imas12), Fundación para la Investigación Biomédica del Hospital 12 de Octubre, Madrid, Spain
| | - Sara Domínguez-Rodríguez
- Instituto de Investigación 12 de Octubre (imas12), Fundación para la Investigación Biomédica del Hospital 12 de Octubre, Madrid, Spain
| | - Juan Miguel Mesa
- Paediatrics Department. Pediatrics Research Group, Hospital Universitario Infanta Sofia, San Sebastian de los Reyes, Madrid, Spain
| | - Paula García-Sánchez
- Emergency Pediatric Department. Instituto Investigación Hospital La Paz (IDIPaz), Hospital Universitario La Paz, Madrid, Spain
| | - María de la Serna
- Paediatrics Department. Pediatrics Research Group, Hospital Universitario Infanta Sofia, San Sebastian de los Reyes, Madrid, Spain
| | - José Antonio Alonso-Cadenas
- Emergency Pediatric Department. Instituto Investigación La Princesa, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Amanda Bermejo
- Pediatric Department, Hospital Universitario de Móstoles, Móstoles, Madrid, Spain
| | - Gema Sabrido
- Pediatric Department, Hospital Universitario Rey Juan Carlos, Móstoles, Madrid, Spain
| | - Leticia Martínez-Campos
- Pediatric Infectious Diseases, Materno Infantil. Hospital Universitario Torrecárdenas, Almeria, Spain
| | | | - Marta Illán-Ramos
- Pediatrics Department, Hospital Universitario Clínico San Carlos, Madrid, Spain
| | - Elena Cobos-Carrascosa
- Instituto de Investigación 12 de Octubre (imas12), Fundación para la Investigación Biomédica del Hospital 12 de Octubre, Madrid, Spain
| | - Álvaro Ballesteros
- Instituto de Investigación 12 de Octubre (imas12), Fundación para la Investigación Biomédica del Hospital 12 de Octubre, Madrid, Spain
| | - Juan Carlos Galán
- Microbiology Department, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS). Centro de Investigación Biomédica en Red en Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Francisco Llorente
- Centro de Investigacion en Sanidad Animal INIA-CSIC, Valdeolmos, Madrid, Spain
| | - David Aguilera-Alonso
- Pediatric Infectious Diseases Unit. Department of Pediatrics, Hospital Universitario Gregorio Marañón. Unidad de Investigación Maternoinfantil Fundación Familia Alonso (UDIMIFFA), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM). CIBER en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Ana Belén Jiménez
- Pediatrics Department, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - Pilar Catalán
- Servicio de Microbiologia Clinica y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Elias Dahdouh
- Clinical Microbiology and Parasitology Department, Hospital Universitario La Paz. IdiPAZ, Madrid, Spain
| | - Ignacio Navarro
- Paediatrics Department. Pediatrics Research Group, Hospital Universitario Infanta Sofia, San Sebastian de los Reyes, Madrid, Spain
| | | | - Pablo Mendoza
- Microbiology Department, Hospital Universitario de Móstoles, Mostoles, Madrid, Spain
| | - Concepción Pérez-Jorge
- Microbiology Department, Hospital Universitario Rey Juan Carlos, Mostoles, Madrid, Spain
| | | | - Daniel Blázquez-Gamero
- Pediatric Infectious Diseases Unit. Department of Pediatrics, Hospital Universitario 12 de Octubre. Pediatric Research and Clinical Trials Unit (UPIC). RITIP (Translational Research Network in Pediatric Infectious Diseases), Madrid, Spain
- Instituto de Investigación 12 de Octubre (imas12), Fundación para la Investigación Biomédica del Hospital 12 de Octubre, Madrid, Spain
- Universidad Complutense, Madrid, Spain
| | - Gonzalo Rivas
- Microbiology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Elena Sáez
- Microbiology Department, UR Salud. Hospital Universitario Infanta Sofía. Paediatrics Research Group, San Sebastian de los Reyes, Madrid, Spain
| | | | - Jesús Lucas-Fernández
- Microbiology Department, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS). Centro de Investigación Biomédica en Red en Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Elisa Pérez-Ramírez
- Centro de Investigacion en Sanidad Animal INIA-CSIC, Valdeolmos, Madrid, Spain
| | - Paloma Merino
- Microbiology Department, Hospital Universitario Clínico San Carlos, Madrid, Spain
| | - Sandra Miragaya
- Clinic Analysis Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Jorge Lorente
- Emergency Pediatric Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Irene Iglesias
- Centro de Investigacion en Sanidad Animal INIA-CSIC, Valdeolmos, Madrid, Spain
| | - Alfredo Tagarro
- Instituto de Investigación 12 de Octubre (imas12), Fundación para la Investigación Biomédica del Hospital 12 de Octubre, Madrid, Spain
- Paediatrics Department. Pediatrics Research Group, Hospital Universitario Infanta Sofia, San Sebastian de los Reyes, Madrid, Spain
| |
Collapse
|
231
|
梁 潇, 杜 信, 周 学. [Latest Research Findings on Health Management Based on Saliva Testing]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2022; 53:1110-1117. [PMID: 36443061 PMCID: PMC10408987 DOI: 10.12182/20221160510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Indexed: 06/16/2023]
Abstract
Being one of the most important exocrine fluids of the human body, saliva can reflect the health status of the body. Soliva collection has various advantages--it is simple, painless, fast, and safe, and soliva can be collected multiple times a day. Therefore, it has been gradually applied in the exploration for biomarkers for disease detection, providing a basis for the monitoring of the course of diseases, medication monitoring, and efficacy evaluation. We should implement health management based on saliva testing, collect the medical data of the healthy and diseased individuals and monitor their whole life cycle health, perform clinical cohort study, aggregate the data on platforms for big data on health and medicine, manage and provide guidance for the health status of populations, pinpoint the high-risk factors for pathogenesis, and provide effective intervention, early warning, and assessment of the vital signs of individuals, thereby reinforcing health management of the whole life cycle and safeguarding people's health in an all-round way. In addition, it drives the development of the health industry and bears strategic significance for the promotion of national economic development. It is becoming a hot research topic promising great potential and impressive applicational prospects. Herein, we reviewed new techniques for clinical saliva testing and health management based on saliva testing.
Collapse
Affiliation(s)
- 潇月 梁
- 口腔疾病研究国家重点实验室 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 牙体牙髓病科 (成都 610041)State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - 信眉 杜
- 口腔疾病研究国家重点实验室 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 牙体牙髓病科 (成都 610041)State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - 学东 周
- 口腔疾病研究国家重点实验室 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 牙体牙髓病科 (成都 610041)State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
232
|
Wang Y, Upadhyay A, Pillai S, Khayambashi P, Tran SD. Saliva as a diagnostic specimen for SARS-CoV-2 detection: A scoping review. Oral Dis 2022; 28 Suppl 2:2362-2390. [PMID: 35445491 PMCID: PMC9115496 DOI: 10.1111/odi.14216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 03/22/2022] [Accepted: 04/12/2022] [Indexed: 12/03/2022]
Abstract
OBJECTIVES This scoping review aims to summarize the diagnostic value of saliva assessed from current studies that (1) compare its performance in reverse transcriptase-polymerase chain reaction testing to nasopharyngeal swabs, (2) evaluate its performance in rapid and point-of-care COVID-19 diagnostic tests, and (3) explore its use as a specimen for detecting anti-SARS-CoV-2 antibodies. MATERIALS AND METHODS A systematic search was performed on the following databases: Medline and Embase (Ovid), World Health Organization, Centers for Disease Control and Prevention, and Global Health (Ovid) from January 2019 to September 2021. Of the 657 publications identified from the searches, n = 146 articles were included in the final scoping review. RESULTS Our findings showcase that salivary samples exceed nasopharyngeal swabs in detecting SARS-CoV-2 using reverse transcriptase-polymerase chain reaction testing in several studies. A select number of rapid antigen and point-of-care tests from the literature were also identified capable of high detection rates using saliva. Moreover, anti-SARS-CoV-2 antibodies have been shown to be detectable in saliva through biochemical assays. CONCLUSION We highlight the potential of saliva as an all-rounded specimen in detecting SARS-CoV-2. However, future large-scale clinical studies will be needed to support its widespread use as a non-invasive clinical specimen for COVID-19 testing.
