1
|
Eigner U, Köffer J, Betz U, Koglin J, Richter E. Evaluation of the cobas MTB and MTB-RIF/INH assay in clinical samples for the detection of Mycobacterium tuberculosis in respiratory specimens. J Clin Microbiol 2025:e0195924. [PMID: 40130921 DOI: 10.1128/jcm.01959-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/06/2025] [Indexed: 03/26/2025] Open
Abstract
The aim of this study was to evaluate the performance of the automated cobas MTB-Real-Time PCR assay for the rapid direct detection of Mycobacterium tuberculosis complex (MTBC) in clinical specimens and the ability of the cobas MTB-RIF/INH assay to correctly detect drug resistance to rifampin (RIF) and isoniazid (INH). The PCR assays were set up on the automated Cobas 6800 system, and the results were compared to liquid culture using BACTEC mycobacteria growth indicator tubes 960 TB system as the gold standard and line probe assays or sequencing results. A total of 500 N-acetyl-L-cysteine/sodium hydroxide (NALC-NaOH)-processed sputum samples were tested with the respective methods. The performance of MTBC detection in pulmonary specimens showed 91.8% sensitivity and 99.3% specificity in comparison to culture. The sensitivity for acid-fast bacteria (AFB) smear-positive specimens and for AFB smear-negative specimens was 100% and 85.1%, respectively. Due to the low prevalence of tuberculosis (TB) resistance in Germany, a collection of resistant TB strains with a wide variety of mutations was analyzed. The cobas MTB-RIF/INH assay detected 19 out of 21 INH-resistant and 22 out of 24 RIF-resistant TB strains. In conclusion, the cobas MTB and the cobas MTB-RIF/INH assays implemented on the automated cobas6800 instrument are reliable and versatile tools for the detection of MTB and RIF/INH resistance. IMPORTANCE Our manuscript addresses the WHO recommendation for the use of "moderate-complexity automated NAATs for detection of TB and resistance to rifampicin and isoniazid" as a part of the WHO End TB Strategy. Rapid detection of tuberculosis (TB) patients is essential to preventing TB transmission and finally reducing TB burden. In this study, we present data on the sensitivity and specificity of the novel cobas MTB assay for TB detection in a low-incidence country, demonstrating highly promising results. Additionally, by analyzing TB strains with various mutations conferring resistance to INH and/or RMP, we assess the opportunities and limitations of the cobas MTB-RIF/INH assay in reliably detecting drug resistance in sputum specimens, thereby facilitating the early onset of appropriate treatment.
Collapse
|
2
|
Kostaki EG. Virology Applications to the COVID-19 Pandemic. Life (Basel) 2025; 15:247. [PMID: 40003655 PMCID: PMC11856160 DOI: 10.3390/life15020247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 01/26/2025] [Indexed: 02/27/2025] Open
Abstract
From the early identification of severe respiratory cases of unknown etiology in Wuhan, China, in late 2019, virology research has played an important role in understanding, management, and prevention of the COVID-19 pandemic [...].
Collapse
Affiliation(s)
- Evangelia Georgia Kostaki
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
3
|
Bai J, Jiang Y, Tan F, Zhu P, Li X, Xiong X, Wang Z, Song T, Xie B, Yang Y, Han J. Electrochemical biosensor for sensitive detection of SARS-CoV-2 gene fragments using Bi 2Se 3 topological insulator. Bioelectrochemistry 2024; 159:108748. [PMID: 38824746 DOI: 10.1016/j.bioelechem.2024.108748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 06/04/2024]
Abstract
In this study, we have designed an electrochemical biosensor based on topological material Bi2Se3 for the sensitive detection of SARS-CoV-2 in the COVID-19 pandemic. Flake-shaped Bi2Se3 was obtained directly from high-quality single crystals using mechanical exfoliation, and the single-stranded DNA was immobilized onto it. Under optimal conditions, the peak current of the differential pulse voltammetry method exhibited a linear relationship with the logarithm of the concentration of target-complementary-stranded DNA, ranging from 1.0 × 10-15 to 1.0 × 10-11 M, with a detection limit of 3.46 × 10-16 M. The topological material Bi2Se3, with Dirac surface states, enhanced the signal-to-interference plus noise ratio of the electrochemical measurements, thereby improving the sensitivity of the sensor. Furthermore, the electrochemical sensor demonstrated excellent specificity in recognizing RNA. It can detect complementary RNA by amplifying and transcribing the initial DNA template, with an initial DNA template concentration ranging from 1.0 × 10-18 to 1.0 × 10-15 M. Furthermore, the sensor also effectively distinguished negative and positive results by detecting splitting-synthetic SARS-CoV-2 pseudovirus with a concentration of 1 copy/μL input. Our work underscores the immense potential of the electrochemical sensing platform based on the topological material Bi2Se3 in the detection of pathogens during the rapid spread of acute infectious diseases.
Collapse
Affiliation(s)
- Jiangyue Bai
- Centre for Quantum Physics, Key Laboratory of Advanced Optoelectronic Quantum Architecture and Measurement (MOE), School of Physics, Beijing Institute of Technology, Beijing 100081, China; International Center for Quantum Materials, Beijing Institute of Technology, Zhuhai, 519000, China; Beijing Key Lab of Nanophotonics and Ultrafine Optoelectronic Systems, School of Physics, Beijing Institute of Technology, Beijing 100081, China
| | - Yujiu Jiang
- Centre for Quantum Physics, Key Laboratory of Advanced Optoelectronic Quantum Architecture and Measurement (MOE), School of Physics, Beijing Institute of Technology, Beijing 100081, China; International Center for Quantum Materials, Beijing Institute of Technology, Zhuhai, 519000, China; Beijing Key Lab of Nanophotonics and Ultrafine Optoelectronic Systems, School of Physics, Beijing Institute of Technology, Beijing 100081, China
| | - Fan Tan
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Peng Zhu
- Centre for Quantum Physics, Key Laboratory of Advanced Optoelectronic Quantum Architecture and Measurement (MOE), School of Physics, Beijing Institute of Technology, Beijing 100081, China; International Center for Quantum Materials, Beijing Institute of Technology, Zhuhai, 519000, China; Beijing Key Lab of Nanophotonics and Ultrafine Optoelectronic Systems, School of Physics, Beijing Institute of Technology, Beijing 100081, China
| | - Xiuxia Li
- Centre for Quantum Physics, Key Laboratory of Advanced Optoelectronic Quantum Architecture and Measurement (MOE), School of Physics, Beijing Institute of Technology, Beijing 100081, China; International Center for Quantum Materials, Beijing Institute of Technology, Zhuhai, 519000, China; Beijing Key Lab of Nanophotonics and Ultrafine Optoelectronic Systems, School of Physics, Beijing Institute of Technology, Beijing 100081, China
| | - Xiaolu Xiong
- Centre for Quantum Physics, Key Laboratory of Advanced Optoelectronic Quantum Architecture and Measurement (MOE), School of Physics, Beijing Institute of Technology, Beijing 100081, China; International Center for Quantum Materials, Beijing Institute of Technology, Zhuhai, 519000, China; Beijing Key Lab of Nanophotonics and Ultrafine Optoelectronic Systems, School of Physics, Beijing Institute of Technology, Beijing 100081, China
| | - Zhiwei Wang
- Centre for Quantum Physics, Key Laboratory of Advanced Optoelectronic Quantum Architecture and Measurement (MOE), School of Physics, Beijing Institute of Technology, Beijing 100081, China; International Center for Quantum Materials, Beijing Institute of Technology, Zhuhai, 519000, China; Beijing Key Lab of Nanophotonics and Ultrafine Optoelectronic Systems, School of Physics, Beijing Institute of Technology, Beijing 100081, China
| | - Tinglu Song
- Experimental Centre of Advanced Materials School of Materials Science & Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Bingteng Xie
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| | - Yanbo Yang
- Centre for Quantum Physics, Key Laboratory of Advanced Optoelectronic Quantum Architecture and Measurement (MOE), School of Physics, Beijing Institute of Technology, Beijing 100081, China; International Center for Quantum Materials, Beijing Institute of Technology, Zhuhai, 519000, China; Beijing Key Lab of Nanophotonics and Ultrafine Optoelectronic Systems, School of Physics, Beijing Institute of Technology, Beijing 100081, China.
| | - Junfeng Han
- Centre for Quantum Physics, Key Laboratory of Advanced Optoelectronic Quantum Architecture and Measurement (MOE), School of Physics, Beijing Institute of Technology, Beijing 100081, China; International Center for Quantum Materials, Beijing Institute of Technology, Zhuhai, 519000, China; Beijing Key Lab of Nanophotonics and Ultrafine Optoelectronic Systems, School of Physics, Beijing Institute of Technology, Beijing 100081, China.
| |
Collapse
|
4
|
Veugen JMJ, Schoenmakers T, van Loo IHM, Haagmans BL, Leers MPG, Lamers MM, Lucchesi M, van Bussel BCT, van Mook WNKA, Nuijts RMMA, Savelkoul PHM, Dickman MM, Wolffs PFG. Advancing COVID-19 diagnostics: rapid detection of intact SARS-CoV-2 using viability RT-PCR assay. Microbiol Spectr 2024; 12:e0016024. [PMID: 39037224 PMCID: PMC11370235 DOI: 10.1128/spectrum.00160-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/10/2024] [Indexed: 07/23/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19). Commonly used methods for both clinical diagnosis of SARS-CoV-2 infection and management of infected patients involve the detection of viral RNA, but the presence of infectious virus particles is unknown. Viability PCR (v-PCR) uses a photoreactive dye to bind non-infectious RNA, ideally resulting in the detection of RNA only from intact virions. This study aimed to develop and validate a rapid v-PCR assay for distinguishing intact and compromised SARS-CoV-2. Propidium monoazide (PMAxx) was used as a photoreactive dye. Mixtures with decreasing percentages of intact SARS-CoV-2 (from 100% to 0%) were prepared from SARS-CoV-2 virus stock and a clinical sample. Each sample was divided into a PMAxx-treated part and a non-PMAxx-treated part. Reverse transcription-PCR (RT-PCR) using an in-house developed SARS-CoV-2 viability assay was then applied to both sample sets. The difference in intact SARS-CoV-2 was determined by subtracting the cycle threshold (Ct) value of the PMAxx-treated sample from the non-PMAxx-treated sample. Mixtures with decreasing concentrations of intact SARS-CoV-2 showed increasingly lower delta Ct values as the percentage of intact SARS-CoV-2 decreased, as expected. This relationship was observed in both high and low viral load samples prepared from cultured SARS-CoV-2 virus stock, as well as for a clinical sample prepared directly from a SARS-CoV-2 positive nasopharyngeal swab. In this study, a rapid v-PCR assay has been validated that can distinguish intact from compromised SARS-CoV-2. The presence of intact virus particles, as determined by v-PCR, may indicate SARS-CoV-2 infectiousness. IMPORTANCE This study developed a novel method that can help determine whether someone who has been diagnosed with coronavirus disease 2019 (COVID-19) is still capable of spreading the virus to others. Current tests only detect the presence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA, but cannot tell whether the particles are still intact and can thus infect cells. The researchers used a dye that selectively blocks the detection of damaged virions and free RNA. They showed that this viability PCR reliably distinguishes intact SARS-CoV-2 capable of infecting from damaged SARS-CoV-2 or free RNA in both cultured virus samples and a clinical sample. Being able to quickly assess contagiousness has important implications for contact tracing and safely ending isolation precautions. This viability PCR technique provides a simple way to obtain valuable information, beyond just positive or negative test results, about the actual risk someone poses of transmitting SARS-CoV-2 through the air or surfaces they come into contact with.
Collapse
Affiliation(s)
- Judith M. J. Veugen
- University Eye Clinic Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, the Netherlands
- Department of Medical Microbiology, Infectious Diseases & Infection Prevention, Maastricht University Medical Center, Maastricht, the Netherlands
- Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, the Netherlands
| | - Tom Schoenmakers
- Department of Clinical Chemistry and Hematology, Zuyderland Medical Center, Sittard-Geleen/Heerlen, the Netherlands
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
| | - Inge H. M. van Loo
- Department of Medical Microbiology, Infectious Diseases & Infection Prevention, Maastricht University Medical Center, Maastricht, the Netherlands
- Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, the Netherlands
| | - Bart L. Haagmans
- Viroscience Department, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Mathie P. G. Leers
- Department of Clinical Chemistry and Hematology, Zuyderland Medical Center, Sittard-Geleen/Heerlen, the Netherlands
- Faculty of Science, Environmental Sciences, Open Universiteit, Heerlen, the Netherlands
| | - Mart M. Lamers
- Viroscience Department, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Mayk Lucchesi
- Department of Medical Microbiology, Infectious Diseases & Infection Prevention, Maastricht University Medical Center, Maastricht, the Netherlands
- Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, the Netherlands
| | - Bas C. T. van Bussel
- Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, the Netherlands
- Department of Intensive Care Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Walther N. K. A. van Mook
- Department of Intensive Care Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School of Health Professions Education (SHE), Maastricht University, Maastricht, the Netherlands
| | - Rudy M. M. A. Nuijts
- University Eye Clinic Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, the Netherlands
- Department of Ophthalmology, Zuyderland Medical Center, Heerlen, the Netherlands
| | - Paul H. M. Savelkoul
- Department of Medical Microbiology, Infectious Diseases & Infection Prevention, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Mor M. Dickman
- University Eye Clinic Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, the Netherlands
| | - Petra F. G. Wolffs
- Department of Medical Microbiology, Infectious Diseases & Infection Prevention, Maastricht University Medical Center, Maastricht, the Netherlands
- Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
5
|
Ramya PR, Halder S, Nagamani K, Singh Chouhan R, Gandhi S. Disposable graphene-oxide screen-printed electrode integrated with portable device for detection of SARS-CoV-2 in clinical samples. Bioelectrochemistry 2024; 158:108722. [PMID: 38697015 DOI: 10.1016/j.bioelechem.2024.108722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/19/2024] [Accepted: 04/27/2024] [Indexed: 05/04/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) diagnosis is the need of the hour, as cases are persistently increasing, and new variants are constantly emerging. The ever-changing nature of the virus leading to multiple variants, has brought an imminent need for early, accurate and rapid detection methods. Herein, we have reported the design and fabrication of Screen-Printed Electrodes (SPEs) with graphene oxide (GO) as working electrode and modified with specific antibodies for SARS-CoV-2 Receptor Binding Domain (RBD). Flexibility of design, and portable nature has made SPEs the superior choice for electrochemical analysis. The developed immunosensor can detect RBD as low as 0.83 fM with long-term storage capacity. The fabricated SPEs immunosensor was tested using a miniaturized portable device and potentiostat on 100 patient nasopharyngeal samples and corroborated with RT-PCR data, displayed 94 % sensitivity. Additionally, the in-house developed polyclonal antibodies detected RBD antigen of the mutated Omicron variant of SARS-CoV-2 successfully. We have not observed any cross-reactivity/binding of the fabricated immunosensor with MERS (cross-reactive antigen) and Influenza A H1N1 (antigen sharing common symptoms). Hence, the developed SPEs sensor may be applied for bedside point-of-care diagnosis of SARS-CoV-2 using miniaturized portable device, in clinical samples.