Collapse
|
233
|
Abavisani M, Rahimian K, Kodori M, Khayami R, Mollapour Sisakht M, Mahmanzar M, Meshkat Z. In silico analysis of the substitution mutations and evolutionary trends of the SARS-CoV-2 structural proteins in Asia. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:1299-1307. [PMID: 36474565 PMCID: PMC9699957 DOI: 10.22038/ijbms.2022.66649.14620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 10/01/2022] [Indexed: 12/23/2022]
Abstract
OBJECTIVES To address a highly mutable pathogen, mutations must be evaluated. SARS-CoV-2 involves changing infectivity, mortality, and treatment and vaccination susceptibility resulting from mutations. MATERIALS AND METHODS We investigated the Asian and worldwide samples of amino-acid sequences (AASs) for envelope (E), membrane (M), nucleocapsid (N), and spike (S) proteins from the announcement of the new coronavirus 2019 (COVID-19) up to January 2022. Sequence alignment to the Wuhan-2019 virus permits tracking mutations in Asian and global samples. Furthermore, we explored the evolutionary tendencies of structural protein mutations and compared the results between Asia and the globe. RESULTS The mutation analyses indicated that 5.81%, 70.63%, 26.59%, and 3.36% of Asian S, E, M, and N samples did not display any mutation. Additionally, the most relative mutations among the S, E, M, and N AASs occurred in the regions of 508 to 635 AA, 7 to 14 AA, 66 to 88 AA, and 164 to 205 AA in both Asian and total samples. D614G, T9I, I82T, and R203M were inferred as the most frequent mutations in S, E, M, and N AASs. Timeline research showed that substitution mutation in the location of 614 among Asian and total S AASs was detected from January 2020. CONCLUSION N protein was the most non-conserved protein, and the most prevalent mutations in S, E, M, and N AASs were D614G, T9I, I82T, and R203M. Screening structural protein mutations is a robust approach for developing drugs, vaccines, and more specific diagnostic tools.
Collapse
Affiliation(s)
- Mohammad Abavisani
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran,Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Karim Rahimian
- Bioinformatics and Computational Omics Lab (BioCOOL), Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University
| | | | - Reza Khayami
- Non communicable Diseases Research Center, Bam University of Medical sciences, Bam, Iran
| | - Mahsa Mollapour Sisakht
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammadamin Mahmanzar
- Department of Biochemistry, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Zahra Meshkat
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran ,Corresponding author: Zahra Meshkat. Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran. Tel: +98-51-38002313; +98-51-38012453;
| |
Collapse
|
234
|
Barbato L, Bernardelli F, Braga G, Clementini M, Di Gioia C, Littarru C, Oreglia F, Raspini M, Brambilla E, Iavicoli I, Pinchi V, Landi L, Sforza NM, Cavalcanti R, Crea A, Cairo F. Surface disinfection and protective masks for SARS-CoV-2 and other respiratory viruses: A review by SIdP COVID-19 task force. Oral Dis 2022; 28 Suppl 2:2317-2325. [PMID: 32946152 PMCID: PMC7646272 DOI: 10.1111/odi.13646] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/04/2020] [Accepted: 09/06/2020] [Indexed: 01/04/2023]
Abstract
OBJECTIVES Primary focused question for this systematic review (SR) was "Which is the evidence about surfaces decontamination and protection masks for SARS-Cov-2 in dental practice?" Secondary question was "Which is the evidence about surfaces decontamination and protection masks against airborne pathogens and directly transmitted viral pathogens causing respiratory infections?" MATERIALS AND METHODS PRISMA guidelines were used. Studies on surface decontamination and protective masks for SARS-CoV-2 in dental practice were considered. Studies on other respiratory viruses were considered for the secondary question. RESULTS No studies are available for SARS-CoV-2. Four studies on surface disinfection against respiratory viruses were included. Ethanol 70% and sodium hypochlorite 0,5% seem to be effective in reducing infectivity by > 3log TCID. Four RCTs compared different types of masks on HCW. The single studies reported no difference for laboratory-diagnosed influenza, laboratory-diagnosed respiratory infection, and influenza-like illness. A meta-analysis was not considered appropriate. CONCLUSIONS There is lack of evidence on the efficacy of surface disinfection and protective masks to reduce the spread of SARS-CoV-2 or other respiratory viruses in dentistry. However, the consistent use of respirator and routine surface disinfection is strongly suggested. There is urgent need of data on the efficacy of specific protection protocols for dental HCW against viral infections.
Collapse
Affiliation(s)
- Luigi Barbato
- Research Unit in Periodontology and Periodontal MedicineDepartment of Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| | | | | | - Marco Clementini
- Department of PeriodontologyUniversità Vita‐Salute San RaffaeleMilanoItaly
| | | | | | | | | | - Eugenio Brambilla
- Department of BiomedicalSurgical and Dental SciencesUniversity of MilanMilanItaly
| | - Ivo Iavicoli
- Section of Occupational MedicineDepartment of Public HealthUniversity of Naples Federico IINaplesItaly
| | - Vilma Pinchi
- Department of Health SciencesSection of Medical Forensic SciencesUniversity of FlorenceFlorenceItaly
| | - Luca Landi
- Private PracticeRomeItaly
- SIdPFlorenceItaly
| | | | | | | | - Francesco Cairo
- SIdPFlorenceItaly
- Research Unit in Periodontology and Periodontal MedicineDepartment of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| |
Collapse
|
235
|
Aleissa MS, AL-Zharani M, Hasnain MS, Alkahtani S. Screening, molecular simulation & in silico kinetics of virtually designed covid-19 main protease inhibitors. JOURNAL OF KING SAUD UNIVERSITY - SCIENCE 2022; 34:102283. [PMID: 36062199 PMCID: PMC9428119 DOI: 10.1016/j.jksus.2022.102283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/27/2022] [Accepted: 08/13/2022] [Indexed: 11/07/2022]
Abstract
Coronavirus (covid-19) infection is considered to be deadliest ever pandemic experienced by the human being. It has very badly affected the socio-economic health of human and stuck the scientific community to think and rethink about its complete eradication. But due to no effective treatment or unavailability of vaccine the health professional could not show any significant improvement to control the pandemic. The situation needs newer molecule, vaccine or effective treatment to control covid-19 infection. Different target in viruses has been explored and proteases enzymes were found to be therapeutically effective target for the design of potential anti-covid-19 molecule as it plays the vital role in viral replication and assembly. Structure-based drug design was employed to discover the small molecule of anti-covid-19. Here we considered the small library of naturally occurring polyphenolic compounds and molecular docking, Molecular dynamics (MD) simulations, free binding energy calculation and in-silico ADME calculations to identify the newer HITs. Based upon their score the two molecules were identified as promising candidate. The docking scores were found to be −7.643 and −7.065 for the HIT1 and HIT-2 respectively. In MD simulations study the RMSD values were found to be 4.3 Å & 4.9 Å respectively. To validate these results MM-GBSA was performed and their binding free energies were computationally determined. The prime energy values of identified HITs (−13412.45 & −13441.8 kJ/mole) were found to be very close proximity to reference molecule (−13493.05 kJ/mole). Then in-silico ADME calculations were performed to calculate the drug likeliness identified HITs. BY considering all the values comparative to reference molecule and obtained in-silico pharmacokinetic properties of identified HITs we can suggest that HIT-1 and HIT-2 would be the most promising molecules that can inhibit the main protease enzyme of covid-19. These two molecules would become the potential drug candidate for the treatment of covid-19 infections.
Collapse
|
236
|
Moser C, Li JZ, Eron JJ, Aga E, Daar ES, Wohl DA, Coombs RW, Javan AC, Bender Ignacio RA, Jagannathan P, Ritz J, Sieg SF, Parikh UM, Hughes MD, Currier JS, Smith DM, Chew KW. Predictors of SARS-CoV-2 RNA From Nasopharyngeal Swabs and Concordance With Other Compartments in Nonhospitalized Adults With Mild to Moderate COVID-19. Open Forum Infect Dis 2022; 9:ofac618. [PMID: 36467293 PMCID: PMC9709705 DOI: 10.1093/ofid/ofac618] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022] Open
Abstract
Background Identifying characteristics associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA shedding may be useful to understand viral compartmentalization, disease pathogenesis, and risks for viral transmission. Methods Participants were enrolled August 2020 to February 2021 in ACTIV-2/A5401, a placebo-controlled platform trial evaluating investigational therapies for mild-to-moderate coronavirus disease 2019 (COVID-19), and underwent quantitative SARS-CoV-2 RNA testing on nasopharyngeal and anterior nasal swabs, oral wash/saliva, and plasma at entry (day 0, pretreatment) and days 3, 7, 14, and 28. Concordance of RNA levels (copies/mL) across compartments and predictors of nasopharyngeal RNA levels were assessed at entry (n = 537). Predictors of changes over time were evaluated among placebo recipients (n = 265) with censored linear regression models. Results Nasopharyngeal and anterior nasal RNA levels at study entry were highly correlated (r = 0.84); higher levels of both were associated with greater detection of RNA in plasma and oral wash/saliva. Older age, White non-Hispanic race/ethnicity, lower body mass index (BMI), SARS-CoV-2 immunoglobulin G seronegativity, and shorter prior symptom duration were associated with higher nasopharyngeal RNA at entry. In adjusted models, body mass index and race/ethnicity associations were attenuated, but the association with age remained (for every 10 years older, mean nasopharyngeal RNA was 0.27 log10 copies/mL higher; P < .001). Examining longitudinal viral RNA levels among placebo recipients, women had faster declines in nasopharyngeal RNA than men (mean change, -2.0 vs -1.3 log10 copies/mL, entry to day 3; P < .001). Conclusions SARS-CoV-2 RNA shedding was concordant across compartments. Age was strongly associated with viral shedding, and men had slower viral clearance than women, which could explain sex differences in acute COVID-19 outcomes.
Collapse
Affiliation(s)
- Carlee Moser
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Jonathan Z Li
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Joseph J Eron
- Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Evgenia Aga
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Eric S Daar
- Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California, USA
| | - David A Wohl
- Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Robert W Coombs
- Department of Laboratory Medicine and Pathology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | | | - Rachel A Bender Ignacio
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, Washington, USA
| | | | - Justin Ritz
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Scott F Sieg
- Department of Medicine, Case Western University, Cleveland, Ohio, USA
| | - Urvi M Parikh
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael D Hughes
- Department of Biostatistics and Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Judith S Currier
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, USA
| | - Davey M Smith
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Kara W Chew
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
237
|
Olmstead AD, Nikiforuk AM, Schwartz S, Márquez AC, Valadbeigy T, Flores E, Saran M, Goldfarb DM, Hayden A, Masud S, Russell SL, Prystajecky N, Jassem AN, Morshed M, Sekirov I. Characterizing Longitudinal Antibody Responses in Recovered Individuals Following COVID-19 Infection and Single-Dose Vaccination: A Prospective Cohort Study. Viruses 2022; 14:2416. [PMID: 36366515 PMCID: PMC9694471 DOI: 10.3390/v14112416] [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: 09/20/2022] [Revised: 10/22/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Investigating antibody titers in individuals who have been both naturally infected with SARS-CoV-2 and vaccinated can provide insight into antibody dynamics and correlates of protection over time. METHODS Human coronavirus (HCoV) IgG antibodies were measured longitudinally in a prospective cohort of qPCR-confirmed, COVID-19 recovered individuals (k = 57) in British Columbia pre- and post-vaccination. SARS-CoV-2 and endemic HCoV antibodies were measured in serum collected between Nov. 2020 and Sept. 2021 (n = 341). Primary analysis used a linear mixed-effects model to understand the effect of single dose vaccination on antibody concentrations adjusting for biological sex, age, time from infection and vaccination. Secondary analysis investigated the cumulative incidence of high SARS-CoV-2 anti-spike IgG seroreactivity equal to or greater than 5.5 log10 AU/mL up to 105 days post-vaccination. No re-infections were detected in vaccinated participants, post-vaccination by qPCR performed on self-collected nasopharyngeal specimens. RESULTS Bivariate analysis (complete data for 42 participants, 270 samples over 472 days) found SARS-CoV-2 spike and RBD antibodies increased 14-56 days post-vaccination (p < 0.001) and vaccination prevented waning (regression coefficient, B = 1.66 [95%CI: 1.45-3.46]); while decline of nucleocapsid antibodies over time was observed (regression coefficient, B = -0.24 [95%CI: -1.2-(-0.12)]). A positive association was found between COVID-19 vaccination and endemic human β-coronavirus IgG titer 14-56 days post vaccination (OC43, p = 0.02 & HKU1, p = 0.02). On average, SARS-CoV-2 anti-spike IgG concentration increased in participants who received one vaccine dose by 2.06 log10 AU/mL (95%CI: 1.45-3.46) adjusting for age, biological sex, and time since infection. Cumulative incidence of high SARS-CoV-2 spike antibodies (>5.5 log10 AU/mL) was 83% greater in vaccinated compared to unvaccinated individuals. CONCLUSIONS Our study confirms that vaccination post-SARS-CoV-2 infection provides multiple benefits, such as increasing anti-spike IgG titers and preventing decay up to 85 days post-vaccination.