Collapse
Affiliation(s)
- P R Ramya
- DBT-National Institute of Animal Biotechnology (NIAB), Hyderabad 500032, Telangana, India; DBT-Regional Centre for Biotechnology (RCB), Faridabad 121001, Haryana, India
| | - Sayanti Halder
- DBT-National Institute of Animal Biotechnology (NIAB), Hyderabad 500032, Telangana, India
| | - K Nagamani
- Department of Microbiology, Gandhi Medical College, Gandhi Hospital, Hyderabad 500025, Telangana, India
| | - Raghuraj Singh Chouhan
- Department of Environmental Sciences, Jožef Stefan Institute, Jamova Cesta 39, 1000 Ljubljana, Slovenia
| | - Sonu Gandhi
- DBT-National Institute of Animal Biotechnology (NIAB), Hyderabad 500032, Telangana, India; DBT-Regional Centre for Biotechnology (RCB), Faridabad 121001, Haryana, India.
| |
Collapse
|
6
|
Wang R, Lu S, Deng F, Wu L, Yang G, Chong S, Liu Y. Enhancing the understanding of SARS-CoV-2 protein with structure and detection methods: An integrative review. Int J Biol Macromol 2024; 270:132237. [PMID: 38734351 DOI: 10.1016/j.ijbiomac.2024.132237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 05/07/2024] [Indexed: 05/13/2024]
Abstract
As the rapid and accurate screening of infectious diseases can provide meaningful information for outbreak prevention and control, as well as owing to the existing limitations of the polymerase chain reaction (PCR), it is imperative to have new and validated detection techniques for SARS-CoV-2. Therefore, the rationale for outlining the techniques used to detect SARS-CoV-2 proteins and performing a comprehensive comparison to serve as a practical benchmark for future identification of similar viral proteins is clear. This review highlights the urgent need to strengthen pandemic preparedness by emphasizing the importance of integrated measures. These include improved tools for pathogen characterization, optimized societal precautions, the establishment of early warning systems, and the deployment of highly sensitive diagnostics for effective surveillance, triage, and resource management. Additionally, with an improved understanding of the virus' protein structure, considerable advances in targeted detection, treatment, and prevention strategies are expected to greatly improve our ability to respond to future outbreaks.
Collapse
Affiliation(s)
- Ruiqi Wang
- Shenyang University of Chemical Technology, Shenyang 110142, China; National Institute of Metrology, Beijing 100029, China
| | - Song Lu
- National Institute of Metrology, Beijing 100029, China
| | - Fanyu Deng
- National Institute of Metrology, Beijing 100029, China; North University of China, Taiyuan 030051, China
| | - Liqing Wu
- National Institute of Metrology, Beijing 100029, China
| | - Guowu Yang
- Shenzhen Academy of Metrology and Quality Inspection, Shenzhen 518055, China
| | - Siying Chong
- Shenyang University of Chemical Technology, Shenyang 110142, China
| | - Yahui Liu
- National Institute of Metrology, Beijing 100029, China.
| |
Collapse
|
7
|
Sepahi N, Samsami S, Mansoori Y, Chenari M, Namavari N, Yazdanpanah A, Ghasemian A, Montaseri Z, Sharifzadeh M, Ranjbar R, Namavari S, Ghanbariasad A. Development of a novel Colorimetric Assay for the rapid diagnosis of Coronavirus disease 2019 from nasopharyngeal samples. Sci Rep 2024; 14:12125. [PMID: 38802360 PMCID: PMC11130264 DOI: 10.1038/s41598-024-53747-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/04/2024] [Indexed: 05/29/2024] Open
Abstract
Emergence of Coronavirus disease 2019 (COVID-19) pandemic has posed a huge threat to public health. Rapid and reliable test to diagnose infected subjects is crucial for disease spread control. We developed a colorimetric test for COVID-19 detection using a Colorimetric Assay based on thiol-linked RNA modified gold nanoparticles (AuNPs) and oligonucleotide probes. This method was conducted on RNA from 200 pharyngeal swab samples initially tested by Real-Time polymerase chain reaction (RT-PCR) as gold standard. A specific oligonucleotide probe designed based on ORF1ab of COVID-19 was functionalized with AuNPs-probe conjugate. The exposure of AuNP-probe to isolated RNA samples was tested using hybridization. In this comparative study, the colorimetric functionalized AuNPs assay exhibited a detection limit of 25 copies/µL. It was higher in comparison to the RT-PCR method, which could only detect 15 copies/µL. The results demonstrated 100% specificity and 96% sensitivity for the developed method. Herein, we developed an incredibly rapid, simple and cost-effective Colorimetric Assay lasting approximately 30 min which could process considerably higher number of COVID-19 samples compared to the RT-PCR. This AuNP-probe conjugate colorimetric method could be considered the optimum alternatives for conventional diagnostic tools especially in over-populated and/or low-income countries.
Collapse
Affiliation(s)
- Neda Sepahi
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Sahar Samsami
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran
| | - Yaser Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Maryam Chenari
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Negin Namavari
- School of Medicine Grenada, St. George's University, St. George's, West Indies, Grenada
| | - Ava Yazdanpanah
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran
| | - Abdolmajid Ghasemian
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Zahra Montaseri
- Department of Infectious Diseases, School of Medicine, Fasa University of Medical Science, Fasa, Iran
| | - Mahboobeh Sharifzadeh
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Razie Ranjbar
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Sahar Namavari
- Department of Medical Biotechnologies, School of Advanced Technologies, Fasa University of Medical Sciences, Avicenna Square, Fasa, Fars, Islamic Republic of Iran
| | - Ali Ghanbariasad
- Department of Medical Biotechnologies, School of Advanced Technologies, Fasa University of Medical Sciences, Avicenna Square, Fasa, Fars, Islamic Republic of Iran.
| |
Collapse
|
8
|
Schädler J, Azeke AT, Ondruschka B, Steurer S, Lütgehetmann M, Fitzek A, Möbius D. Concordance between MITS and conventional autopsies for pathological and virological diagnoses. Int J Legal Med 2024; 138:431-442. [PMID: 37837537 PMCID: PMC10861633 DOI: 10.1007/s00414-023-03088-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 09/04/2023] [Indexed: 10/16/2023]
Abstract
In pandemics or to further study highly contagious infectious diseases, new strategies are needed for the collection of post-mortem tissue samples to identify the pathogen as well as its morphological impact. In this study, an ultrasound-guided minimally invasive tissue sampling (MITS) protocol was developed and validated for post-mortem use. The histological and microbiological qualities of post-mortem specimens were evaluated and compared between MITS and conventional autopsy (CA) in a series of COVID-19 deaths. Thirty-six ultrasound-guided MITS were performed. In five cases more, specimens for histological and virological examination were also obtained and compared during the subsequently performed CA. Summary statistics and qualitative interpretations (positive, negative) were calculated for each organ tissue sample from MITS and CA, and target genes were determined for both human cell count (beta-globin) and virus (SARS-CoV-2 specific E gene). There are no significant differences between MITS and CA with respect to the detectability of viral load in individual organs, which is why MITS can be of utmost importance and an useful alternative, especially during outbreaks of infectious diseases.
Collapse
Affiliation(s)
- Julia Schädler
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Akhator Terence Azeke
- Department of Anatomic Pathology, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Benjamin Ondruschka
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marc Lütgehetmann
- Institute of Medical Microbiology, Virology, and Hygiene, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Antonia Fitzek
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dustin Möbius
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
9
|
Qi L, Zhang Z, Wang M, Ke Z, Mao H, Deng G, Wang J. One-plasmid double-expression system for preparation of MS2 virus-like particles packaging SARS-CoV-2 RNA. Front Cell Infect Microbiol 2023; 13:1238543. [PMID: 38094745 PMCID: PMC10716189 DOI: 10.3389/fcimb.2023.1238543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023] Open
Abstract
COVID-19 is a disease caused by a virus named SARS-CoV-2. SARS-CoV-2 is a single-stranded positive-sense RNA virus. Reverse transcription quantitative PCR (RT-qPCR) assays are the gold standard molecular test for detection of RNA viruses. The aim of this study was to construct an RNA-positive control based on MS2 phage-like particles (MS2 VLPs) to detect SARS-CoV-2 RNA. pCDFDuet-1 was used as a one-plasmid double-expression system to construct MS2 VLPs containing ssRNA of SARS-CoV-2. The sequence encoding one copy of maturase, His-tag and coat protein dimer was cloned and inserted into MCS1 of the plasmid; the fragment encoding protein N and ORF1ab from SARS-CoV-2 was cloned and inserted into MCS2. The prepared plasmid was transformed into Escherichia coli strain BL2 (DE3), and expression of the construct was induced by 1 mM isopropyl-L-thio-D-galactopyranoside (IPTG) at 30°C for 12 hours. MS2 VLPs were purified and collected with Ni-NTA affinity chromatography columns. The size and shape of the MS2 VLPs were verified by transmission electron microscopy, and the stability of MS2 VLP packaged RNA was evaluated by treatment with RNase A. Effects of storage temperature and buffer on MS2 VLP stability were also investigated. The results showed that SARS-CoV-2 MS2 VLPs could be successfully produced by this one-plasmid double-expression system. MS2 VLPs showed high stability and may be used as a positive control in molecular diagnosis of COVID-19.
Collapse
Affiliation(s)
- Lili Qi
- School of Biological and Chemical Engineering, NingboTech University, Ningbo, Zhejiang, China
| | - Zheng Zhang
- School of Biological and Chemical Engineering, NingboTech University, Ningbo, Zhejiang, China
| | - Mengting Wang
- School of Biological and Chemical Engineering, NingboTech University, Ningbo, Zhejiang, China
| | - Zhijian Ke
- School of Biological and Chemical Engineering, NingboTech University, Ningbo, Zhejiang, China
| | - Haiguang Mao
- School of Biological and Chemical Engineering, NingboTech University, Ningbo, Zhejiang, China
| | - Gang Deng
- Blood Transfusion Research Institute, Ningbo Central Blood Station, Ningbo, Zhejiang, China
| | - Jinbo Wang
- School of Biological and Chemical Engineering, NingboTech University, Ningbo, Zhejiang, China
| |
Collapse
|
10
|
Kim KW, Lee B, Eom S, Shin D, Park C, Kim S, Yi H. Universal primers for rift valley fever virus whole-genome sequencing. Sci Rep 2023; 13:18688. [PMID: 37907670 PMCID: PMC10618441 DOI: 10.1038/s41598-023-45848-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 10/24/2023] [Indexed: 11/02/2023] Open
Abstract
Rift Valley fever (RVF) is a mosquito-borne zoonotic disease causing acute hemorrhagic fever. Accurate identification of mutations and phylogenetic characterization of RVF virus (RVFV) require whole-genome analysis. Universal primers to amplify the entire RVFV genome from clinical samples with low copy numbers are currently unavailable. Thus, we aimed to develop universal primers applicable for all known RVFV strains. Based on the genome sequences available from public databases, we designed eight pairs of universal PCR primers covering the entire RVFV genome. To evaluate primer universality, four RVFV strains (ZH548, Kenya 56 (IB8), BIME-01, and Lunyo), encompassing viral phylogenetic diversity, were chosen. The nucleic acids of the test strains were chemically synthesized or extracted via cell culture. These RNAs were evaluated using the PCR primers, resulting in successful amplification with expected sizes (0.8-1.7 kb). Sequencing confirmed that the products covered the entire genome of the RVFV strains tested. Primer specificity was confirmed via in silico comparison against all non-redundant nucleotide sequences using the BLASTn alignment tool in the NCBI database. To assess the clinical applicability of the primers, mock clinical specimens containing human and RVFV RNAs were prepared. The entire RVFV genome was successfully amplified and sequenced at a viral concentration of 108 copies/mL. Given the universality, specificity, and clinical applicability of the primers, we anticipate that the RVFV universal primer pairs and the developed method will aid in RVFV phylogenomics and mutation detection.
Collapse
Affiliation(s)
- Kwan Woo Kim
- Department of Public Health Sciences, Graduate School, Korea University, Seoul, Republic of Korea
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul, Republic of Korea
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Banseok Lee
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul, Republic of Korea
- Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul, Republic of Korea
| | - Sujeong Eom
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul, Republic of Korea
- Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul, Republic of Korea
| | - Donghoon Shin
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul, Republic of Korea
- Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul, Republic of Korea
| | - Changwoo Park
- Microbiological Analysis Team, Group for Biometrology, Korea Research Institute of Standards and Science (KRISS), Daejeon, Republic of Korea
- Convergent Research Center for Emerging Virus Infection, Korea Research Institute of Chemical Technology (KRICT), Daejeon, Republic of Korea
- Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Seil Kim
- Microbiological Analysis Team, Group for Biometrology, Korea Research Institute of Standards and Science (KRISS), Daejeon, Republic of Korea.
- Convergent Research Center for Emerging Virus Infection, Korea Research Institute of Chemical Technology (KRICT), Daejeon, Republic of Korea.
- Department of Bio-Analysis Science, University of Science and Technology, Daejeon, Republic of Korea.
| | - Hana Yi
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul, Republic of Korea.
- Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul, Republic of Korea.
- School of Biosystems and Biomedical Sciences, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
11
|
Naorungroj S, Srisomwat C, Khamcharoen W, Jampasa S, Pasomsub E, Shin K, Vilaivan T, Chailapakul O. Sequential Flow Controllable Microfluidic Device for G-Quadruplex DNAzyme-Based Electrochemical Detection of SARS-CoV-2 Using a Pyrrolidinyl Peptide Nucleic Acid. Anal Chem 2023; 95:12794-12801. [PMID: 37590190 DOI: 10.1021/acs.analchem.3c01758] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been a significant health issue globally. Point-of-care (POC) testing that can offer a rapid and accurate diagnosis of SARS-CoV-2 at the early stage of infection is highly desirable to constrain this outbreak, especially in resource-limited settings. Herein, we present a G-quadruplex DNAzyme-based electrochemical assay that is integrated with a sequential flow controllable microfluidic device for the detection of SARS-CoV-2 cDNA. According to the detection principle, a pyrrolidinyl peptide nucleic acid probe is immobilized on a screen-printed graphene electrode for capturing SARS-CoV-2 DNA. The captured DNA subsequently hybridizes with another DNA probe that carries a G-quadruplex DNAzyme as the signaling unit. The G-quadruplex DNAzyme catalyzes the H2O2-mediated oxidation of hydroquinone to benzoquinone that can be detected using square-wave voltammetry to give a signal that corresponds to the target DNA concentration. The assay exhibited high selectivity for SARS-CoV-2 DNA and showed a good experimental detection limit at 30 pM. To enable automation, the DNAzyme-based assay was combined with a capillary-driven microfluidic device featuring a burst valve technology to allow sequential sample and reagent delivery as well as the DNA target hybridization and enzymatic reaction to be operated in a precisely controlled fashion. The developed microfluidic device was successfully applied for the detection of SARS-CoV-2 from nasopharyngeal swab samples. The results were in good agreement with the standard RT-PCR method and could be performed within 20 min. Thus, this platform offers desirable characteristics that make it an alternative POC tool for COVID-19 diagnosis.
Collapse
Affiliation(s)
- Sarida Naorungroj
- Electrochemistry and Optical Spectroscopy Center of Excellence (EOSCE), Department of Chemistry, Faculty of Science, Chulalongkorn University, Phayathai Road, Pathumwan, Bangkok 10330, Thailand
| | - Chawin Srisomwat
- Department of Chemistry, Faculty of Science and Technology, Thammasat University, Pathumthani 12121, Thailand
| | - Wisarut Khamcharoen
- Electrochemistry and Optical Spectroscopy Center of Excellence (EOSCE), Department of Chemistry, Faculty of Science, Chulalongkorn University, Phayathai Road, Pathumwan, Bangkok 10330, Thailand
| | - Sakda Jampasa
- Electrochemistry and Optical Spectroscopy Center of Excellence (EOSCE), Department of Chemistry, Faculty of Science, Chulalongkorn University, Phayathai Road, Pathumwan, Bangkok 10330, Thailand
| | - Ekawat Pasomsub
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Kwanwoo Shin
- Department of Chemistry and Institute of Biological Interfaces, Sogang University, Seoul 04107, Republic of Korea
| | - Tirayut Vilaivan
- Organic Synthesis Research Unit, Department of Chemistry, Faculty of Science, Chulalongkorn University, Phayathai Road, Patumwan, Bangkok 10330, Thailand
| | - Orawon Chailapakul
- Electrochemistry and Optical Spectroscopy Center of Excellence (EOSCE), Department of Chemistry, Faculty of Science, Chulalongkorn University, Phayathai Road, Pathumwan, Bangkok 10330, Thailand
| |
Collapse
|
12
|
Terence Azeke A, Schädler J, Ondruschka B, Steurer S, Möbius D, Fitzek A. Minimally Invasive Tissue Sampling via Post Mortem Ultrasound: A Feasible Tool (Not Only) in Infectious Diseases-A Case Report. Diagnostics (Basel) 2023; 13:2643. [PMID: 37627902 PMCID: PMC10453131 DOI: 10.3390/diagnostics13162643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
In the past years the number of hospital autopsies have declined steadily, becoming almost excluded from medical training. Medicolegal (forensic) autopsies account for almost all autopsies, whereas hospital autopsies are becoming increasingly rare. Minimally invasive tissue sampling (MITS) using post mortem ultrasound offers the opportunity to increase the number of post mortem examinations in a clinical and even forensic context. MITS is a needle-based post mortem procedure that uses (radiological) imaging techniques to examine major organs of the body, acquire tissue samples and aspirate fluid from the body cavities or hollow organs. In this study, MITS was used to determine the presence of other co-existing diseases in a deceased infected 97-year-old woman with severe acute respiratory syndrome coronavirus 2. The examination of her body was carried out using ultrasound as an imaging tool and to gather ultrasound-guided biopsies as conventional autopsy was rejected by the next of kin. Ultrasound and histology identified an intravesical mass leading to an obstruction of the urinary outlet resulting in bilateral hydronephrosis and purulent pyelonephritis, which was unknown during her lifetime. Histopathological examination revealed the tumor mass to be a squamous cell carcinoma. This study has shown that MITS can be used to determine the cause of death and the presence of concomitant diseases in the infectious deceased.