Collapse
Affiliation(s)
- Andrea D. Olmstead
- Department of Pathology and Laboratory Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC V6T 1Z7, Canada
- British Columbia Centre for Disease Control Public Health Laboratory, Provincial Health Services Authority, 655 West 12th Ave, Vancouver, BC V5Z 4R4, Canada
| | - Aidan M. Nikiforuk
- British Columbia Centre for Disease Control Public Health Laboratory, Provincial Health Services Authority, 655 West 12th Ave, Vancouver, BC V5Z 4R4, Canada
- School of Population and Public Health, University of British Columbia, 2206 E Mall, Vancouver, BC V6T 1Z3, Canada
| | - Sydney Schwartz
- British Columbia Centre for Disease Control Public Health Laboratory, Provincial Health Services Authority, 655 West 12th Ave, Vancouver, BC V5Z 4R4, Canada
| | - Ana Citlali Márquez
- British Columbia Centre for Disease Control Public Health Laboratory, Provincial Health Services Authority, 655 West 12th Ave, Vancouver, BC V5Z 4R4, Canada
| | - Tahereh Valadbeigy
- British Columbia Centre for Disease Control Public Health Laboratory, Provincial Health Services Authority, 655 West 12th Ave, Vancouver, BC V5Z 4R4, Canada
| | - Eri Flores
- British Columbia Centre for Disease Control Public Health Laboratory, Provincial Health Services Authority, 655 West 12th Ave, Vancouver, BC V5Z 4R4, Canada
| | - Monika Saran
- Department of Pathology and Laboratory Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC V6T 1Z7, Canada
| | - David M. Goldfarb
- Department of Pathology and Laboratory Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC V6T 1Z7, Canada
- Department of Pathology and Laboratory Medicine, British Columbia Children’s and Women’s Hospital, 4500 Oak Street, Vancouver, BC V6H 3N1, Canada
| | - Althea Hayden
- Office of the Chief Medical Health Officer, Vancouver Coastal Health, Vancouver, BC V5Z 4C2, Canada
| | - Shazia Masud
- Department of Pathology and Laboratory Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC V6T 1Z7, Canada
- Department of Pathology and Laboratory Medicine, Surrey Memorial Hospital, Surrey, BC V3V 1Z2, Canada
| | - Shannon L. Russell
- Department of Pathology and Laboratory Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC V6T 1Z7, Canada
- British Columbia Centre for Disease Control Public Health Laboratory, Provincial Health Services Authority, 655 West 12th Ave, Vancouver, BC V5Z 4R4, Canada
| | - Natalie Prystajecky
- Department of Pathology and Laboratory Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC V6T 1Z7, Canada
- British Columbia Centre for Disease Control Public Health Laboratory, Provincial Health Services Authority, 655 West 12th Ave, Vancouver, BC V5Z 4R4, Canada
| | - Agatha N. Jassem
- Department of Pathology and Laboratory Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC V6T 1Z7, Canada
- British Columbia Centre for Disease Control Public Health Laboratory, Provincial Health Services Authority, 655 West 12th Ave, Vancouver, BC V5Z 4R4, Canada
| | - Muhammad Morshed
- Department of Pathology and Laboratory Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC V6T 1Z7, Canada
- British Columbia Centre for Disease Control Public Health Laboratory, Provincial Health Services Authority, 655 West 12th Ave, Vancouver, BC V5Z 4R4, Canada
| | - Inna Sekirov
- Department of Pathology and Laboratory Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC V6T 1Z7, Canada
- British Columbia Centre for Disease Control Public Health Laboratory, Provincial Health Services Authority, 655 West 12th Ave, Vancouver, BC V5Z 4R4, Canada
| |
Collapse
|
238
|
Chen S, Murphy EA, Pendergrass AG, Sukhu AC, Eng D, Jurkiewicz M, Mohammed I, Rand S, White LJ, Hupert N, Yang YJ. Estimating the Effectiveness of Shielding during Pregnancy against SARS-CoV-2 in New York City during the First Year of the COVID-19 Pandemic. Viruses 2022; 14:2408. [PMID: 36366506 PMCID: PMC9697040 DOI: 10.3390/v14112408] [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: 09/27/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 01/31/2023] Open
Abstract
Pregnant patients have increased morbidity and mortality in the setting of SARS-CoV-2 infection. The exposure of pregnant patients in New York City to SARS-CoV-2 is not well understood due to early lack of access to testing and the presence of asymptomatic COVID-19 infections. Before the availability of vaccinations, preventative (shielding) measures, including but not limited to wearing a mask and quarantining at home to limit contact, were recommended for pregnant patients. Using universal testing data from 2196 patients who gave birth from April through December 2020 from one institution in New York City, and in comparison, with infection data of the general population in New York City, we estimated the exposure and real-world effectiveness of shielding in pregnant patients. Our Bayesian model shows that patients already pregnant at the onset of the pandemic had a 50% decrease in exposure compared to those who became pregnant after the onset of the pandemic and to the general population.
Collapse
Affiliation(s)
- Siyu Chen
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX1 3QD, UK
| | - Elisabeth A. Murphy
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | | | - Ashley C. Sukhu
- New York Presbyterian-Weill Cornell Medicine, New York, NY 10065, USA
| | - Dorothy Eng
- New York Presbyterian-Weill Cornell Medicine, New York, NY 10065, USA
| | | | - Iman Mohammed
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Sophie Rand
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lisa J. White
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX1 3QD, UK
| | - Nathaniel Hupert
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX1 3QD, UK
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yawei J. Yang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- New York Presbyterian-Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
239
|
Kombe Kombe AJ, Biteghe FAN, Ndoutoume ZN, Jin T. CD8 + T-cell immune escape by SARS-CoV-2 variants of concern. Front Immunol 2022; 13:962079. [PMID: 36389664 PMCID: PMC9647062 DOI: 10.3389/fimmu.2022.962079] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 10/03/2022] [Indexed: 07/30/2023] Open
Abstract
Despite the efficacy of antiviral drug repositioning, convalescent plasma (CP), and the currently available vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the worldwide coronavirus disease 2019 (COVID-19) pandemic is still challenging because of the ongoing emergence of certain new SARS-CoV-2 strains known as variants of concern (VOCs). Mutations occurring within the viral genome, characterized by these new emerging VOCs, confer on them the ability to efficiently resist and escape natural and vaccine-induced humoral and cellular immune responses. Consequently, these VOCs have enhanced infectivity, increasing their stable spread in a given population with an important fatality rate. While the humoral immune escape process is well documented, the evasion mechanisms of VOCs from cellular immunity are not well elaborated. In this review, we discussed how SARS-CoV-2 VOCs adapt inside host cells and escape anti-COVID-19 cellular immunity, focusing on the effect of specific SARS-CoV-2 mutations in hampering the activation of CD8+ T-cell immunity.
Collapse
Affiliation(s)
- Arnaud John Kombe Kombe
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | | | - Zélia Nelly Ndoutoume
- The Second Clinical School, Medical Imaging, Chongqing Medical University, Chongqing, China
| | - Tengchuan Jin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Laboratory of Structural Immunology, Chinese Academic of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Chinese Academic of Sciences (CAS) Center for Excellence in Molecular Cell Science, Chinese Academy of Science, Shanghai, China
| |
Collapse
|
240
|
Gouvea MDPG, Moulaz IR, Gouveia TM, Lança KEM, Lacerda BSDP, Thompson BP, Polese J, de Lima MD, Ribeiro-Rodrigues R, Mill JG, Valim V. Anti-SARS-CoV-2 antibody immunoreactivity profiles during COVID-19 recurrence. Rev Soc Bras Med Trop 2022; 55:e01062022. [PMID: 36287469 PMCID: PMC9592100 DOI: 10.1590/0037-8682-0106-2022] [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: 03/03/2022] [Accepted: 09/09/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND This study aimed to evaluate IgG and IgM levels in COVID-19 recurrence. METHODS The serum antibody levels and clinical data from 73 healthcare workers with SARS-CoV-2 divided into seroconverted (n=51) and non-seroconverted (n=22) groups were assessed. The presence of specific anti-nucleocapsid (anti-N) IgM and IgG for SARS-CoV-2 was evaluated. IgG antibodies to the SARS-CoV-2 spike receptor-binding domain were used to confirm non-seroconversion in all negative anti-N. RESULTS Four recurrent cases displayed mild symptoms and were non-seroconverted until the recurrence of symptoms. CONCLUSIONS Undetectable anti-nucleocapsid IgM and IgG levels may be correlated with symptomatic COVID-19 recurrence.
Collapse
Affiliation(s)
| | - Isac Ribeiro Moulaz
- Universidade Federal do Espírito Santo, Centro de Ciências da Saúde, Vitória, ES, Brasil
| | - Thayná Martins Gouveia
- Universidade Federal do Espírito Santo, Centro de Ciências da Saúde, Vitória, ES, Brasil
| | | | | | - Beatriz Paoli Thompson
- Universidade Federal do Espírito Santo, Centro de Ciências da Saúde, Vitória, ES, Brasil
| | - Jéssica Polese
- Universidade Federal do Espírito Santo, Centro de Ciências da Saúde, Departamento de Pneumologia, Vitória, ES, Brasil
| | - Marina Deorce de Lima
- Universidade Federal do Espírito Santo, Centro de Ciências da Saúde, Vitória, ES, Brasil
| | - Rodrigo Ribeiro-Rodrigues
- Departamento de Saúde do Estado do Espírito Santo, Laboratório de Saúde Pública do Estado do Espírito Santo, Vitória, ES, Brasil., Universidade Federal do Espírito Santo, Centro de Ciências da Saúde, Centro de Doenças Infecciosas, Vitória, ES, Brasil
| | - José Geraldo Mill
- Universidade Federal do Espírito Santo, Centro de Ciências da Saúde, Departamento de Ciências Fisiológicas, Vitória, ES, Brasil
| | - Valéria Valim
- Universidade Federal do Espírito Santo, Hospital Universitário Cassiano Antonio Moraes, Divisão de Reumatologia, Vitória, ES, Brasil
| |
Collapse
|
241
|
Wang Q, Ma J, Zhang L, Xie L. Diagnostic performance of corona virus disease 2019 chest computer tomography image recognition based on deep learning: Systematic review and meta-analysis. Medicine (Baltimore) 2022; 101:e31346. [PMID: 36281129 PMCID: PMC9592148 DOI: 10.1097/md.0000000000031346] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND To analyze the diagnosis performance of deep learning model used in corona virus disease 2019 (COVID-19) computer tomography(CT) chest scans. The included sample contains healthy people, confirmed COVID-19 patients and unconfirmed suspected patients with corresponding symptoms. METHODS PubMed, Web of Science, Wiley, China National Knowledge Infrastructure, WAN FANG DATA, and Cochrane Library were searched for articles. Three researchers independently screened the literature, extracted the data. Any differences will be resolved by consulting the third author to ensure that a highly reliable and useful research paper is produced. Data were extracted from the final articles, including: authors, country of study, study type, sample size, participant demographics, type and name of AI software, results (accuracy, sensitivity, specificity, ROC, and predictive values), other outcome(s) if applicable. RESULTS Among the 3891 searched results, 32 articles describing 51,392 confirmed patients and 7686 non-infected individuals met the inclusion criteria. The pooled sensitivity, the pooled specificity, positive likelihood ratio, negative likelihood ratio and the pooled diagnostic odds ratio (OR) is 0.87(95%CI [confidence interval]: 0.85, 0.89), 0.85(95%CI: 0.82, 0.87), 6.7(95%CI: 5.7, 7.8), 0.14(95%CI: 0.12, 0.16), and 49(95%CI: 38, 65). Further, the AUROC (area under the receiver operating characteristic curve) is 0.94(95%CI: 0.91, 0.96). Secondary outcomes are specific sensitivity and specificity within subgroups defined by different models. Resnet has the best diagnostic performance, which has the highest sensitivity (0.91[95%CI: 0.87, 0.94]), specificity (0.90[95%CI: 0.86, 0.93]) and AUROC (0.96[95%CI: 0.94, 0.97]), according to the AUROC, we can get the rank Resnet > Densenet > VGG > Mobilenet > Inception > Effficient > Alexnet. CONCLUSIONS Our study findings show that deep learning models have immense potential in accurately stratifying COVID-19 patients and in correctly differentiating them from patients with other types of pneumonia and normal patients. Implementation of deep learning-based tools can assist radiologists in correctly and quickly detecting COVID-19 and, consequently, in combating the COVID-19 pandemic.
Collapse
Affiliation(s)
- Qiaolan Wang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- West China-PUMC C.C. Chen Institute of Health, Sichuan University, Chengdu, China
| | - Jingxuan Ma
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- West China-PUMC C.C. Chen Institute of Health, Sichuan University, Chengdu, China
| | - Luoning Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- West China-PUMC C.C. Chen Institute of Health, Sichuan University, Chengdu, China
| | - Linshen Xie
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- West China-PUMC C.C. Chen Institute of Health, Sichuan University, Chengdu, China
- *Correspondence: Linshen Xie, West China School of Public Health and West China Fourth Hospital, Sichuan University, 610041 Chengdu, China (e-mail: )
| |
Collapse
|
242
|
Alsalameh S, Alnajjar K, Makhzoum T, Al Eman N, Shakir I, Mir TA, Alkattan K, Chinnappan R, Yaqinuddin A. Advances in Biosensing Technologies for Diagnosis of COVID-19. BIOSENSORS 2022; 12:898. [PMID: 36291035 PMCID: PMC9599206 DOI: 10.3390/bios12100898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/11/2022] [Accepted: 10/14/2022] [Indexed: 06/16/2023]
Abstract
The COVID-19 pandemic has severely impacted normal human life worldwide. Due to its rapid community spread and high mortality statistics, the development of prompt diagnostic tests for a massive number of samples is essential. Currently used traditional methods are often expensive, time-consuming, laboratory-based, and unable to handle a large number of specimens in resource-limited settings. Because of its high contagiousness, efficient identification of SARS-CoV-2 carriers is crucial. As the advantages of adopting biosensors for efficient diagnosis of COVID-19 increase, this narrative review summarizes the recent advances and the respective reasons to consider applying biosensors. Biosensors are the most sensitive, specific, rapid, user-friendly tools having the potential to deliver point-of-care diagnostics beyond traditional standards. This review provides a brief introduction to conventional methods used for COVID-19 diagnosis and summarizes their advantages and disadvantages. It also discusses the pathogenesis of COVID-19, potential diagnostic biomarkers, and rapid diagnosis using biosensor technology. The current advancements in biosensing technologies, from academic research to commercial achievements, have been emphasized in recent publications. We covered a wide range of topics, including biomarker detection, viral genomes, viral proteins, immune responses to infection, and other potential proinflammatory biomolecules. Major challenges and prospects for future application in point-of-care settings are also highlighted.
Collapse
Affiliation(s)
| | - Khalid Alnajjar
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Tariq Makhzoum
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Noor Al Eman
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Ismail Shakir
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Tanveer Ahmad Mir
- Laboratory of Tissue/Organ Bioengineering and BioMEMS, Organ Transplant Centre of Excellence, Transplant Research and Innovation Department, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Khaled Alkattan
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Raja Chinnappan
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Ahmed Yaqinuddin
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
243
|
Gunawardana M, Webster S, Rivera S, Cortez JM, Breslin J, Pinales C, Buser C, Ibarrondo FJ, Yang OO, Bobardt M, Gallay PA, Adler AP, Ramirez CM, Anton PA, Baum MM. Early SARS-CoV-2 dynamics and immune responses in unvaccinated participants of an intensely sampled longitudinal surveillance study. COMMUNICATIONS MEDICINE 2022; 2:129. [PMID: 36238348 PMCID: PMC9553075 DOI: 10.1038/s43856-022-00195-4] [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: 03/10/2022] [Accepted: 09/29/2022] [Indexed: 11/08/2022] Open
Abstract
Background A comprehensive understanding of the SARS-CoV-2 infection dynamics and the ensuing host immune responses is needed to explain the pathogenesis as it relates to viral transmission. Knowledge gaps exist surrounding SARS-CoV-2 in vivo kinetics, particularly in the earliest stages after exposure. Methods An ongoing, workplace clinical surveillance study was used to intensely sample a small cohort longitudinally. Nine study participants who developed COVID-19 between November, 2020 and March, 2021 were monitored at high temporal resolution for three months in terms of viral loads as well as associated inflammatory biomarker and antibody responses. CD8 + T cells targeting SARS-CoV-2 in blood samples from study participants were evaluated. Results Here we show that the resulting datasets, supported by Bayesian modeling, allowed the underlying kinetic processes to be described, yielding a number of unexpected findings. Early viral replication is rapid (median doubling time, 3.1 h), providing a narrow window between exposure and viral shedding, while the clearance phase is slow and heterogeneous. Host immune responses different widely across participants. Conclusions Results from our small study give a rare insight into the life-cycle of COVID-19 infection and hold a number of important biological, clinical, and public health implications.