Collapse
Affiliation(s)
- Akhator Terence Azeke
- Department of Anatomic Pathology, Irrua Specialist Teaching Hospital, KM 87 Benin Auchi Rd, Irrua 310115, Nigeria
| | - Julia Schädler
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Butenfeld 34, D-22529 Hamburg, Germany
| | - Benjamin Ondruschka
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Butenfeld 34, D-22529 Hamburg, Germany
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Dustin Möbius
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Butenfeld 34, D-22529 Hamburg, Germany
| | - Antonia Fitzek
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Butenfeld 34, D-22529 Hamburg, Germany
| |
Collapse
|
13
|
Tang YN, Jiang D, Wang X, Liu Y, Wei D. Recent progress on rapid diagnosis of COVID-19 by point-of-care testing platforms. CHINESE CHEM LETT 2023; 35:108688. [PMID: 37362324 PMCID: PMC10266891 DOI: 10.1016/j.cclet.2023.108688] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 05/25/2023] [Accepted: 06/12/2023] [Indexed: 06/28/2023]
Abstract
The outbreak of COVID-19 has drawn great attention around the world. SARS-CoV-2 is a highly infectious virus with occult transmission by many mutations and a long incubation period. In particular, the emergence of asymptomatic infections has made the epidemic even more severe. Therefore, early diagnosis and timely management of suspected cases are essential measures to control the spread of the virus. Developing simple, portable, and accurate diagnostic techniques for SARS-CoV-2 is the key to epidemic prevention. The advantages of point-of-care testing technology make it play an increasingly important role in viral detection and screening. This review summarizes the point-of-care testing platforms developed by nucleic acid detection, immunological detection, and nanomaterial-based biosensors detection. Furthermore, this paper provides a prospect for designing future highly accurate, cheap, and convenient SARS-CoV-2 diagnostic technology.
Collapse
Affiliation(s)
- Ya-Nan Tang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Institute of Molecular Materials and Devices, Fudan University, Shanghai 200433, China
| | - Dingding Jiang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Institute of Molecular Materials and Devices, Fudan University, Shanghai 200433, China
| | - Xuejun Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Institute of Molecular Materials and Devices, Fudan University, Shanghai 200433, China
| | - Yunqi Liu
- Institute of Molecular Materials and Devices, Fudan University, Shanghai 200433, China
| | - Dacheng Wei
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Institute of Molecular Materials and Devices, Fudan University, Shanghai 200433, China
| |
Collapse
|
14
|
Hosseinpour Moghadam N, Najafi R, Ghanbariasad A, Shiralizadeh Dezfuli A, Jalali A. Improving the selective naked-eye detection of COVID-19 mediated by simultaneously using three different target oligonucleotides coated on plasmonic AuNPs/hexagonal Ag@AuNPs. J Biomol Struct Dyn 2023; 41:14372-14381. [PMID: 36995117 DOI: 10.1080/07391102.2023.2193989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 02/10/2023] [Indexed: 03/31/2023]
Abstract
The localized surface plasmon resonance (LSPR) phenomenon provides a versatile property in biosensor technology. This uncommon feature was utilized to produce a homogeneous optical biosensor to detect COVID-19 by the naked-eye readout. In this work, we synthesized two types of plasmonic nanoparticles: (i) AuNPs and (ii) hexagonal core-shell nanoparticles-Au shell on AgNPs (Au@AgNPs). We report herein the development of two colorimetric biosensors employing the efficient targeting and the binding ability for three regions of the COVID-19 genome, that is, S-gene, N-gene and E-gene, at the same time. Two AuNPs and Ag@AuNPs individually coated with three different targets oligonucleotide sequence (TOs) (AuNPs-TOs-mix and Ag@AuNPs-TOs-mix) for simultaneous detection of S-gene, N-gene and E-gene of the COVID-19 virus, using the LSPR and naked-eye methods in the laboratory and biological samples. The target COVID-19 genome RNA detected using the AuNPs-TOs-mix and Ag@AuNPs-TOs-mix can achieve the same sensitivity. The detection ranges by the AuNPs-TOs-mix and Ag@AuNPs-TOs-mix are both sufficiently improved in equal amounts in comparison to any of the AuNPs-TOs and Ag@AuNPs-TOs. The sensitivity of the current COVID-19 biosensors were 94% and 96% based on the number of positive samples detected for AuNPs-TOs-mix and Ag@AuNPs-TOs-mix, respectively. Moreover, all the real-time PCR confirmed negative samples obtained the same results by the biosensor; accordingly, the specificity of this approach got to 100%. The current study reports a selective, reliable, reproducible and visual 'naked-eye' detection of COVID-19, devoid of the requirement of any sophisticated instrumental techniques.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | - Rezvan Najafi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Ghanbariasad
- Department of Medical Biotechnology, Fasa University of Medical Sciences, Fasa, Iran
| | | | - Akram Jalali
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
15
|
A triple-target reverse transcription loop-mediated isothermal amplification (RT-LAMP) for rapid and accurate detection of SARS-CoV-2 virus. Anal Chim Acta 2023; 1255:341146. [PMID: 37032059 PMCID: PMC10039734 DOI: 10.1016/j.aca.2023.341146] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023]
Abstract
The spreading of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) across the world has impacted people's health and lives worldwide in recent years. Rapid and accurate diagnosis is crucial for curbing the pandemic of coronavirus disease 2019 (COVID-19). Reverse transcription loop-mediated isothermal amplification (RT-LAMP) has great potential for SARS-CoV-2 detection but fails to completely replace conventional PCR due to the high false-positive rate (FPR). We proposed a triple-target RT-LAMP method for dual-signal, sensitive, and simultaneous detection of conserved genes of SARS-CoV-2. Multiple LAMP primer sets were designed for N, E, and M genes and their amplification efficacy were screened. Then, using artificial plasmids and RNA, the optimal primer set for each gene was examined on specificity, sensitivity, and detection range. The RT-LAMP initiated by these primer sets exhibited better specificity and sensitivity than that of RT-qPCR, and the triple-target RT-LAMP could determine different variants of SARS-CoV-2. By testing 78 artificial RNA samples, the total FPR of triple-target RT-LAMP was eliminated compared with that of mono-target RT-LAMP. The triple-target RT-LAMP method precisely identified throat swab specimens through colorimetry and fluorescent signals within 60 min, and the limit of detection (LOD) was as low as 187 copies/reaction. In the future, the triple-target RT-LAMP can be applied to in-field and on-site diagnosis of symptomatic and asymptomatic virus carriers.
Collapse
|
16
|
Castro-Balado A, Novo-Veleiro I, Vázquez-Agra N, Barbeito-Castiñeiras G, Estany-Gestal A, Trastoy-Pena R, González-Barcia M, Zarra-Ferro I, del Río-Garma MC, Crespo-Diz C, Delgado-Sánchez O, Otero-Espinar FJ, Mondelo-García C, Pose-Reino A, Fernández-Ferreiro A. Efficacy and Safety of Inhaled Ethanol in Early-Stage SARS-CoV-2 Infection in Older Adults: A Phase II Randomized Clinical Trial. Pharmaceutics 2023; 15:pharmaceutics15020667. [PMID: 36839987 PMCID: PMC9966500 DOI: 10.3390/pharmaceutics15020667] [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: 12/25/2022] [Revised: 02/06/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
BACKGROUND Inhaled ethanol in the early stages of SARS-CoV-2 infection may reduce the viral load, decreasing progression and improving prognosis. The ALCOVID-19 trial was designed to study the efficacy and safety of inhaled ethanol in older adults at initial phases of infection. METHODS Randomized, triple-blind, placebo-controlled phase II clinical trial. Experimental group (n = 38) inhaled 65° ethanol through an oxygen flow, while in the control group (n = 37), water for injection was used. General endpoint was to evaluate disease progression according to the modified World Health Organization (WHO) Clinical Progression Scale. Specific effectiveness endpoints were body temperature, oxygen saturation, viral load assessed by cycle threshold (Ct) on real-time polymerase chain reaction (RT-PCR), analytical biomarkers and use of antibiotics or corticosteroids. Specific safety outcomes were the absence of ethanol in plasma, electrographic, analytical, or respiratory alterations. RESULTS In the intention-to-treat population, no differences were found regarding disease progression. Mean Ct values increased over time in both groups, being numerically higher in the ethanol group, reaching a value above 33 only in the ethanol group on day 14, a value above which patients are considered non-infective. No differences were found in the other specific effectiveness endpoints. Inhaled ethanol was proven to be safe as no plasma ethanol was detected, and there were no electrocardiographic, analytical, or respiratory alterations. CONCLUSIONS The efficacy of inhaled ethanol in terms of the progression of SARS-CoV-2 infection was not demonstrated in the present trial. However, it is positioned as a safe treatment for elderly patients with early-stage COVID-19.
Collapse
Affiliation(s)
- Ana Castro-Balado
- Pharmacy Department, University Clinical Hospital of Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain
- Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Pharmacology, Pharmacy and Pharmaceutical Technology Department, Faculty of Pharmacy, University of Santiago de Compostela (USC), 15782 Santiago de Compostela, Spain
| | - Ignacio Novo-Veleiro
- Internal Medicine Department, University Clinical Hospital of Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain
| | - Néstor Vázquez-Agra
- Internal Medicine Department, University Clinical Hospital of Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain
| | - Gema Barbeito-Castiñeiras
- Microbiology Department, University Clinical Hospital of Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain
| | - Ana Estany-Gestal
- Epidemiology and Clinical Research Unit, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Rocío Trastoy-Pena
- Microbiology Department, University Clinical Hospital of Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain
| | - Miguel González-Barcia
- Pharmacy Department, University Clinical Hospital of Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain
- Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Irene Zarra-Ferro
- Pharmacy Department, University Clinical Hospital of Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain
- Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - María Carmen del Río-Garma
- Clinical Analytic Department, University Clinical Hospital of Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain
| | - Carlos Crespo-Diz
- Pharmacy Department, University Clinical Hospital of Pontevedra (SERGAS), 36162 Pontevedra, Spain
| | | | - Francisco J. Otero-Espinar
- Pharmacology, Pharmacy and Pharmaceutical Technology Department, Faculty of Pharmacy, University of Santiago de Compostela (USC), 15782 Santiago de Compostela, Spain
| | - Cristina Mondelo-García
- Pharmacy Department, University Clinical Hospital of Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain
- Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Correspondence: (C.M.-G.); (A.P.-R.); (A.F.-F.); Tel.: +34-981-951-423 (A.F.-F.)
| | - Antonio Pose-Reino
- Internal Medicine Department, University Clinical Hospital of Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain
- Correspondence: (C.M.-G.); (A.P.-R.); (A.F.-F.); Tel.: +34-981-951-423 (A.F.-F.)
| | - Anxo Fernández-Ferreiro
- Pharmacy Department, University Clinical Hospital of Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain
- Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Correspondence: (C.M.-G.); (A.P.-R.); (A.F.-F.); Tel.: +34-981-951-423 (A.F.-F.)
| |
Collapse
|
17
|
Truong PL, Yin Y, Lee D, Ko SH. Advancement in COVID-19 detection using nanomaterial-based biosensors. EXPLORATION (BEIJING, CHINA) 2023; 3:20210232. [PMID: 37323622 PMCID: PMC10191025 DOI: 10.1002/exp.20210232] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 05/11/2022] [Indexed: 06/17/2023]
Abstract
Coronavirus disease 2019 (COVID-19) pandemic has exemplified how viral growth and transmission are a significant threat to global biosecurity. The early detection and treatment of viral infections is the top priority to prevent fresh waves and control the pandemic. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been identified through several conventional molecular methodologies that are time-consuming and require high-skill labor, apparatus, and biochemical reagents but have a low detection accuracy. These bottlenecks hamper conventional methods from resolving the COVID-19 emergency. However, interdisciplinary advances in nanomaterials and biotechnology, such as nanomaterials-based biosensors, have opened new avenues for rapid and ultrasensitive detection of pathogens in the field of healthcare. Many updated nanomaterials-based biosensors, namely electrochemical, field-effect transistor, plasmonic, and colorimetric biosensors, employ nucleic acid and antigen-antibody interactions for SARS-CoV-2 detection in a highly efficient, reliable, sensitive, and rapid manner. This systematic review summarizes the mechanisms and characteristics of nanomaterials-based biosensors for SARS-CoV-2 detection. Moreover, continuing challenges and emerging trends in biosensor development are also discussed.
Collapse
Affiliation(s)
- Phuoc Loc Truong
- Laser and Thermal Engineering LabDepartment of Mechanical EngineeringGachon UniversitySeongnamKorea
| | - Yiming Yin
- New Materials InstituteDepartment of MechanicalMaterials and Manufacturing EngineeringUniversity of Nottingham Ningbo ChinaNingboChina
- Applied Nano and Thermal Science LabDepartment of Mechanical EngineeringSeoul National UniversityGwanak‐guSeoulKorea
| | - Daeho Lee
- Laser and Thermal Engineering LabDepartment of Mechanical EngineeringGachon UniversitySeongnamKorea
| | - Seung Hwan Ko
- Applied Nano and Thermal Science LabDepartment of Mechanical EngineeringSeoul National UniversityGwanak‐guSeoulKorea
- Institute of Advanced Machinery and Design (SNU‐IAMD)/Institute of Engineering ResearchSeoul National UniversityGwanak‐guSeoulKorea
| |
Collapse
|
18
|
Maryam S, Ul Haq I, Yahya G, Ul Haq M, Algammal AM, Saber S, Cavalu S. COVID-19 surveillance in wastewater: An epidemiological tool for the monitoring of SARS-CoV-2. Front Cell Infect Microbiol 2023; 12:978643. [PMID: 36683701 PMCID: PMC9854263 DOI: 10.3389/fcimb.2022.978643] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 11/03/2022] [Indexed: 01/06/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has prompted a lot of questions globally regarding the range of information about the virus's possible routes of transmission, diagnostics, and therapeutic tools. Worldwide studies have pointed out the importance of monitoring and early surveillance techniques based on the identification of viral RNA in wastewater. These studies indicated the presence of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA in human feces, which is shed via excreta including mucus, feces, saliva, and sputum. Subsequently, they get dumped into wastewater, and their presence in wastewater provides a possibility of using it as a tool to help prevent and eradicate the virus. Its monitoring is still done in many regions worldwide and serves as an early "warning signal"; however, a lot of limitations of wastewater surveillance have also been identified.