Collapse
Affiliation(s)
- Manjula Gunawardana
- Department of Chemistry, Oak Crest Institute of Science, 128–132W. Chestnut Ave., Monrovia, CA USA
| | - Simon Webster
- Department of Chemistry, Oak Crest Institute of Science, 128–132W. Chestnut Ave., Monrovia, CA USA
| | - Sofia Rivera
- Department of Chemistry, Oak Crest Institute of Science, 128–132W. Chestnut Ave., Monrovia, CA USA
| | - John M. Cortez
- Department of Chemistry, Oak Crest Institute of Science, 128–132W. Chestnut Ave., Monrovia, CA USA
| | - Jessica Breslin
- Department of Chemistry, Oak Crest Institute of Science, 128–132W. Chestnut Ave., Monrovia, CA USA
| | - Cristian Pinales
- Department of Chemistry, Oak Crest Institute of Science, 128–132W. Chestnut Ave., Monrovia, CA USA
| | - Christopher Buser
- Department of Chemistry, Oak Crest Institute of Science, 128–132W. Chestnut Ave., Monrovia, CA USA
| | - F. Javier Ibarrondo
- University of California, Los Angeles (UCLA), Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA USA
| | - Otto O. Yang
- University of California, Los Angeles (UCLA), Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA USA
- University of California, Los Angeles (UCLA), Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA USA
| | - Michael Bobardt
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA USA
| | - Philippe A. Gallay
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA USA
| | - Amy P. Adler
- Jumpstart Research Consulting, LLC, Santa Fe, NM USA
| | - Christina M. Ramirez
- University of California, Los Angeles (UCLA), Department of Biostatistics, Fielding School of Public Health, UCLA, Los Angeles, CA USA
| | - Peter A. Anton
- Department of Chemistry, Oak Crest Institute of Science, 128–132W. Chestnut Ave., Monrovia, CA USA
| | - Marc M. Baum
- Department of Chemistry, Oak Crest Institute of Science, 128–132W. Chestnut Ave., Monrovia, CA USA
| |
Collapse
|
244
|
Waites W, Cavaliere M, Danos V, Datta R, Eggo RM, Hallett TB, Manheim D, Panovska-Griffiths J, Russell TW, Zarnitsyna VI. Compositional modelling of immune response and virus transmission dynamics. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2022; 380:20210307. [PMID: 35965463 PMCID: PMC9376723 DOI: 10.1098/rsta.2021.0307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Transmission models for infectious diseases are typically formulated in terms of dynamics between individuals or groups with processes such as disease progression or recovery for each individual captured phenomenologically, without reference to underlying biological processes. Furthermore, the construction of these models is often monolithic: they do not allow one to readily modify the processes involved or include the new ones, or to combine models at different scales. We show how to construct a simple model of immune response to a respiratory virus and a model of transmission using an easily modifiable set of rules allowing further refining and merging the two models together. The immune response model reproduces the expected response curve of PCR testing for COVID-19 and implies a long-tailed distribution of infectiousness reflective of individual heterogeneity. This immune response model, when combined with a transmission model, reproduces the previously reported shift in the population distribution of viral loads along an epidemic trajectory. This article is part of the theme issue 'Technical challenges of modelling real-life epidemics and examples of overcoming these'.
Collapse
Affiliation(s)
- W. Waites
- Department of Computer and Information Sciences, University of Strathclyde, Glasgow, UK
- Centre for Mathematical Modelling of Infectious Disease, London School of Hygiene and Tropical Medicine, London, UK
| | - M. Cavaliere
- Department of Computing and Mathematics, Manchester Metropolitan University, Manchester, UK
| | - V. Danos
- Département d’Informatique, École Normale Supérieure, Paris, France
| | - R. Datta
- Datta Enterprises LLC, San Francisco, CA, USA
| | - R. M. Eggo
- Department of Computer and Information Sciences, University of Strathclyde, Glasgow, UK
| | - T. B. Hallett
- MRC Centre for Global Infectious Disease Analysis, Imperial College London, London, UK
| | - D. Manheim
- Technion, Israel Institute of Technology, Haifa, Israel
| | - J. Panovska-Griffiths
- The Big Data Institute and the Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- The Queen’s College, University of Oxford, Oxford, UK
| | - T. W. Russell
- Department of Computer and Information Sciences, University of Strathclyde, Glasgow, UK
| | - V. I. Zarnitsyna
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
245
|
Aljarba NH, Hasnain MS, Bin-Meferij MM, Alkahtani S. An in-silico investigation of potential natural polyphenols for the targeting of COVID main protease inhibitor. JOURNAL OF KING SAUD UNIVERSITY. SCIENCE 2022; 34:102214. [PMID: 35811756 PMCID: PMC9250415 DOI: 10.1016/j.jksus.2022.102214] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/07/2022] [Accepted: 06/28/2022] [Indexed: 05/04/2023]
Abstract
The deadliest recent pandemic outbreak of COVID-19 disease has severely damaged the socio-economic health of the people globally. Due to unavailability of any effective vaccine or treatment the human beings are still struggling to overcome the pandemic condition. In an attempt to discover anti-COVID molecule, we used in-silico approach and reported 160 natural polyphenols to identify the most promising druggable HITs that can further used for drug discovery process. The co-crystallized structure COVID protease enzyme (PDB id 6LU7) was used. HTVS, MD simulation, binding energy calculations and in-silico ADME calculation were done and analyzed. Depending upon the scores three compounds galangin, nalsudaldain and rhamnezine were identified and the docking score were found to be -7.704, -6.51, -4.212 respectively. These docked complexes were further subjected to MD simulation runs over a 100 ns time and the RMSD and RMSF values were determined. The RMSD values of three compounds were found to be 2.9 Å, 7.6 Å & 9.5 Å respectively and the lowest RMSF values suggested the steady stability of ligand-protein complexes. The binding free energies (ΔG) of compounds with protein were found to be -49.8, -56.45, -62.87 kJ/mole. Moreover, in-silico ADME calculations indicated the drug likeliness properties of these molecules. By considering all these in-silico results the identified HITs would be the most probable anti-COVID drug molecules that can be further taken in wet lab and can act as lead for development of newer inhibitor of COVID-19 main protease enzyme.