Collapse
Affiliation(s)
- Sajida Maryam
- Department of Biosciences, The Commission on Science and Technology for Sustainable Development in the South (COMSATS) University Islamabad (CUI), Islamabad, Pakistan
| | - Ihtisham Ul Haq
- Department of Biosciences, The Commission on Science and Technology for Sustainable Development in the South (COMSATS) University Islamabad (CUI), Islamabad, Pakistan
- Department of Physical Chemistry and Polymers Technology, Silesian University of Technology, Gliwice, Poland
- Joint Doctoral School, Silesian University of Technology, Gliwice, Poland
| | - Galal Yahya
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mehboob Ul Haq
- Department of Biosciences, The Commission on Science and Technology for Sustainable Development in the South (COMSATS) University Islamabad (CUI), Islamabad, Pakistan
| | - Abdelazeem M. Algammal
- Department of Bacteriology, Immunology, and Mycology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
19
|
Fonseca Brito L, Tödter S, Kottlau J, Cermann K, Spier A, Petersen E, Schäfer I, Twerenbold R, Aepfelbacher M, Lütgehetmann M, Stahl FR. Performance of an interferon-γ release assay-based test for cell-mediated immunity to SARS-CoV-2. Front Immunol 2023; 14:1069968. [PMID: 36875076 PMCID: PMC9978494 DOI: 10.3389/fimmu.2023.1069968] [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: 10/17/2022] [Accepted: 01/31/2023] [Indexed: 02/18/2023] Open
Abstract
In search for immunological correlates of protection against acute coronavirus disease 2019 (COVID-19) there is a need for high through-put assays for cell-mediated immunity (CMI) to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. We established an interferon-γ release assay -based test for detection of CMI against SARS-CoV-2 spike (S) or nucleocapsid (NC) peptides. Blood samples obtained from 549 healthy or convalescent individuals were measured for interferon-γ (IFN-γ) production after peptide stimulation using a certified chemiluminescence immunoassay. Test performance was calculated applying cutoff values with the highest Youden indices in receiver-operating-characteristics curve analysis and compared to a commercially available serologic test. Potential confounders and clinical correlates were assessed for all test systems. 522 samples obtained from 378 convalescent in median 298 days after PCR-confirmed SARS-CoV-2 infection and 144 healthy control individuals were included in the final analysis. CMI testing had a sensitivity and specificity of up to 89% and 74% for S peptides and 89% and 91% for NC peptides, respectively. High white blood cell counts correlated negatively with IFN-γ responses but there was no CMI decay in samples obtained up to one year after recovery. Severe clinical symptoms at time of acute infection were associated with higher measures of adaptive immunity and reported hair loss at time of examination. This laboratory-developed test for CMI to SARS-CoV-2 NC peptides exhibits excellent test performance, is suitable for high through-put routine diagnostics, and should be evaluated for clinical outcome prediction in prospective pathogen re-exposure.
Collapse
Affiliation(s)
- Luís Fonseca Brito
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Virus-Host-Interaction, Leibniz Institute of Virology, Hamburg, Germany
| | - Silvia Tödter
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julian Kottlau
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kathrin Cermann
- Institute of Microbiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anthea Spier
- Institute of Microbiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elina Petersen
- Department of Cardiology, University Heart and Vascular Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Population Health Research Department, University Heart and Vascular Center, Hamburg, Germany
| | - Ines Schäfer
- Department of Cardiology, University Heart and Vascular Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Population Health Research Department, University Heart and Vascular Center, Hamburg, Germany
| | - Raphael Twerenbold
- Department of Cardiology, University Heart and Vascular Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Population Health Research Department, University Heart and Vascular Center, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK) Partner Site Hamburg-Kiel-Lübeck, Hamburg, Germany
| | - Martin Aepfelbacher
- Institute of Microbiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marc Lütgehetmann
- Institute of Microbiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Felix R Stahl
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Virus-Host-Interaction, Leibniz Institute of Virology, Hamburg, Germany
| |
Collapse
|
20
|
Dunay GA, Barroso M, Woidy M, Danecka MK, Engels G, Hermann K, Neumann FS, Paul K, Beime J, Escherich G, Fehse K, Grinstein L, Haniel F, Haupt LJ, Hecher L, Kehl T, Kemen C, Kemper MJ, Kobbe R, Kohl A, Klokow T, Nörz D, Olfe J, Schlenker F, Schmiesing J, Schrum J, Sibbertsen F, Stock P, Tiede S, Vettorazzi E, Zazara DE, Zapf A, Lütgehetmann M, Oh J, Mir TS, Muntau AC, Gersting SW. Long-Term Antibody Response to SARS-CoV-2 in Children. J Clin Immunol 2023; 43:46-56. [PMID: 36121535 PMCID: PMC9483535 DOI: 10.1007/s10875-022-01355-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 08/18/2022] [Indexed: 01/21/2023]
Abstract
Almost 2 years into the pandemic and with vaccination of children significantly lagging behind adults, long-term pediatric humoral immune responses to SARS-CoV-2 are understudied. The C19.CHILD Hamburg (COVID-19 Child Health Investigation of Latent Disease) Study is a prospective cohort study designed to identify and follow up children and their household contacts infected in the early 2020 first wave of SARS-CoV-2. We screened 6113 children < 18 years by nasopharyngeal swab-PCR in a low-incidence setting after general lockdown, from May 11 to June 30, 2020. A total of 4657 participants underwent antibody testing. Positive tests were followed up by repeated PCR and serological testing of all household contacts over 6 months. In total, the study identified 67 seropositive children (1.44%); the median time after infection at first presentation was 83 days post-symptom onset (PSO). Follow-up of household contacts showed less than 100% seroprevalence in most families, with higher seroprevalence in families with adult index cases compared to pediatric index cases (OR 1.79, P = 0.047). Most importantly, children showed sustained seroconversion up to 9 months PSO, and serum antibody concentrations persistently surpassed adult levels (ratio serum IgG spike children vs. adults 90 days PSO 1.75, P < 0.001; 180 days 1.38, P = 0.01; 270 days 1.54, P = 0.001). In a low-incidence setting, SARS-CoV-2 infection and humoral immune response present distinct patterns in children including higher antibody levels, and lower seroprevalence in families with pediatric index cases. Children show long-term SARS-CoV-2 antibody responses. These findings are relevant to novel variants with increased disease burden in children, as well as for the planning of age-appropriate vaccination strategies.
Collapse
Affiliation(s)
- Gabor A. Dunay
- University Children’s Research, UCR@Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Madalena Barroso
- University Children’s Research, UCR@Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Mathias Woidy
- University Children’s Research, UCR@Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Marta K. Danecka
- University Children’s Research, UCR@Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Geraldine Engels
- University Children’s Research, UCR@Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Katharina Hermann
- Department of Pediatrics, Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Friederike S. Neumann
- University Children’s Research, UCR@Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Kevin Paul
- University Children’s Research, UCR@Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Jan Beime
- Department of Pediatrics, Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Gabriele Escherich
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Kristin Fehse
- University Children’s Research, UCR@Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Lev Grinstein
- Department of Pediatrics, Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Franziska Haniel
- Department of Pediatric Cardiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Luka J. Haupt
- University Children’s Research, UCR@Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Laura Hecher
- Department of Pediatrics, Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Torben Kehl
- Department of Pediatric Cardiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Christoph Kemen
- Wilhelmstift Children’s Hospital, Liliencronstraße 130, 22149 Hamburg, Germany
| | - Markus J. Kemper
- Asklepios Klinik Nord – Heidberg, Tangstedter Landstraße 400, 22417 Hamburg, Germany
| | - Robin Kobbe
- Institute for Infection Research and Vaccine Development (IIRVD), University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Aloisa Kohl
- Department of Pediatrics, Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Thomas Klokow
- University Children’s Research, UCR@Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Dominik Nörz
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Jakob Olfe
- Department of Pediatric Cardiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Friderike Schlenker
- Department of Pediatrics, Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Jessica Schmiesing
- University Children’s Research, UCR@Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Johanna Schrum
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Freya Sibbertsen
- University Children’s Research, UCR@Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Philippe Stock
- Altona Children’s Hospital, Bleickenallee 38, 22763 Hamburg, Germany
| | - Stephan Tiede
- University Children’s Research, UCR@Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Eik Vettorazzi
- Institute of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Dimitra E. Zazara
- Department of Pediatrics, Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany ,Department of Obstetrics and Prenatal Medicine, Division for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Antonia Zapf
- Institute of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Marc Lütgehetmann
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Jun Oh
- Department of Pediatrics, Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Thomas S. Mir
- Department of Pediatric Cardiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Ania C. Muntau
- Department of Pediatrics, Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | - Søren W. Gersting
- University Children’s Research, UCR@Kinder-UKE, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251 Hamburg, Germany
| | | |
Collapse
|
21
|
Song T, Chen C, Bao S, Du B, Wang X, Liu J, Wang F, Ma W, Yao G, Wan X, Zhang X, Wang J, Jiang H. An immobilization-based, loop-mediated isothermal amplification device for nucleic acid detection of SARS-CoV-2 N gene. BIOTECHNOL BIOTEC EQ 2022. [DOI: 10.1080/13102818.2022.2134822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Affiliation(s)
- Tianyu Song
- State Key Laboratory of NBC Protection for Civilian, Beijing, PR China
| | - Chang Chen
- State Key Laboratory of NBC Protection for Civilian, Beijing, PR China
| | - Shaoheng Bao
- State Key Laboratory of NBC Protection for Civilian, Beijing, PR China
| | - Bin Du
- State Key Laboratory of NBC Protection for Civilian, Beijing, PR China
| | - Xiaokun Wang
- State Key Laboratory of NBC Protection for Civilian, Beijing, PR China
| | - Jiajia Liu
- State Key Laboratory of NBC Protection for Civilian, Beijing, PR China
| | - Fuli Wang
- State Key Laboratory of NBC Protection for Civilian, Beijing, PR China
| | - Wei Ma
- State Key Laboratory of NBC Protection for Civilian, Beijing, PR China
| | - Ge Yao
- State Key Laboratory of NBC Protection for Civilian, Beijing, PR China
| | - Xiukun Wan
- State Key Laboratory of NBC Protection for Civilian, Beijing, PR China
| | - Xinlong Zhang
- State Key Laboratory of NBC Protection for Civilian, Beijing, PR China
| | - Jingjing Wang
- State Key Laboratory of NBC Protection for Civilian, Beijing, PR China
| | - Hui Jiang
- State Key Laboratory of NBC Protection for Civilian, Beijing, PR China
| |
Collapse
|
22
|
Guan PC, Zhang H, Li ZY, Xu SS, Sun M, Tian XM, Ma Z, Lin JS, Gu MM, Wen H, Zhang FL, Zhang YJ, Yu GJ, Yang C, Wang ZX, Song Y, Li JF. Rapid Point-of-Care Assay by SERS Detection of SARS-CoV-2 Virus and Its Variants. Anal Chem 2022; 94:17795-17802. [PMID: 36511436 PMCID: PMC9762416 DOI: 10.1021/acs.analchem.2c03437] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/25/2022] [Indexed: 12/15/2022]
Abstract
Addressing the spread of coronavirus disease 2019 (COVID-19) has highlighted the need for rapid, accurate, and low-cost diagnostic methods that detect specific antigens for SARS-CoV-2 infection. Tests for COVID-19 are based on reverse transcription PCR (RT-PCR), which requires laboratory services and is time-consuming. Here, by targeting the SARS-CoV-2 spike protein, we present a point-of-care SERS detection platform that specifically detects SARS-CoV-2 antigen in one step by captureing substrates and detection probes based on aptamer-specific recognition. Using the pseudovirus, without any pretreatment, the SARS-CoV-2 virus and its variants were detected by a handheld Raman spectrometer within 5 min. The limit of detection (LoD) for the pseudovirus was 124 TU μL-1 (18 fM spike protein), with a linear range of 250-10,000 TU μL-1. Moreover, this assay can specifically recognize the SARS-CoV-2 antigen without cross reacting with specific antigens of other coronaviruses or influenza A. Therefore, the platform has great potential for application in rapid point-of-care diagnostic assays for SARS-CoV-2.
Collapse
Affiliation(s)
- Peng-Cheng Guan
- College
of Materials, State Key Laboratory for Physical Chemistry of Solid
Surfaces, iChEM, MOE Key Laboratory of Spectrochemical Analysis and
Instrumentation, College of Chemistry and Chemical Engineering, College
of Energy, The First Affiliated Hospital, Xiamen University, Xiamen 361005, China
| | - Hong Zhang
- Shanghai
Children’s Hospital, Shanghai Jiao
Tong University, Shanghai 200062, China
| | - Zhi-Yong Li
- College
of Materials, State Key Laboratory for Physical Chemistry of Solid
Surfaces, iChEM, MOE Key Laboratory of Spectrochemical Analysis and
Instrumentation, College of Chemistry and Chemical Engineering, College
of Energy, The First Affiliated Hospital, Xiamen University, Xiamen 361005, China
| | - Shan-Shan Xu
- College
of Materials, State Key Laboratory for Physical Chemistry of Solid
Surfaces, iChEM, MOE Key Laboratory of Spectrochemical Analysis and
Instrumentation, College of Chemistry and Chemical Engineering, College
of Energy, The First Affiliated Hospital, Xiamen University, Xiamen 361005, China
| | - Miao Sun
- College
of Materials, State Key Laboratory for Physical Chemistry of Solid
Surfaces, iChEM, MOE Key Laboratory of Spectrochemical Analysis and
Instrumentation, College of Chemistry and Chemical Engineering, College
of Energy, The First Affiliated Hospital, Xiamen University, Xiamen 361005, China
| | - Xian-Min Tian
- Shanghai
Children’s Hospital, Shanghai Jiao
Tong University, Shanghai 200062, China
| | - Zhan Ma
- Shanghai
Children’s Hospital, Shanghai Jiao
Tong University, Shanghai 200062, China
| | - Jia-Sheng Lin
- College
of Materials, State Key Laboratory for Physical Chemistry of Solid
Surfaces, iChEM, MOE Key Laboratory of Spectrochemical Analysis and
Instrumentation, College of Chemistry and Chemical Engineering, College
of Energy, The First Affiliated Hospital, Xiamen University, Xiamen 361005, China
| | - Man-Man Gu
- Department
of Optics and Electronic Technology, China
Jiliang University, Hangzhou 310018, China
| | - Huan Wen
- College
of Materials, State Key Laboratory for Physical Chemistry of Solid
Surfaces, iChEM, MOE Key Laboratory of Spectrochemical Analysis and
Instrumentation, College of Chemistry and Chemical Engineering, College
of Energy, The First Affiliated Hospital, Xiamen University, Xiamen 361005, China
| | - Fan-Li Zhang
- Department
of Optics and Electronic Technology, China
Jiliang University, Hangzhou 310018, China
| | - Yue-Jiao Zhang
- College
of Materials, State Key Laboratory for Physical Chemistry of Solid
Surfaces, iChEM, MOE Key Laboratory of Spectrochemical Analysis and
Instrumentation, College of Chemistry and Chemical Engineering, College
of Energy, The First Affiliated Hospital, Xiamen University, Xiamen 361005, China
| | - Guang-Jun Yu
- Shanghai
Children’s Hospital, Shanghai Jiao
Tong University, Shanghai 200062, China
| | - Chaoyong Yang
- College
of Materials, State Key Laboratory for Physical Chemistry of Solid
Surfaces, iChEM, MOE Key Laboratory of Spectrochemical Analysis and
Instrumentation, College of Chemistry and Chemical Engineering, College
of Energy, The First Affiliated Hospital, Xiamen University, Xiamen 361005, China
- Innovation
Laboratory for Sciences and Technologies of Energy Materials of Fujian
Province (IKKEM), Xiamen 361005, China
| | - Zhan-Xiang Wang
- College
of Materials, State Key Laboratory for Physical Chemistry of Solid
Surfaces, iChEM, MOE Key Laboratory of Spectrochemical Analysis and
Instrumentation, College of Chemistry and Chemical Engineering, College
of Energy, The First Affiliated Hospital, Xiamen University, Xiamen 361005, China
| | - Yanling Song
- College
of Materials, State Key Laboratory for Physical Chemistry of Solid
Surfaces, iChEM, MOE Key Laboratory of Spectrochemical Analysis and
Instrumentation, College of Chemistry and Chemical Engineering, College
of Energy, The First Affiliated Hospital, Xiamen University, Xiamen 361005, China
| | - Jian-Feng Li
- College
of Materials, State Key Laboratory for Physical Chemistry of Solid
Surfaces, iChEM, MOE Key Laboratory of Spectrochemical Analysis and
Instrumentation, College of Chemistry and Chemical Engineering, College
of Energy, The First Affiliated Hospital, Xiamen University, Xiamen 361005, China
- Innovation
Laboratory for Sciences and Technologies of Energy Materials of Fujian
Province (IKKEM), Xiamen 361005, China
- Department
of Optics and Electronic Technology, China
Jiliang University, Hangzhou 310018, China
| |
Collapse
|
23
|
Chensue SW, Siler AF, Kim PS, Dimcheff DE, Daghfal DJ, Prostko J, Frias E, Linder KA, Schildhouse RJ. SARS-CoV-2 Anti-Spike IgG Antibody and ACE2 Receptor Binding Inhibition Levels among Breakthrough Stage Veteran Patients. Microbiol Spectr 2022; 10:e0274722. [PMID: 36409132 PMCID: PMC9769865 DOI: 10.1128/spectrum.02747-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/29/2022] [Indexed: 11/23/2022] Open
Abstract
SARS-CoV-2 mRNA vaccines have been critical to curbing pandemic COVID-19; however, a major shortcoming has been the inability to assess levels of protection after vaccination. This study assessed serologic status of breakthrough infections in vaccinated patients at a Veterans Administration medical center from June through December 2021 during a SARS-CoV-2 delta variant wave. Breakthrough occurred mostly beyond 150 days after two-dose vaccination with a mean of 239 days. Anti-SARS-CoV-2 spike (S) IgG levels were low at 0 to 2 days postsymptoms but increased in subjects presenting thereafter. Population measurements of anti-S IgG and angiotensin converting enzyme-2 receptor (ACE2-R) binding inhibition among uninfected, vaccinated patients suggested immune decay occurred after 150 days with 62% having anti-S IgG levels at or below 1,000 AU comparable with breakthrough patients at 0 to 2 days postsymptom onset. In contrast, vaccination after resolved infection conferred robust enduring anti-S IgG levels (5,000 to >50,000 AU) with >90% ACE2-R binding inhibition. However, monoclonal antibody (MAb)-treated patients did not benefit from their prior infection suggesting impaired establishment of B cell memory. Analysis of boosted patients confirmed the benefit of a third vaccine dose with most having anti-S IgG levels above 5,000 AU with >90% ACE2-R binding inhibition, but a subset had levels <5,000 AU. Anti-S IgG levels >5,000 AU were associated with >90% ACE2-R binding inhibition and no documented breakthrough infections, whereas levels falling below 5,000 AU and approaching 1,000 AU were associated with breakthrough infections. Thus, quantitative antibody measurements may provide a means to guide vaccination intervals for the individual. IMPORTANCE Currently, clinicians have no guidance for the serologic assessment of SARS-Cov-2 postvaccination status regarding protection and risk of infection. Vaccination and boosters are administered blindly without evaluation of need or outcome at the individual level. The recent development of automated quantitative assays for anti-SARS-CoV-2 spike protein IgG antibodies permits accurate measurement of humoral immunity in standardized units. Clinical studies, such as reported here, will help establish protective antibody levels allowing identification and targeted management of poor vaccine responders and vaccinated subjects undergoing immune decay.
Collapse
Affiliation(s)
- Stephen W. Chensue
- VA Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | - Paul S. Kim
- VA Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
- Division of Hospital Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Derek E. Dimcheff
- VA Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
- Division of Hospital Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - David J. Daghfal
- Abbott Laboratories, CoreLab Division, Abbott Park, Illinois, USA
| | - John Prostko
- Abbott Laboratories, CoreLab Division, Abbott Park, Illinois, USA
| | - Edwin Frias
- Abbott Laboratories, CoreLab Division, Abbott Park, Illinois, USA
| | - Kathleen A. Linder
- VA Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
- Division of Infectious Disease, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Richard J. Schildhouse
- VA Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
- Division of Hospital Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
24
|
Knobloch JK, Pfefferle S, Lütgehetmann M, Nörz D, Klupp EM, Belmar Campos CE, Kluge S, Aepfelbacher M, Knobling B, Franke G. Infectivity of SARS-CoV-2 on Inanimate Surfaces: Don't Trust Ct Value. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:17074. [PMID: 36554950 PMCID: PMC9779331 DOI: 10.3390/ijerph192417074] [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: 11/03/2022] [Revised: 12/08/2022] [Accepted: 12/16/2022] [Indexed: 06/17/2023]
Abstract
SARS-CoV-2 RNA is frequently identified in patient rooms and it was speculated that the viral load quantified by PCR might correlate with infectivity of surfaces. To evaluate Ct values for the prediction of infectivity, we investigated contaminated surfaces and Ct-value changes after disinfection. Viral RNA was detected on 37 of 143 investigated surfaces of an ICU. However, virus isolation failed for surfaces with a high viral RNA load. Also, SARS-CoV-2 could not be cultivated from surfaces artificially contaminated with patient specimens. In order to evaluate the significance of Ct values more precisely, we used surrogate enveloped bacteriophage Φ6. A strong reduction in Φ6 was achieved by three different disinfection methods. Despite a strong reduction in viability almost no change in the Ct values was observed for UV-C and alcoholic surface disinfectant. Disinfection using ozone resulted in a lack of Φ6 recovery as well as a detectable shift in Ct values indicating strong degradation of the viral RNA. The observed lack of significant effects on the detectable viral RNA after effective disinfection suggest that quantitative PCR is not suitable for predicting the infectivity of SARS-CoV-2 on inanimate surfaces. Ct values should therefore not be considered as markers for infectivity in this context.
Collapse
Affiliation(s)
- Johannes K. Knobloch
- Institute for Medical Microbiology, Virology and Hygiene, Department for Infection Prevention and Control, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Susanne Pfefferle
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Marc Lütgehetmann
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Dominik Nörz
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Eva M. Klupp
- Institute for Medical Microbiology, Virology and Hygiene, Department for Infection Prevention and Control, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Cristina E. Belmar Campos
- Institute for Medical Microbiology, Virology and Hygiene, Department for Infection Prevention and Control, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Stefan Kluge
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Martin Aepfelbacher
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Birte Knobling
- Institute for Medical Microbiology, Virology and Hygiene, Department for Infection Prevention and Control, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Gefion Franke
- Institute for Medical Microbiology, Virology and Hygiene, Department for Infection Prevention and Control, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| |
Collapse
|
25
|
Development of an optimized colorimetric RT-LAMP for SARS-CoV-2 assay with enhanced procedure controls for remote diagnostics. Sci Rep 2022; 12:21424. [PMID: 36503901 PMCID: PMC9741705 DOI: 10.1038/s41598-022-25872-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
The coronavirus pandemic accentuated the need for molecular diagnostic tests. A technique highly used to this end is the Polymerase Chain Reaction (PCR)-a sensitive and specific technique commonly used as the gold standard for molecular diagnostics. However, it demands highly trained personnel and high-maintenance equipment and is relatively time-consuming. An alternative is the Loop-Mediated Isothermal Amplification (LAMP) technique, which doesn't need sample purification or expensive equipment, and is similar to PCR when compared in sensitivity and specificity. In this paper, we developed an optimized colorimetric Reverse Transcriptase Loop-Mediated Isothermal Amplification (RT-LAMP) Point-of-Care test using a portable device to diagnose COVID-19. Variables such as concentration of primers, magnesium sulfate, betaine, hydrochloride guanidine, Bst, and temperature of the reactions were tested. We also created a pipetting quality control system-using a combination of dyes-to avoid false negatives due to a lack of samples added to the reaction test tube. Mineral oil was incorporated in the composition of the RT-LAMP reactions to avoid evaporation when a heating lid isn't available. The final RT-LAMP test is tenfold more sensitive when compared to the WarmStart Colorimetric Master mix from New England Biolabs with a sensitivity of 5 copies per μL.
Collapse
|
26
|
Yilmaz A, Turan N, Kocazeybek BS, Dinc HO, Tali HE, Aydin O, Tali HB, Yilmaz SG, Konukoglu D, Borekci S, Bold D, Roman Sosa G, Gungordu N, Vardaloglu I, Gareayaghi N, Guzel M, Guner E, Sadeyen JR, Chang P, Iqbal M, Richt JA, Yilmaz H. Development of in House ELISAs to Detect Antibodies to SARS-CoV-2 in Infected and Vaccinated Humans by Using Recombinant S, S1 and RBD Proteins. Diagnostics (Basel) 2022; 12:3085. [PMID: 36553092 PMCID: PMC9777145 DOI: 10.3390/diagnostics12123085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/24/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
(1) Background: The aim of this study was to produce in-house ELISAs which can be used to determine SARS-CoV-2-specific antibody levels directed against the spike protein (S), the S1 subunit of S and the receptor binding domain (RBD) of S in SARS-CoV-2 vaccinated and infected humans. (2) Methods: Three in-house ELISAs were developed by using recombinant proteins of SARS-CoV-2, namely the S, S1 and RBD proteins. Specificity and sensitivity evaluations of these tests were performed using sera from SARS-CoV-2-infected (n = 70) and SARS-CoV-2-vaccinated (n = 222; CoronaVac vaccine) humans in Istanbul, Turkey. The analyses for the presence of SARS-CoV-2-specific antibodies were performed using the in-house ELISAs, a commercial ELISA (Abbott) and a commercial surrogate virus neutralization test (sVNT). We also analyzed archival human sera (n = 50) collected before the emergence of COVID-19 cases in Turkey. (3) Results: The sensitivity of the in-house S, S1 and RBD ELISAs was found to be 88.44, 90.17 and 95.38%, while the specificity was 72.27, 89.08 and 89.92%, respectively, when compared to the commercial SARS-CoV-2 antibody test kit. The area under curve (AUC) values were 0.777 for the in-house S ELISA, 0.926 for the S1 ELISA, and 0.959 for the RBD ELISA. The kappa values were 0.62, 0.79 and 0.86 for the S, S1 and RBD ELISAs, respectively. (4) Conclusions: The in-house S1 and RBD ELISAs developed in this study have acceptable performance characteristics in terms of sensitivity, specificity, AUC and kappa values. In particular, the RBD ELISA seems viable to determine SARS-CoV-2-specific antibody levels, both in infected and vaccinated people, and help mitigate SARS-CoV-2 outbreaks and spread.
Collapse
Affiliation(s)
- Aysun Yilmaz
- Department of Virology, Veterinary Faculty, Istanbul University-Cerrahpasa, Hadimkoy, Istanbul 34098, Turkey
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK
| | - Nuri Turan
- Department of Virology, Veterinary Faculty, Istanbul University-Cerrahpasa, Hadimkoy, Istanbul 34098, Turkey
| | - Bekir Sami Kocazeybek
- Department of Medical Microbiology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul 34098, Turkey
| | - Harika Oyku Dinc
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Bezmialem Vakif University, Istanbul 34098, Turkey
| | - Hasan Emre Tali
- Department of Virology, Veterinary Faculty, Istanbul University-Cerrahpasa, Hadimkoy, Istanbul 34098, Turkey
| | - Ozge Aydin
- Department of Virology, Veterinary Faculty, Istanbul University-Cerrahpasa, Hadimkoy, Istanbul 34098, Turkey
| | - Hamid Besim Tali
- Department of Virology, Veterinary Faculty, Istanbul University-Cerrahpasa, Hadimkoy, Istanbul 34098, Turkey
| | - Semaha Gul Yilmaz
- Department of Virology, Veterinary Faculty, Istanbul University-Cerrahpasa, Hadimkoy, Istanbul 34098, Turkey
| | - Dildar Konukoglu
- Department of Biocehmistry, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul 34098, Turkey
| | - Sermin Borekci
- Department of Pulmonary Diseases, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul 34098, Turkey
| | - Dashzeveg Bold
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Gleyder Roman Sosa
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Nejdiye Gungordu
- Department of Pulmonary Diseases, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul 34098, Turkey
- Department of Occupational Diseases, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul 34098, Turkey
| | - Ilgim Vardaloglu
- Department of Pulmonary Diseases, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul 34098, Turkey
| | - Nesrin Gareayaghi
- Sisli, Hamidiye Etfal Training and Research Hospiatal, Blood Center, Istanbul 34098, Turkey
| | - Mine Guzel
- Biruni Laboratories, Esentepe, Istanbul 34098, Turkey
| | - Ebru Guner
- Biruni Laboratories, Esentepe, Istanbul 34098, Turkey
| | | | - Pengxiang Chang
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK
| | - Munir Iqbal
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK
| | - Juergen A. Richt
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Huseyin Yilmaz
- Department of Virology, Veterinary Faculty, Istanbul University-Cerrahpasa, Hadimkoy, Istanbul 34098, Turkey
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK
| |
Collapse
|
27
|
Ferrobotic swarms enable accessible and adaptable automated viral testing. Nature 2022; 611:570-577. [PMID: 36352231 PMCID: PMC9645323 DOI: 10.1038/s41586-022-05408-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 10/04/2022] [Indexed: 11/10/2022]
Abstract
Expanding our global testing capacity is critical to preventing and containing pandemics1–9. Accordingly, accessible and adaptable automated platforms that in decentralized settings perform nucleic acid amplification tests resource-efficiently are required10–14. Pooled testing can be extremely efficient if the pooling strategy is based on local viral prevalence15–20; however, it requires automation, small sample volume handling and feedback not available in current bulky, capital-intensive liquid handling technologies21–29. Here we use a swarm of millimetre-sized magnets as mobile robotic agents (‘ferrobots’) for precise and robust handling of magnetized sample droplets and high-fidelity delivery of flexible workflows based on nucleic acid amplification tests to overcome these limitations. Within a palm-sized printed circuit board-based programmable platform, we demonstrated the myriad of laboratory-equivalent operations involved in pooled testing. These operations were guided by an introduced square matrix pooled testing algorithm to identify the samples from infected patients, while maximizing the testing efficiency. We applied this automated technology for the loop-mediated isothermal amplification and detection of the SARS-CoV-2 virus in clinical samples, in which the test results completely matched those obtained off-chip. This technology is easily manufacturable and distributable, and its adoption for viral testing could lead to a 10–300-fold reduction in reagent costs (depending on the viral prevalence) and three orders of magnitude reduction in instrumentation cost. Therefore, it is a promising solution to expand our testing capacity for pandemic preparedness and to reimagine the automated clinical laboratory of the future. A handheld printed circuit board-based programmable platform using ferrobots can perform the complex, laboratory-equivalent procedures involved in multiplexed and pooled nucleic acid amplification testing, allowing for the decentralization of viral diagnostics.
Collapse
|
28
|
Loop-Mediated Isothermal Amplification-Based Microfluidic Platforms for the Detection of Viral Infections. Curr Infect Dis Rep 2022; 24:205-215. [PMID: 36341307 PMCID: PMC9628606 DOI: 10.1007/s11908-022-00790-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2022] [Indexed: 11/09/2022]
Abstract
Purpose of Review Easy-to-use, fast, and accurate virus detection method is essential for patient management and epidemic surveillance, especially during severe pandemics. Loop-mediated isothermal amplification (LAMP) on a microfluidic platform is suitable for detecting infectious viruses, regardless of the availability of medical resources. The purpose of this review is to introduce LAMP-based microfluidic devices for virus detection, including their detection principles, methods, and application. Recent Findings Facing the uncontrolled spread of viruses, the large-scale deployment of LAMP-based microfluidic platforms at the grassroots level can help expand the coverage of nucleic acid testing and shorten the time to obtain test reports. Microfluidic chip technology is highly integrated and miniaturized, enabling precise fluid control for effective virus detection. Performing LAMP on miniaturized systems can reduce analysis time, reagent consumption and risk of sample contamination, and improve analytical performance. Summary Compared to traditional benchtop protocols, LAMP-based microfluidic devices reduce the testing time, reagent consumption, and the risk of sample contamination. In addition to simultaneous detection of multiple target genes by special channel design, microfluidic chips can also integrate digital LAMP to achieve absolute quantification of target genes.