Collapse
Affiliation(s)
- Nada H Aljarba
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Md Saquib Hasnain
- Department of Pharmacy, Palamau Institute of Pharmacy, Chianki, Daltonganj 822102, Jharkhand, India
| | | | - Saad Alkahtani
- Department of Zoology, College of Science, King Saud University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
246
|
Sourander B, Andersson LM, Brink M, Yilmaz A, Sundell N, Marklund E, Nilsson S, Lindh M, Robertson J, Gisslén M. No effect of remdesivir or betamethasone on upper respiratory tract SARS-CoV-2 RNA kinetics in hospitalised COVID-19 patients: a retrospective observational study. Infect Dis (Lond) 2022; 54:703-712. [PMID: 35708280 DOI: 10.1080/23744235.2022.2081716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The viral kinetics of SARS-CoV-2 has been considered clinically important. While remdesivir and corticosteroids are recommended for COVID-19 patients requiring oxygen support, there is a limited number of published reports on viral kinetics in hospitalised patients with COVID-19 treated with remdesivir or corticosteroids. METHODS We conducted a retrospective study by collecting longitudinal samples from the nasopharynx/throat of 123 hospitalised patients (median age 55 years, 74% male) with COVID-19, to evaluate the effects of remdesivir and corticosteroid treatment on viral RNA levels. The subjects were divided into four groups: those receiving remdesivir (n = 25), betamethasone (n = 41), both (n = 15), or neither (n = 42). Time to viral RNA clearance was analysed using Kaplan-Meier plots, categorical data were analysed using Fisher's exact test, and Kruskal-Wallis for continuous data. Viral RNA decline rate was analysed using a mixed effect model. RESULTS We found no significant difference in SARS-CoV-2 RNA decline rate or time to SARS-CoV-2 RNA clearance between the groups. Moreover, clinical status at baseline was not correlated with time to viral clearance. CONCLUSIONS Since SARS-CoV-2 RNA kinetics was not affected by treatment, repeated sampling from the upper respiratory tract cannot be used to evaluate treatment response.
Collapse
Affiliation(s)
- Birger Sourander
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Infectious Diseases, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lars-Magnus Andersson
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Infectious Diseases, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Magnus Brink
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Infectious Diseases, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Aylin Yilmaz
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Infectious Diseases, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Nicklas Sundell
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Infectious Diseases, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Emelie Marklund
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Infectious Diseases, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Staffan Nilsson
- Mathematical Sciences, Chalmers University of Technology, Gothenburg, Sweden
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Magnus Lindh
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Josefina Robertson
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Infectious Diseases, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Magnus Gisslén
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Infectious Diseases, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
247
|
Avuçlu E. COVID-19 detection using X-ray images and statistical measurements. MEASUREMENT : JOURNAL OF THE INTERNATIONAL MEASUREMENT CONFEDERATION 2022; 201:111702. [PMID: 35942188 PMCID: PMC9349030 DOI: 10.1016/j.measurement.2022.111702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 07/14/2022] [Accepted: 07/30/2022] [Indexed: 06/15/2023]
Abstract
The COVID-19 pandemic spread all over the world, starting in China in late 2019, and significantly affected life in all aspects. As seen in SARS, MERS, COVID-19 outbreaks, coronaviruses pose a great threat to world health. The COVID-19 epidemic, which caused pandemics all over the world, continues to seriously threaten people's lives. Due to the rapid spread of COVID-19, many countries' healthcare sectors were caught off guard. This situation put a burden on doctors and healthcare professionals that they could not handle. All of the studies on COVID-19 in the literature have been done to help experts to recognize COVID-19 more accurately, to use more accurate diagnosis and appropriate treatment methods. The alleviation of this workload will be possible by developing computer aided early and accurate diagnosis systems with machine learning. Diagnosis and evaluation of pneumonia on computed tomography images provide significant benefits in investigating possible complications and in case follow-up. Pneumonia and lesions occurring in the lungs should be carefully examined as it helps in the diagnostic process during the pandemic period. For this reason, the first diagnosis and medications are very important to prevent the disease from progressing. In this study, a dataset consisting of Pneumonia and Normal images was used by proposing a new image preprocessing process. These preprocessed images were reduced to 15x15 unit size and their features were extracted according to their RGB values. Experimental studies were carried out by performing both normal values and feature reduction among these features. RGB values of the images were used in train and test processes for MLAs. In experimental studies, 5 different Machine Learning Algorithms (MLAs) (Multi Class Support Vector Machine (MC-SVM), k Nearest Neighbor (k-NN), Decision Tree (DT), Multinominal Logistic Regression (MLR), Naive Bayes (NB)). The following accuracy rates were obtained in train operations for MLAs, respectively; 1, 1, 1, 0.746377, 0.963768. Accuracy results in test operations were obtained as follows; 0.87755, 0.857143, 0.857143, 0.877551, 0.938776.
Collapse
Affiliation(s)
- Emre Avuçlu
- Department of Software Engineering, Faculty of Engineering, Aksaray University, Aksaray TURKEY
| |
Collapse
|
248
|
Dzananovic B, Williamson M, Nwaigwe C, Routray C. Clinical significance of anti-nucleocapsid-IgG sero-positivity in SARS-CoV-2 infection in hospitalized patients in North Dakota. World J Clin Infect Dis 2022; 12:50-60. [DOI: 10.5495/wjcid.v12.i2.50] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 08/02/2022] [Accepted: 09/21/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND During the peak of the coronavirus diseases 2019 (COVID-19) pandemic, clinicians actively studied the utility of various epidemiologic-clinical parameters to determine the prognosis for patients hospitalized with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Serum IgG antibody level, D-Dimer, C-reactive protein and neutrophil to lymphocyte ratio, etc. were studied to assess their association with the clinical course in hospitalized patients and predict who may be at increased risk for poor clinical outcome. However, the influence of SARS-CoV-2-anti-nucleocapsid-IgG antibody (IgG-N) sero-positivity on the clinical outcome of patients with COVID-19 is largely unknown.
AIM To study the influence of SARS-CoV-2 anti-nucleocapsid-IgG seropositivity on clinical course and diseases severity in hospitalized COVID-19 patients.
METHODS We conducted a retrospective study of adults admitted to a tertiary care community hospital in North Dakota with COVID-19. Included patients had severe COVID-19 disease or worse and so required supplemental oxygen on admission. They were serologically tested for SARS-CoV-2-anti-nuceocapsid-IgG (IgG-N). The IgG-N positive group were 26 patients and the IgG-N negative group had 33 patients. The groups received similar treatment for COVID-19 as approved by our healthcare system from Day 1 of admission until discharge or death. Measurable parameters for monitoring the patients’ clinical course included the following: Length of hospitalization (LOS), use of high flow nasal canula (HFNC), use of noninvasive bilevel positive pressure ventilation (BiPAP), admission into the intensive care unit, need for mechanical ventilation (VENT); and the patient outcome/discharge or death. Other variables included were age, gender and body-mass-index, and duration of symptoms before presentation. For each variable, the outcome was modeled as a function of SARS-CoV-2-IgG-N status (positive or negative) using a generalized linear model. For LOS-days, a negative binomial distribution was used as it had a better fit than a Poisson or Gaussian distribution as evidenced by a Pearson chi-square/df value closer to 1.0. All other outcomes utilized a binary logistic regression model.
RESULTS After a thorough examination of patient data, it was found that admission rates to the Intensive Care Unit, as well as the usage of BiPAP, HFNC and VENT support, in conjunction with patient outcomes, were not significantly different across IgG-N status. However, the LOS variable when assessed by IgG-N status was found to be significant (t value = 2.16, P value = 0.0349). IgG-N negative patients had higher than average LOS in comparison to IgG-N positive patients (15.12 vs 9.35 d). Even when removing the extreme value (an LOS of 158 d), IgG-N negative patients still had slightly higher than average stays (10.66 vs 9.35 d) but the relationship was no longer significant. For patient outcome/death, only age (numerical) was a significant predictor (F value = 4.66, P value = 0.0352). No other variables for any of the outcomes were significant predictors of clinical course or disease severity.