Collapse
|
29
|
Lin Q, Zhang J, Liu L, Kong J, Fang X. Simultaneous Rapid Nucleic Acid and Protein Detection in a Lateral Chromatography Chip for COVID-19 Diagnosis. ACS OMEGA 2022; 7:38409-38416. [PMID: 36340144 PMCID: PMC9631717 DOI: 10.1021/acsomega.2c03499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 08/16/2022] [Indexed: 06/16/2023]
Abstract
In this work, we report a fast, portable, and economical microfluidic platform for the simultaneous detection of nucleic acid and proteins. Using SARS-CoV-2 as a target, this microfluidic chip enabled to simultaneously detect the SARS-CoV-2 RNA (N gene) antigen (or specific IgG antibody) with respective detection limits of 1 copy/μL for nucleic acid, 0.85 ng/mL for antigen, and 5.80 ng/mL for IgG within 30 min with high stability and anti-interference ability. The capability of this system in clinical applications was further evaluated using clinical samples, displaying 100% sensitivity and 100% specificity for COVID-19 diagnosis. These findings demonstrate the potential of this method to be used for the detection and subsequent control of pathogens.
Collapse
Affiliation(s)
- Qiuyuan Lin
- Department
of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, P. R. China
| | - Jin Zhang
- Qingdao
International Travel Healthcare Center, Qingdao Customs, Qingdao 266071, P. R. China
| | - Liling Liu
- Shanghai
Suxin Biotechnology Co. Ltd., and IgeneTec Diagnostic Products Co.
Ltd., Shanghai 201318, P. R. China
| | - Jilie Kong
- Department
of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, P. R. China
| | - Xueen Fang
- Department
of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, P. R. China
| |
Collapse
|
30
|
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
|
31
|
Heyer A, Günther T, Robitaille A, Lütgehetmann M, Addo MM, Jarczak D, Kluge S, Aepfelbacher M, Schulze Zur Wiesch J, Fischer N, Grundhoff A. Remdesivir-induced emergence of SARS-CoV2 variants in patients with prolonged infection. Cell Rep Med 2022; 3:100735. [PMID: 36075217 PMCID: PMC9378267 DOI: 10.1016/j.xcrm.2022.100735] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/19/2022] [Accepted: 08/12/2022] [Indexed: 04/09/2023]
Abstract
We here investigate the impact of antiviral treatments such as remdesivir on intra-host genomic diversity and emergence of SARS-CoV2 variants in patients with a prolonged course of infection. Sequencing and variant analysis performed in 112 longitudinal respiratory samples from 14 SARS-CoV2-infected patients with severe disease progression show that major frequency variants do not generally arise during prolonged infection. However, remdesivir treatment can increase intra-host genomic diversity and result in the emergence of novel major variant species harboring fixed mutations. This is particularly evident in a patient with B cell depletion who rapidly developed mutations in the RNA-dependent RNA polymerase gene following remdesivir treatment. Remdesivir treatment-associated emergence of novel variants is of great interest in light of current treatment guidelines for hospitalized patients suffering from severe SARS-CoV2 disease, as well as the potential use of remdesivir to preventively treat non-hospitalized patients at high risk for severe disease progression.
Collapse
Affiliation(s)
- Andreas Heyer
- I. Department of Medicine, Gastroenterology and Hepatology, Sections of Infectious Diseases and Tropical Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | - Marc Lütgehetmann
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marylyn M Addo
- I. Department of Medicine, Gastroenterology and Hepatology, Sections of Infectious Diseases and Tropical Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Center for Infection Research (DZIF), Hamburg-Borstel-Lübeck-Riems, Germany; Institute of Infection Research and Vaccine Development (IIRVD), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dominik Jarczak
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Kluge
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Aepfelbacher
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julian Schulze Zur Wiesch
- I. Department of Medicine, Gastroenterology and Hepatology, Sections of Infectious Diseases and Tropical Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Center for Infection Research (DZIF), Hamburg-Borstel-Lübeck-Riems, Germany
| | - Nicole Fischer
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | | |
Collapse
|
32
|
Nörz D, Tang HT, Emmerich P, Giersch K, Fischer N, Schmiedel S, Addo MM, Aepfelbacher M, Pfefferle S, Lütgehetmann M. Rapid Adaptation of Established High-Throughput Molecular Testing Infrastructure for Monkeypox Virus Detection. Emerg Infect Dis 2022; 28:1765-1769. [PMID: 35905463 PMCID: PMC9423910 DOI: 10.3201/eid2809.220917] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Beginning in May 2022, a rising number of monkeypox cases were reported in non-monkeypox-endemic countries in the Northern Hemisphere. We adapted 2 published quantitative PCRs for use as a dual-target monkeypox virus test on widely used automated high-throughput PCR systems. We determined analytic performance by serial dilutions of monkeypox virus reference material, which we quantified by digital PCR. We found the lower limit of detection for the combined assays was 4.795 (95% CI 3.6-8.6) copies/mL. We compared clinical performance against a commercial manual orthopoxvirus research use only PCR kit by using clinical remnant swab samples. Our assay showed 100% positive (n = 11) and 100% negative (n = 56) agreement. Timely and scalable PCR tests are crucial for limiting further spread of monkeypox. The assay we provide streamlines high-throughput molecular testing for monkeypox virus on existing broadly established platforms used for SARS-CoV-2 diagnostic testing.
Collapse
|
33
|
Browne DJ, Kelly AM, Brady JL, Doolan DL. A high-throughput screening RT-qPCR assay for quantifying surrogate markers of immunity from PBMCs. Front Immunol 2022; 13:962220. [PMID: 36110843 PMCID: PMC9469018 DOI: 10.3389/fimmu.2022.962220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
Immunoassays that quantitate cytokines and other surrogate markers of immunity from peripheral blood mononuclear cells (PBMCs), such as flow cytometry or Enzyme-Linked Immunosorbent Spot (ELIspot), allow highly sensitive measurements of immune effector function. However, those assays consume relatively high numbers of cells and expensive reagents, precluding comprehensive analyses and high-throughput screening (HTS). To address this issue, we developed a sensitive and specific reverse transcription-quantitative PCR (RT-qPCR)-based HTS assay, specifically designed to quantify surrogate markers of immunity from very low numbers of PBMCs. We systematically evaluated the volumes and concentrations of critical reagents within the RT-qPCR protocol, miniaturizing the assay and ultimately reducing the cost by almost 90% compared to current standard practice. We assessed the suitability of this cost-optimized RT-qPCR protocol as an HTS tool and determined the assay exceeds HTS uniformity and signal variance testing standards. Furthermore, we demonstrate this technique can effectively delineate a hierarchy of responses from as little as 50,000 PBMCs stimulated with CD4+ or CD8+ T cell peptide epitopes. Finally, we establish that this HTS-optimized protocol has single-cell analytical sensitivity and a diagnostic sensitivity equivalent to detecting 1:10,000 responding cells (i.e., 100 Spot Forming Cells/106 PBMCs by ELIspot) with over 90% accuracy. We anticipate this assay will have widespread applicability in preclinical and clinical studies, especially when samples are limited, and cost is an important consideration.
Collapse
|
34
|
Maia R, Carvalho V, Faria B, Miranda I, Catarino S, Teixeira S, Lima R, Minas G, Ribeiro J. Diagnosis Methods for COVID-19: A Systematic Review. MICROMACHINES 2022; 13:1349. [PMID: 36014271 PMCID: PMC9415914 DOI: 10.3390/mi13081349] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/11/2022] [Accepted: 08/16/2022] [Indexed: 05/15/2023]
Abstract
At the end of 2019, the coronavirus appeared and spread extremely rapidly, causing millions of infections and deaths worldwide, and becoming a global pandemic. For this reason, it became urgent and essential to find adequate tests for an accurate and fast diagnosis of this disease. In the present study, a systematic review was performed in order to provide an overview of the COVID-19 diagnosis methods and tests already available, as well as their evolution in recent months. For this purpose, the Science Direct, PubMed, and Scopus databases were used to collect the data and three authors independently screened the references, extracted the main information, and assessed the quality of the included studies. After the analysis of the collected data, 34 studies reporting new methods to diagnose COVID-19 were selected. Although RT-PCR is the gold-standard method for COVID-19 diagnosis, it cannot fulfill all the requirements of this pandemic, being limited by the need for highly specialized equipment and personnel to perform the assays, as well as the long time to get the test results. To fulfill the limitations of this method, other alternatives, including biological and imaging analysis methods, also became commonly reported. The comparison of the different diagnosis tests allowed to understand the importance and potential of combining different techniques, not only to improve diagnosis but also for a further understanding of the virus, the disease, and their implications in humans.
Collapse
Affiliation(s)
- Renata Maia
- Microelectromechanical Systems Research Unit (CMEMS-UMinho), School of Engineering, Campus de Azurém, University of Minho, Guimarães, Portugal
- LABBELS-Associate Laboratory, Braga/Guimarães, Portugal
| | - Violeta Carvalho
- Microelectromechanical Systems Research Unit (CMEMS-UMinho), School of Engineering, Campus de Azurém, University of Minho, Guimarães, Portugal
- LABBELS-Associate Laboratory, Braga/Guimarães, Portugal
- MEtRICs, Mechanical Engineering Department, Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal
- ALGORITMI, Production and Systems Department, School of Engineering, Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal
| | - Bernardo Faria
- Microelectromechanical Systems Research Unit (CMEMS-UMinho), School of Engineering, Campus de Azurém, University of Minho, Guimarães, Portugal
- LABBELS-Associate Laboratory, Braga/Guimarães, Portugal
| | - Inês Miranda
- Microelectromechanical Systems Research Unit (CMEMS-UMinho), School of Engineering, Campus de Azurém, University of Minho, Guimarães, Portugal
- LABBELS-Associate Laboratory, Braga/Guimarães, Portugal
| | - Susana Catarino
- Microelectromechanical Systems Research Unit (CMEMS-UMinho), School of Engineering, Campus de Azurém, University of Minho, Guimarães, Portugal
- LABBELS-Associate Laboratory, Braga/Guimarães, Portugal
| | - Senhorinha Teixeira
- ALGORITMI, Production and Systems Department, School of Engineering, Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal
| | - Rui Lima
- MEtRICs, Mechanical Engineering Department, Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal
- CEFT, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
- ALiCE, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
| | - Graça Minas
- Microelectromechanical Systems Research Unit (CMEMS-UMinho), School of Engineering, Campus de Azurém, University of Minho, Guimarães, Portugal
- LABBELS-Associate Laboratory, Braga/Guimarães, Portugal
| | - João Ribeiro
- ALiCE, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
- Campus de Santa Apolónia, Instituto Politécnico de Bragança, 5300-253 Bragança, Portugal
- Centro de Investigação de Montanha (CIMO), Campus de Santa Apolónia, Instituto Politécnico de Bragança, 5300-253 Bragança, Portugal
- Laboratório Associado para a Sustentabilidade e Tecnologia em Regiões de Montanha (SusTEC), Campus de Santa Apolónia, Instituto Politécnico de Bragança, 5300-253 Bragança, Portugal
| |
Collapse
|
35
|
Bouchra B, Wissal M, Amine C, Wail H, Younes Z, Hakima K, Meryem S, Hanae D, Khaoula H, Afaf A, Amina B. RAPID IMPLEMENTATION OF REAL-TIME REVERSE-TRANSCRIPTION POLYMERASE CHAIN REACTION (REAL-TIME RT-PCR) ASSAY FOR THE DETECTION OF SARS-COV-2 IN A MOROCCAN HOSPITAL. Afr J Infect Dis 2022; 16:72-77. [PMID: 36124329 PMCID: PMC9480888 DOI: 10.21010/ajid.v16i2s.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 11/07/2022] Open
Abstract
Background The main challenge faced in the African countries was to implement efficient molecular diagnostic facilities and start Covid-19 diagnosis as fast as possible to handle the rapid and unpredictable rise of cases. Materials Methods and Results We describe our experience in implementing a molecular biology unit at Sheikh Zaïd International University Hospital in Rabat, with a delay as short as one week, and starting real-time RT-PCR assay for the detection of SARS-Cov-2 infection, since the outbreak widened in Morocco in mid-March, 2020. Conclusion The challenges encountered in the first period of Covid-19 pandemic are still present. This work aims to give an example of a rapid and adaptive response in order to maintain our diagnosis ability for Covid-19 and for other pathogens.
Collapse
Affiliation(s)
- Belefquih Bouchra
- Biolife Medical Analysis Laboratory, Harhoura, Temara, Morocco,Microbiology Laboratory, Cheikh-Zaid University Hospital, Abulcasis University of Health Sciences, Rabat, Morocco,Corresponding author’s E-Mail:
| | - Maher Wissal
- Microbiology Laboratory, Cheikh-Zaid University Hospital, Abulcasis University of Health Sciences, Rabat, Morocco
| | - Cheikh Amine
- Microbiology Laboratory, Cheikh-Zaid University Hospital, Abulcasis University of Health Sciences, Rabat, Morocco
| | - Hamdani Wail
- Microbiology Laboratory, Cheikh-Zaid University Hospital, Abulcasis University of Health Sciences, Rabat, Morocco
| | - Zaid Younes
- Microbiology Laboratory, Cheikh-Zaid University Hospital, Abulcasis University of Health Sciences, Rabat, Morocco,Laboratory of Biology of Human Pathologies, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| | - Kabbaj Hakima
- Central Virology Laboratory, Ibn Sina University Hospital Center, Rabat Morocco
| | - Seffar Meryem
- Central Virology Laboratory, Ibn Sina University Hospital Center, Rabat Morocco
| | - Dakka Hanae
- Biolife Medical Analysis Laboratory, Harhoura, Temara, Morocco,Microbiology Laboratory, Cheikh-Zaid University Hospital, Abulcasis University of Health Sciences, Rabat, Morocco,Laboratory of Biology of Human Pathologies, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| | - Hadami Khaoula
- Biolife Medical Analysis Laboratory, Harhoura, Temara, Morocco
| | - Allaoui Afaf
- Microbiology Laboratory, Cheikh-Zaid University Hospital, Abulcasis University of Health Sciences, Rabat, Morocco
| | - Benouda Amina
- Microbiology Laboratory, Cheikh-Zaid University Hospital, Abulcasis University of Health Sciences, Rabat, Morocco
| |
Collapse
|
36
|
Mukherjee MD, Kumar A, Solanki PR, Verma D, Yadav AK, Chaudhary N, Kumar P. Recent Advances in Understanding SARS-CoV-2 Infection and Updates on
Potential Diagnostic and Therapeutics for COVID-19. CORONAVIRUSES 2022; 3. [DOI: 10.2174/2666796703666220302143102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 11/09/2021] [Accepted: 12/13/2021] [Indexed: 09/25/2023]
Abstract
Abstract:
A more focused approach is needed to understand the SARS-CoV-2 virulence, structure, and
genomics to devise more effective diagnostic and treatment interventions as this virus can evade the immune
attack and causes life-threatening complications such as cytokine storm. The spread of the virus is
still amplifying and causing thousands of new cases worldwide. It is essential to review current diagnostics
and treatment approaches to pave the way to correct or modify our current practices to make more
effective interventions against COVID-19. COVID-19 vaccine development has moved at a breakneck
pace since the outbreak began, utilizing practically all possible platforms or tactics to ensure the success
of vaccines. A total of 42 vaccine candidates have already entered clinical trials, including promising
results from numerous vaccine candidates in phase 1 or phase 2 trials. Further, many existing drugs are
being explored on broad-spectrum antiviral medications for their use in clinical recovery against COVID-
19. The present review attempts to re-examine the SARS-CoV-2 structure, its viral life cycle, clinical
symptoms and pathogenesis, mode of transmission, diagnostics, and treatment strategies that may be useful
for resorting to more effective approaches for controlling COVID-19. Various antiviral drugs and
vaccination strategies with their strengths and weaknesses are also discussed in the paper to augment our
understanding of COVID-19 management.
Collapse
Affiliation(s)
- Maumita D. Mukherjee
- Amity Institute of Applied Sciences, Amity University, Noida, Uttar Pradesh-201313, India
| | - Anil Kumar
- National Institute of Immunology, New Delhi-110067, India
| | - Pratima R. Solanki
- Nano-Bio Laboratory, Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi-110067, India
| | - Damini Verma
- Nano-Bio Laboratory, Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi-110067, India
- Amity Institute of Applied Sciences, Amity University, Noida, Uttar Pradesh-201313, India
| | - Amit K. Yadav
- Nano-Bio Laboratory, Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi-110067, India
| | - Navneet Chaudhary
- Department of Biotechnology,
Delhi Technological University, Delhi-110042, India
| | - Pramod Kumar
- Sri Aurobindo College, Delhi University, New Delhi-110017,
India
| |
Collapse
|
37
|
Grewal S, Syed Gurcoo M, Sudhan Sharma S. Comparative analysis of specificity and sensitivity between Cobas 6800 system and SARS-CoV-2 rRT-PCR to detect COVID-19 infection in clinical samples. Arch Microbiol 2022; 204:502. [PMID: 35852637 PMCID: PMC9295087 DOI: 10.1007/s00203-022-03118-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 11/25/2022]
Abstract
Fast and reliable testing for the COVID 19 infection is the need of the hour for the development of effective and reliable tools and assays. However, it is difficult to find the performance relativity among all these tests which are poorly understood. In this study, we aimed to evaluate the two different platforms where we determine the difference of sensitivity and specificity between the fully automated analyzer (Roche Diagnostics Cobas 6800 SARS-CoV-2 test) under FDA Emergency Use Authorization (EUA) and the laboratory designed test (SARS-CoV-2 rRT-PCR) based on the protocol developed by ICMR (Indian Council for Medical Research). The study was conducted for individual samples. We performed our study with two different approaches, first with validation method consisting of 188 samples (2 batches) on cobas 6800 instrument (Roche Molecular Systems, Branchburg, NJ) soon after we received US FDA EUA on 1 June 2021, all these samples were tested earlier with laboratory designed tests on 25th and 26th May 2021. Over all agreement between the two tests is of 88% and the coefficient of agreement between the two testing platform Cohen’sκ coefficient was found to be 0.76 (95% CI, 2.5897–13.4103) suggesting the substantial agreement between the two platforms. However, in some of the cases, both tests have shown a little disagreement. An overall discordance rate between two systems was found 11.1%. The difference may be due to the limit of detection, variation in the sequences of the primer design or may be due to other factors depicting the importance of comparing the two platforms used in the testing for SARS-CoV-2. Second approach includes head to head evaluation which comprises 1631 samples showed overall agreement of 99% and kappa value of 0.98. These results showed that cobas is effective and reliable assay for the detection of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Simmi Grewal
- Department of Microbiology, Government Medical College, Jammu, 180001, Jammu and Kashmir, India
| | - Mehreen Syed Gurcoo
- Department of Microbiology, Government Medical College, Jammu, 180001, Jammu and Kashmir, India
| | - Shashi Sudhan Sharma
- Department of Microbiology, Government Medical College, Jammu, 180001, Jammu and Kashmir, India.
| |
Collapse
|
38
|
Point-of-care COVID-19 testing: colorimetric diagnosis using rapid and ultra-sensitive ramified rolling circle amplification. Anal Bioanal Chem 2022; 414:5907-5915. [PMID: 35715585 PMCID: PMC9205388 DOI: 10.1007/s00216-022-04156-7] [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: 03/25/2022] [Revised: 05/17/2022] [Accepted: 05/30/2022] [Indexed: 12/24/2022]
Abstract
In this paper, we report a molecular diagnostic system—combining a colorimetric probe (RHthio-CuSO4) for pyrophosphate sensing and isothermal gene amplification (ramified rolling circle amplification)—that operates with high selectivity and sensitivity for clinical point-of-care diagnosis of SARS-CoV-2. During the polymerase phase of the DNA amplification process, pyrophosphate was released from the nucleotide triphosphate as a side product, which was then sensed by our RHthio-CuSO4 probe with a visible color change. This simple colorimetric diagnostic system allowed highly sensitive (1.13 copies/reaction) detection of clinical SARS-CoV-2 within 1 h, while also displaying high selectivity, as evidenced by its discrimination of two respiratory viral genomes (human rhino virus and respiratory syncytial virus) from that of SARS-CoV-2. All of the reactions in this system were performed at a single temperature, with positive identification being made by the naked eye, without requiring any instrumentation. The high sensitivity and selectivity, short detection time (1 h), simple treatment (one-pot reaction), isothermal amplification, and colorimetric detection together satisfy the requirements for clinical point-of-care detection of SARS-CoV-2. Therefore, we believe that this combination of a colorimetric probe and isothermal amplification will be useful for point-of-care testing to prevent the propagation of COVID-19.
Collapse
|
39
|
Brehm TT, Pfefferle S, von Possel R, Karolyi M, Zoufaly A, Wichmann D, Kobbe R, Emmerich P, Nörz D, Aepfelbacher M, Wiesch JSZ, Addo MM, Stefan, Schmiedel, Lütgehetmann M. Clinical efficacy and in vitro neutralization capacity of monoclonal antibodies for SARS-CoV-2 delta and omicron variants. J Med Virol 2022; 94:5038-5043. [PMID: 35662058 PMCID: PMC9347884 DOI: 10.1002/jmv.27916] [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: 05/06/2022] [Revised: 05/24/2022] [Accepted: 06/01/2022] [Indexed: 11/07/2022]
Abstract
We aimed to provide in vitro data on the neutralization capacity of different monoclonal antibody (mAb) preparations against the severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) delta and omicron variant, respectively, and describe the in vivo RNA kinetics of coronavirus disease 2019 (COVID‐19) patients treated with the respective mAbs. Virus neutralization assays were performed to assess the neutralizing effect of the mAb formulations casirivimab/imdevimab and sotrovimab on the SARS‐CoV‐2 delta and omicron variant. Additionally, respiratory tract SARS‐CoV‐2 RNA kinetics are provided for 25 COVID‐19 patients infected with either delta variant (n = 18) or omicron variant (n = 7) treated with the respective mAb formulations during their hospital stay. In the virus neutralization assay, sotrovimab exhibits neutralizing capacity at therapeutically achievable concentrations against the SARS‐CoV‐2 delta and omicron variant. In contrast, casivirimab/imdevimab had neutralizing capacity against the delta variant but failed neutralization against the omicron variant except for a very high concentration above the currently recommended therapeutic dosage. In patients with delta variant infections treated with casivirimab/imdevimab, we observed a rapid decrease of respiratory viral RNA at day 3 after mAb therapy. In contrast, no such prompt decline was observed in patients with delta variant or omicron variant infections receiving sotrovimab.
Collapse
Affiliation(s)
- Thomas Theo Brehm
- Division of Infectious Diseases, I. Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel Riems
| | - Susanne Pfefferle
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel Riems.,Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Straße 74, 20359, Hamburg, Germany.,Center for Diagnostics, Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronald von Possel
- Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Straße 74, 20359, Hamburg, Germany
| | - Mario Karolyi
- Department of Medicine 4, Klinik Favoriten, Vienna, Austria
| | - Alexander Zoufaly
- Department of Medicine 4, Klinik Favoriten, Vienna, Austria.,Faculty of Medicine, Sigmund Freud University, Vienna, Austria
| | - Dominic Wichmann
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Robin Kobbe
- Division of Infectious Diseases, I. Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Petra Emmerich
- Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Straße 74, 20359, Hamburg, Germany
| | - Dominik Nörz
- Center for Diagnostics, Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Aepfelbacher
- Center for Diagnostics, Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julian Schulze Zur Wiesch
- Division of Infectious Diseases, I. Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel Riems
| | - Marylyn M Addo
- Division of Infectious Diseases, I. Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel Riems.,Institute for Infection Research and Vaccine Development, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Schmiedel
- Division of Infectious Diseases, I. Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel Riems
| | - Marc Lütgehetmann
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel Riems.,Center for Diagnostics, Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
40
|
Molecularly Imprinted Polymer-Based Sensors for SARS-CoV-2: Where Are We Now? Biomimetics (Basel) 2022; 7:biomimetics7020058. [PMID: 35645185 PMCID: PMC9149885 DOI: 10.3390/biomimetics7020058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 01/27/2023] Open
Abstract
Since the first reported case of COVID-19 in 2019 in China and the official declaration from the World Health Organization in March 2021 as a pandemic, fast and accurate diagnosis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has played a major role worldwide. For this reason, various methods have been developed, comprising reverse transcriptase-polymerase chain reaction (RT-PCR), immunoassays, clustered regularly interspaced short palindromic repeats (CRISPR), reverse transcription loop-mediated isothermal amplification (RT-LAMP), and bio(mimetic)sensors. Among the developed methods, RT-PCR is so far the gold standard. Herein, we give an overview of the MIP-based sensors utilized since the beginning of the pandemic.
Collapse
|
41
|
Impact of Oral Rinsing with Octenidine Based Solution on SARS-CoV-2 Loads in Saliva of Infected Patients an Exploratory Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19095582. [PMID: 35564977 PMCID: PMC9099814 DOI: 10.3390/ijerph19095582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023]
Abstract
Objective: In this study, the in-vivo effect of an antiseptic mouth rinse with Octenisept plus phenoxyethanol (OCT + PE) on the oral SARS-CoV-2 load was investigated. Material and Methods: In eight COVID-19 patients, saliva samples were obtained before mouth rinsing and at five time points post rinsing with OCT + PE (n = 47 saliva samples in total). SARS-CoV-2 RNA was detected and quantified by RT-qPCR and virus isolation in cell culture was performed to assess for infectivity. Results: Immediately after mouth rinsing (1 min), a significant reduction of the SARS-CoV-2 RNA loads in saliva was achieved (p = 0.03) with 7/8 participants having SARS-CoV-2 RNA levels undetectable by RT-qPCR. At later time points, RNA levels returned to baseline levels in all study participants. Infectivity of saliva samples was demonstrated by successful virus isolation from saliva samples collected at later time points. Conclusions: This study highlights that saliva samples from COVID-19 patients are infectious and demonstrates that mouth rinsing with OCT + PE temporarily leads to a significant reduction of the SARS-CoV-2 load in saliva. Clinical relevance: Mouth rinsing with OCT + PE could provide a simple, rapid, and efficient method for SARS-CoV-2 infection prevention, particularly in the field of dental and respiratory medicine
Collapse
|
42
|
DeFina SM, Wang J, Yang L, Zhou H, Adams J, Cushing W, Tuohy B, Hui P, Liu C, Pham K. SaliVISION: a rapid saliva-based COVID-19 screening and diagnostic test with high sensitivity and specificity. Sci Rep 2022; 12:5729. [PMID: 35388102 PMCID: PMC8986854 DOI: 10.1038/s41598-022-09718-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 03/21/2022] [Indexed: 12/12/2022] Open
Abstract
The Coronavirus disease 2019 (COVID-19) pandemic-caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)– has posed a global threat and presented with it a multitude of economic and public-health challenges. Establishing a reliable means of readily available, rapid diagnostic testing is of paramount importance in halting the spread of COVID-19, as governments continue to ease lockdown restrictions. The current standard for laboratory testing utilizes reverse transcription quantitative polymerase chain reaction (RT-qPCR); however, this method presents clear limitations in requiring a longer run-time as well as reduced on-site testing capability. Therefore, we investigated the feasibility of a reverse transcription looped-mediated isothermal amplification (RT-LAMP)-based model of rapid COVID-19 diagnostic testing which allows for less invasive sample collection, named SaliVISION. This novel, two-step, RT-LAMP assay utilizes a customized multiplex primer set specifically targeting SARS-CoV-2 and a visual report system that is ready to interpret within 40 min from the start of sample processing and does not require a BSL-2 level testing environment or special laboratory equipment. When compared to the SalivaDirect and Thermo Fisher Scientific TaqPath RT-qPCR testing platforms, the respective sensitivities of the SaliVISION assay are 94.29% and 98.28% while assay specificity was 100% when compared to either testing platform. Our data illustrate a robust, rapid diagnostic assay in our novel RT-LAMP test design, with potential for greater testing throughput than is currently available through laboratory testing and increased on-site testing capability.
Collapse
Affiliation(s)
- Samuel M DeFina
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Jianhui Wang
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Lei Yang
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Han Zhou
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Jennifer Adams
- Department of Laboratory Medicine, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - William Cushing
- Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT, USA.,Yale New Haven Hospital, New Haven, CT, USA
| | - Beth Tuohy
- Yale University Health Services, Yale University, New Haven, CT, USA
| | - Pei Hui
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Chen Liu
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA.
| | - Kien Pham
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA.
| |
Collapse
|
43
|
Choi MH, Lee J, Seo YJ. Dual-site ligation-assisted loop-mediated isothermal amplification (dLig-LAMP) for colorimetric and point-of-care determination of real SARS-CoV-2. Mikrochim Acta 2022; 189:176. [PMID: 35381892 PMCID: PMC8982663 DOI: 10.1007/s00604-022-05293-7] [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: 12/19/2021] [Accepted: 03/21/2022] [Indexed: 12/24/2022]
Abstract
A probing system has been developed based on dual-site ligation-assisted loop-mediated isothermal amplification (dLig-LAMP) for the selective colorimetric detection of SARS-CoV-2. This approach can induce false-positive and -negative detection in real clinical samples; dLig-LAMP operates with improved selectivity. Unlike RT-LAMP, the selectivity of dLig-LAMP is determined in both the ligation and primer binding steps, not in the reverse transcription step. With this selective system in hand, we developed a colorimetric signaling system for point-of-care detection. We also developed a colorimetric probe for sensing pyrophosphate, which arises as a side product during the LAMP DNA amplification. Thus, dLig-LAMP appears to be an alternative method for improving the selectivity problems associated with reverse transcription. In addition, combining dLig-LAMP with colorimetric pyrophosphate probing allows point-of-care detection of SARS-CoV-2 within 1 h with high selectivity.
Collapse
Affiliation(s)
- Moon Hyeok Choi
- Department of Chemistry, Jeonbuk National University, Jeonju, 54896, South Korea
| | - Jaehyeon Lee
- Department of Laboratory Medicine, Jeonbuk National University Medical School and Hospital, Jeonju, 54896, South Korea
| | - Young Jun Seo
- Department of Chemistry, Jeonbuk National University, Jeonju, 54896, South Korea.
| |
Collapse
|
44
|
Wanner N, Andrieux G, Badia-I-Mompel P, Edler C, Pfefferle S, Lindenmeyer MT, Schmidt-Lauber C, Czogalla J, Wong MN, Okabayashi Y, Braun F, Lütgehetmann M, Meister E, Lu S, Noriega MLM, Günther T, Grundhoff A, Fischer N, Bräuninger H, Lindner D, Westermann D, Haas F, Roedl K, Kluge S, Addo MM, Huber S, Lohse AW, Reiser J, Ondruschka B, Sperhake JP, Saez-Rodriguez J, Boerries M, Hayek SS, Aepfelbacher M, Scaturro P, Puelles VG, Huber TB. Molecular consequences of SARS-CoV-2 liver tropism. Nat Metab 2022; 4:310-319. [PMID: 35347318 PMCID: PMC8964418 DOI: 10.1038/s42255-022-00552-6] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 02/14/2022] [Indexed: 02/07/2023]
Abstract
Extrapulmonary manifestations of COVID-19 have gained attention due to their links to clinical outcomes and their potential long-term sequelae1. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) displays tropism towards several organs, including the heart and kidney. Whether it also directly affects the liver has been debated2,3. Here we provide clinical, histopathological, molecular and bioinformatic evidence for the hepatic tropism of SARS-CoV-2. We find that liver injury, indicated by a high frequency of abnormal liver function tests, is a common clinical feature of COVID-19 in two independent cohorts of patients with COVID-19 requiring hospitalization. Using autopsy samples obtained from a third patient cohort, we provide multiple levels of evidence for SARS-CoV-2 liver tropism, including viral RNA detection in 69% of autopsy liver specimens, and successful isolation of infectious SARS-CoV-2 from liver tissue postmortem. Furthermore, we identify transcription-, proteomic- and transcription factor-based activity profiles in hepatic autopsy samples, revealing similarities to the signatures associated with multiple other viral infections of the human liver. Together, we provide a comprehensive multimodal analysis of SARS-CoV-2 liver tropism, which increases our understanding of the molecular consequences of severe COVID-19 and could be useful for the identification of organ-specific pharmacological targets.