CONCLUSION Our study demonstrated that IgG-N seroconversion had no significant association with clinical outcomes in hospitalized COVID-19 patients.
Collapse
Affiliation(s)
- Bakir Dzananovic
- Department of Medicine, Idaho College of Osteopathic Medicine, Meridian, ID 83642, United States
| | - Mark Williamson
- Department of Biostatistics, Epidemiology and Research Design Core, University of North Dakota, Grand Forks, ND 58202, United States
| | - Casmiar Nwaigwe
- Department of Internal Medicine, University of North Dakota School of Medicine and Health Sciences, Minot, ND 58701, United States
| | - Chittaranjan Routray
- Family Medicine, University of North Dakota School of Medicine and Health Sciences, Minot, ND 58701, United States
- Department of Internal Medicine, Trinity Health, Minot, ND 58701, United States
| |
Collapse
|
249
|
Salu OB, Akase IE, Anyanwu RA, Orenolu MR, Abdullah MA, Giwa-Tubosun T, Oloko SA, Oshinjo AM, Abiola AA, Oyedeji KS, Omilabu SA. Saliva sample for detection of SARS-CoV-2: A possible alternative for mass testing. PLoS One 2022; 17:e0275201. [PMID: 36170269 PMCID: PMC9518879 DOI: 10.1371/journal.pone.0275201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 09/12/2022] [Indexed: 11/19/2022] Open
Abstract
Molecular diagnostic testing has played a critical role in the global response to the novel Coronavirus disease (COVID-19) pandemic, since its first outbreak in late 2019. At the inception of the COVID-19 pandemic, nasopharyngeal swab sample analysis for COVID-19 diagnosis using the real-time polymerase chain reaction (RT-PCR) technique was the most widely used. However, due to the high cost and difficulty of sample collection, the number of available sample types for COVID-19 diagnosis is rapidly increasing, as is the COVID-19 diagnostic literature. The use of nasal swabs, saliva, and oral fluids as viable sample options for the effective detection of SARS-CoV-2 has been implemented successfully in different settings since 2020. These alternative sample type provides a plethora of advantages including decreasing the high exposure risk to frontline workers, enhancing the chances of home self-sampling, reducing the cost, and significantly increasing testing capacity. This study sought to ascertain the effectiveness of Saliva samples as an alternative for COVID-19 diagnosis in Nigeria. Demographic data, paired samples of Nasopharyngeal Swab and Drooling Saliva were obtained from 309 consenting individuals aged 8-83 years presenting for COVID-19 testing. All samples were simultaneously assayed for the detection of SARS-CoV-2 RdRp, N, and E genes using the GeneFinder™ COVID-19 Plus RT-PCR test kit. Out of 309 participants, only 299 with valid RT-PCR results comprising 159 (53.2%) males and 140 (46.8%) females were analyzed in this study using the R Statistical package. Among the 299 samples analyzed, 39 (13.0%) had SARS-CoV-2 detected in at least one specimen type. Both swabs and saliva were positive in 20 (51.3%) participants. Ten participants (25.6%) had swab positive/saliva-negative results and 9 participants (23.1%) had saliva positive/swab-negative results. The percentage of positive and negative agreement of the saliva samples with the nasopharyngeal swab were 67% and 97% respectively with positive and negative predictive values as 69% and 96% respectively. The findings indicate that drooling saliva samples have good and comparable diagnostic accuracy to the nasopharyngeal swabs with moderate sensitivities and high specificities.
Collapse
Affiliation(s)
- Olumuyiwa Babalola Salu
- Centre for Human and Zoonotic Virology, Central Research Laboratory, College of Medicine of the University of Lagos, Idi-Araba, Lagos, Nigeria
- Department of Medical Microbiology and Parasitology, College of Medicine of the University of Lagos, Idi-Araba, Lagos, Nigeria
| | - Iorhen Ephraim Akase
- Department of Medicine, Infectious Disease Unit, Lagos University Teaching, Idi-Araba, Lagos State, Nigeria
| | - Roosevelt Amaobichukwu Anyanwu
- Centre for Human and Zoonotic Virology, Central Research Laboratory, College of Medicine of the University of Lagos, Idi-Araba, Lagos, Nigeria
| | - Mercy Remilekun Orenolu
- Centre for Human and Zoonotic Virology, Central Research Laboratory, College of Medicine of the University of Lagos, Idi-Araba, Lagos, Nigeria
| | - Maryam Abiodun Abdullah
- Centre for Human and Zoonotic Virology, Central Research Laboratory, College of Medicine of the University of Lagos, Idi-Araba, Lagos, Nigeria
| | | | | | | | | | - Kolawole Solomon Oyedeji
- Centre for Human and Zoonotic Virology, Central Research Laboratory, College of Medicine of the University of Lagos, Idi-Araba, Lagos, Nigeria
- Department of Medical Laboratory Science, College of Medicine of the University of Lagos, Idi-Araba, Lagos, Nigeria
| | - Sunday Aremu Omilabu
- Centre for Human and Zoonotic Virology, Central Research Laboratory, College of Medicine of the University of Lagos, Idi-Araba, Lagos, Nigeria
- Department of Medical Microbiology and Parasitology, College of Medicine of the University of Lagos, Idi-Araba, Lagos, Nigeria
| |
Collapse
|
250
|
Boldova AE, Korobkin JD, Nechipurenko YD, Sveshnikova AN. Theoretical Explanation for the Rarity of Antibody-Dependent Enhancement of Infection (ADE) in COVID-19. Int J Mol Sci 2022; 23:11364. [PMID: 36232664 PMCID: PMC9569501 DOI: 10.3390/ijms231911364] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/19/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
Global vaccination against the SARS-CoV-2 virus has proved to be highly effective. However, the possibility of antibody-dependent enhancement of infection (ADE) upon vaccination remains underinvestigated. Here, we aimed to theoretically determine conditions for the occurrence of ADE in COVID-19. We developed a series of mathematical models of antibody response: model Ab-a model of antibody formation; model Cv-a model of infection spread in the body; and a complete model, which combines the two others. The models describe experimental data on SARS-CoV and SARS-CoV-2 infections in humans and cell cultures, including viral load dynamics, seroconversion times and antibody concentration kinetics. The modelling revealed that a significant proportion of macrophages can become infected only if they bind antibodies with high probability. Thus, a high probability of macrophage infection and a sufficient amount of pre-existing antibodies are necessary for the development of ADE in SARS-CoV-2 infection. However, from the point of view of the dynamics of pneumocyte infection, the two cases where the body has a high concentration of preexisting antibodies and a high probability of macrophage infection and where there is a low concentration of antibodies in the body and no macrophage infection are indistinguishable. This conclusion could explain the lack of confirmed ADE cases for COVID-19.
Collapse
Affiliation(s)
- Anna E. Boldova
- Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya Str., 109029 Moscow, Russia
| | - Julia D. Korobkin
- Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya Str., 109029 Moscow, Russia
| | - Yury D. Nechipurenko
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anastasia N. Sveshnikova
- Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya Str., 109029 Moscow, Russia
- Department of Normal Physiology, Sechenov First Moscow State Medical University, 8/2 Trubetskaya St., 119991 Moscow, Russia
- Faculty of Fundamental Physico-Chemical Engineering, Lomonosov Moscow State University, 1/51 Leninskie Gory, 119991 Moscow, Russia
| |
Collapse
|