Collapse
Affiliation(s)
- Nicola Wanner
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Pau Badia-I-Mompel
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, BioQuant, Heidelberg, Germany
| | - Carolin Edler
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Susanne Pfefferle
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maja T Lindenmeyer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Jan Czogalla
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Milagros N Wong
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yusuke Okabayashi
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fabian Braun
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marc Lütgehetmann
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elisabeth Meister
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Shun Lu
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maria L M Noriega
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Günther
- Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Adam Grundhoff
- Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Nicole Fischer
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hanna Bräuninger
- Department of Cardiology, University Heart and Vascular Centre Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Diana Lindner
- Department of Cardiology, University Heart and Vascular Centre Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Dirk Westermann
- Department of Cardiology, University Heart and Vascular Centre Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Fabian Haas
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kevin Roedl
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Kluge
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marylyn M Addo
- I. Department of Medicine, University Medical Hospital Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research, Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Samuel Huber
- I. Department of Medicine, University Medical Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Ansgar W Lohse
- I. Department of Medicine, University Medical Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Benjamin Ondruschka
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan P Sperhake
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julio Saez-Rodriguez
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, BioQuant, Heidelberg, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium and German Cancer Research Center, Partner Site Freiburg, Freiburg, Germany
| | - Salim S Hayek
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Martin Aepfelbacher
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Pietro Scaturro
- Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Victor G Puelles
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
45
|
Tombuloglu H, Sabit H, Al-Khallaf H, Kabanja JH, Alsaeed M, Al-Saleh N, Al-Suhaimi E. Multiplex real-time RT-PCR method for the diagnosis of SARS-CoV-2 by targeting viral N, RdRP and human RP genes. Sci Rep 2022; 12:2853. [PMID: 35181721 PMCID: PMC8857243 DOI: 10.1038/s41598-022-06977-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 02/03/2022] [Indexed: 12/13/2022] Open
Abstract
Corona Virus Disease 2019 (COVID-19) is a disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This pandemic has brought the world to a standstill and threatened human lives. Many methods are known to date to detect this virus. Due to their relative sensitivity, polymerase chain reaction (PCR)-based assays are the most frequently applied and considered the gold standard. However, due to the rapid mutation rate of the viral genome and the emergence of new variants, existing protocols need to be updated and improved. Designing a fast and accurate PCR-based assay is of great importance for the early detection of this virus and more efficient control of the spread of this disease. This study describes a fast, reliable, easy-to-use, and high-throughput multiplex SARS-CoV-2 RT-PCR detection method. The assay was designed to detect two viral genes (N and RdRP) and a human gene (RP) simultaneously. The performance and the sensitivity of the assay were tested in 28 SARS-CoV-2 positive samples and compared with commercial kits, which showed 100% positive percent agreement with a limit of detection (LOD) value of 1.40 and 0.81 copies/µL or 35.13 and 20.31 copies/reaction for RdRP and N genes, respectively. The current assay is found accurate, reliable, simple, sensitive, and specific. It can be used as an optimized SARS-CoV-2 diagnostic assay in hospitals, medical centers, and diagnostic laboratories as well as for research purposes.
Collapse
Affiliation(s)
- Huseyin Tombuloglu
- Department of Genetics Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia.
| | - Hussein Sabit
- Department of Genetics Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| | - Hamoud Al-Khallaf
- Department of Pathology and Laboratory Medicine, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Juma H Kabanja
- Department of Pathology and Laboratory Medicine, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Moneerah Alsaeed
- Department of Genetics Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| | - Najat Al-Saleh
- Department of Family and Community Medicine, King Fahad Hospital of the University, Imam Abdulrahman Bin Faisal University, Dammam, 31441, Saudi Arabia
| | - Ebtesam Al-Suhaimi
- Department of Biology, College of Science and Institute of Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, 31441, Saudi Arabia
| |
Collapse
|
46
|
Levine-Tiefenbrun M, Yelin I, Uriel H, Kuint J, Schreiber L, Herzel E, Katz R, Ben-Tov A, Gazit S, Patalon T, Chodick G, Kishony R. SARS-CoV-2 RT-qPCR Test Detection Rates Are Associated with Patient Age, Sex, and Time since Diagnosis. J Mol Diagn 2022; 24:112-119. [PMID: 34826637 PMCID: PMC8608683 DOI: 10.1016/j.jmoldx.2021.10.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/16/2021] [Accepted: 10/04/2021] [Indexed: 02/02/2023] Open
Abstract
Quantifying the detection rate of the widely used quantitative RT-PCR (RT-qPCR) test for severe acute respiratory syndrome coronavirus 2 and its dependence on patient demographic characteristics and disease progression is key in designing epidemiologic strategies. Analyzing 843,917 test results of 521,696 patients, a "positive period" was defined for each patient between diagnosis of coronavirus disease 2019 and the last positive test result. The fraction of positive test results within this period was then used to estimate detection rate. Regression analyses were used to determine associations of detection with time of sampling after diagnosis, patient demographic characteristics, and viral RNA copy number based on RT-qPCR cycle threshold values of the next positive test result. The overall detection rate in tests performed within 14 days after diagnosis was 83.1%. This rate was higher at days 0 to 5 after diagnosis (89.3%). Furthermore, detection rate was strongly associated with age and sex. Finally, the detection rate with the Allplex 2019-nCoV RT-qPCR kit was associated, at the single-patient level, with viral RNA copy number (P < 10-9). These results show that the reliability of the test result is reduced in later days as well as for women and younger patients, in whom the viral loads are typically lower.
Collapse
Affiliation(s)
| | - Idan Yelin
- Biology Faculty, Technion-Israel Institute of Technology, Haifa, Israel.
| | - Hedva Uriel
- Faculty of Computer Science, Technion-Israel Institute of Technology, Haifa, Israel
| | - Jacob Kuint
- Maccabitech, Maccabi Health Services, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | - Esma Herzel
- Maccabitech, Maccabi Health Services, Tel Aviv, Israel
| | - Rachel Katz
- Maccabitech, Maccabi Health Services, Tel Aviv, Israel
| | - Amir Ben-Tov
- Maccabitech, Maccabi Health Services, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Sivan Gazit
- Maccabitech, Maccabi Health Services, Tel Aviv, Israel
| | - Tal Patalon
- Maccabitech, Maccabi Health Services, Tel Aviv, Israel
| | - Gabriel Chodick
- Maccabitech, Maccabi Health Services, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Roy Kishony
- Biology Faculty, Technion-Israel Institute of Technology, Haifa, Israel; Faculty of Computer Science, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
47
|
Development of the DNA-based biosensors for high performance in detection of molecular biomarkers: More rapid, sensitive, and universal. Biosens Bioelectron 2022; 197:113739. [PMID: 34781175 PMCID: PMC8553638 DOI: 10.1016/j.bios.2021.113739] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/25/2021] [Indexed: 02/07/2023]
Abstract
The molecular biomarkers are molecules that are closely related to specific physiological states. Numerous molecular biomarkers have been identified as targets for disease diagnosis and biological research. To date, developing highly efficient probes for the precise detection of biomarkers has become an attractive research field which is very important for biological and biochemical studies. During the past decades, not only the small chemical probe molecules but also the biomacromolecules such as enzymes, antibodies, and nucleic acids have been introduced to construct of biosensor platform to achieve the detection of biomarkers in a highly specific and highly efficient way. Nevertheless, improving the performance of the biosensors, especially in clinical applications, is still in urgent demand in this field. A noteworthy example is the Corona Virus Disease 2019 (COVID-19) that breaks out globally in a short time in 2020. The COVID-19 was caused by the virus called SARS-CoV-2. Early diagnosis is very important to block the infection of the virus. Therefore, during these months scientists have developed dozens of methods to achieve rapid and sensitive detection of the virus. Nowadays some of these new methods have been applied for producing the commercial detection kit and help people against the disease worldwide. DNA-based biosensors are useful tools that have been widely applied in the detection of molecular biomarkers. The good stability, high specificity, and excellent biocompatibility make the DNA-based biosensors versatile in application both in vitro and in vivo. In this paper, we will review the major methods that emerged in recent years on the design of DNA-based biosensors and their applications. Moreover, we will also briefly discuss the possible future direction of DNA-based biosensors design. We believe this is helpful for people interested in not only the biosensor field but also in the field of analytical chemistry, DNA nanotechnology, biology, and disease diagnosis.
Collapse
|
48
|
Vierbaum L, Wojtalewicz N, Grunert HP, Lindig V, Duehring U, Drosten C, Corman V, Niemeyer D, Ciesek S, Rabenau HF, Berger A, Obermeier M, Nitsche A, Michel J, Mielke M, Huggett J, O’Sullivan D, Busby E, Cowen S, Vallone PM, Cleveland MH, Falak S, Kummrow A, Keller T, Schellenberg I, Zeichhardt H, Kammel M. RNA reference materials with defined viral RNA loads of SARS-CoV-2-A useful tool towards a better PCR assay harmonization. PLoS One 2022; 17:e0262656. [PMID: 35051208 PMCID: PMC8775330 DOI: 10.1371/journal.pone.0262656] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 01/03/2022] [Indexed: 12/19/2022] Open
Abstract
SARS-CoV-2, the cause of COVID-19, requires reliable diagnostic methods to track the circulation of this virus. Following the development of RT-qPCR methods to meet this diagnostic need in January 2020, it became clear from interlaboratory studies that the reported Ct values obtained for the different laboratories showed high variability. Despite this the Ct values were explored as a quantitative cut off to aid clinical decisions based on viral load. Consequently, there was a need to introduce standards to support estimation of SARS-CoV-2 viral load in diagnostic specimens. In a collaborative study, INSTAND established two reference materials (RMs) containing heat-inactivated SARS-CoV-2 with SARS-CoV-2 RNA loads of ~107 copies/mL (RM 1) and ~106 copies/mL (RM 2), respectively. Quantification was performed by RT-qPCR using synthetic SARS-CoV-2 RNA standards and digital PCR. Between November 2020 and February 2021, German laboratories were invited to use the two RMs to anchor their Ct values measured in routine diagnostic specimens, with the Ct values of the two RMs. A total of 305 laboratories in Germany were supplied with RM 1 and RM 2. The laboratories were requested to report their measured Ct values together with details on the PCR method they used to INSTAND. This resultant 1,109 data sets were differentiated by test system and targeted gene region. Our findings demonstrate that an indispensable prerequisite for linking Ct values to SARS-CoV-2 viral loads is that they are treated as being unique to an individual laboratory. For this reason, clinical guidance based on viral loads should not cite Ct values. The RMs described were a suitable tool to determine the specific laboratory Ct for a given viral load. Furthermore, as Ct values can also vary between runs when using the same instrument, such RMs could be used as run controls to ensure reproducibility of the quantitative measurements.
Collapse
Affiliation(s)
- Laura Vierbaum
- INSTAND e.V., Society for Promoting Quality Assurance in Medical Laboratories, Duesseldorf, North Rhine-Westphalia, Germany
| | - Nathalie Wojtalewicz
- INSTAND e.V., Society for Promoting Quality Assurance in Medical Laboratories, Duesseldorf, North Rhine-Westphalia, Germany
| | | | - Vanessa Lindig
- IQVD GmbH, Institut fuer Qualitaetssicherung in der Virusdiagnostik, Berlin, Germany
| | - Ulf Duehring
- GBD Gesellschaft fuer Biotechnologische Diagnostik mbH, Berlin, Germany
| | - Christian Drosten
- Institute of Virology, Charité - University Medicine Berlin; National Consultant Laboratory for Coronaviruses; German Centre for Infection Research, Berlin, Germany
| | - Victor Corman
- Institute of Virology, Charité - University Medicine Berlin; National Consultant Laboratory for Coronaviruses; German Centre for Infection Research, Berlin, Germany
| | - Daniela Niemeyer
- Institute of Virology, Charité - University Medicine Berlin; National Consultant Laboratory for Coronaviruses; German Centre for Infection Research, Berlin, Germany
| | - Sandra Ciesek
- Institute for Medical Virology, University Hospital, Goethe University Frankfurt, Frankfurt, Hesse, Germany
- German Centre for Infection Research, External partner site Frankfurt, Hesse, Germany
- Fraunhofer Institute for Molecular Biology and Applied Ecology, Branch Translational Medicine and Pharmacology, Frankfurt, Hesse, Germany
| | - Holger F. Rabenau
- Institute for Medical Virology, University Hospital, Goethe University Frankfurt, Frankfurt, Hesse, Germany
| | - Annemarie Berger
- Institute for Medical Virology, University Hospital, Goethe University Frankfurt, Frankfurt, Hesse, Germany
| | | | - Andreas Nitsche
- Robert Koch-Institute, Centre for Biological Threats and Special Pathogens, Berlin, Germany
| | - Janine Michel
- Robert Koch-Institute, Centre for Biological Threats and Special Pathogens, Berlin, Germany
| | - Martin Mielke
- Robert Koch-Institute, Department for Infectious Diseases, Berlin, Germany
| | - Jim Huggett
- National Measurement Laboratory, LGC, Teddington, Middlesex, United Kingdom
- Faculty of Health & Medical Science, School of Biosciences & Medicine, University of Surrey, Guildford, United Kingdom
| | - Denise O’Sullivan
- National Measurement Laboratory, LGC, Teddington, Middlesex, United Kingdom
| | - Eloise Busby
- National Measurement Laboratory, LGC, Teddington, Middlesex, United Kingdom
| | - Simon Cowen
- National Measurement Laboratory, LGC, Teddington, Middlesex, United Kingdom
| | - Peter M. Vallone
- Materials Measurement Laboratory, Biomolecular Measurement Division, NIST, National Institute of Standards and Technology, Applied Genetics Group, Gaithersburg, Massachusetts, United States of America
| | - Megan H. Cleveland
- Materials Measurement Laboratory, Biomolecular Measurement Division, NIST, National Institute of Standards and Technology, Applied Genetics Group, Gaithersburg, Massachusetts, United States of America
| | - Samreen Falak
- Physikalisch-Technische Bundesanstalt, Berlin, Germany
| | | | | | - Ingo Schellenberg
- INSTAND e.V., Society for Promoting Quality Assurance in Medical Laboratories, Duesseldorf, North Rhine-Westphalia, Germany
- Institute of Bioanalytical Sciences, Center of Life Sciences, Anhalt University of Applied Sciences, Bernburg, Saxony-Anhalt, Germany
| | - Heinz Zeichhardt
- INSTAND e.V., Society for Promoting Quality Assurance in Medical Laboratories, Duesseldorf, North Rhine-Westphalia, Germany
- GBD Gesellschaft fuer Biotechnologische Diagnostik mbH, Berlin, Germany
- IQVD GmbH, Institut fuer Qualitaetssicherung in der Virusdiagnostik, Berlin, Germany
| | - Martin Kammel
- INSTAND e.V., Society for Promoting Quality Assurance in Medical Laboratories, Duesseldorf, North Rhine-Westphalia, Germany
- IQVD GmbH, Institut fuer Qualitaetssicherung in der Virusdiagnostik, Berlin, Germany
| |
Collapse
|
49
|
Drobysh M, Ramanaviciene A, Viter R, Chen CF, Samukaite-Bubniene U, Ratautaite V, Ramanavicius A. Biosensors for the Determination of SARS-CoV-2 Virus and Diagnosis of COVID-19 Infection. Int J Mol Sci 2022; 23:666. [PMID: 35054850 PMCID: PMC8776074 DOI: 10.3390/ijms23020666] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/29/2021] [Accepted: 01/03/2022] [Indexed: 02/04/2023] Open
Abstract
Monitoring and tracking infection is required in order to reduce the spread of the coronavirus disease 2019 (COVID-19), induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). To achieve this goal, the development and deployment of quick, accurate, and sensitive diagnostic methods are necessary. The determination of the SARS-CoV-2 virus is performed by biosensing devices, which vary according to detection methods and the biomarkers which are inducing/providing an analytical signal. RNA hybridisation, antigen-antibody affinity interaction, and a variety of other biological reactions are commonly used to generate analytical signals that can be precisely detected using electrochemical, electrochemiluminescence, optical, and other methodologies and transducers. Electrochemical biosensors, in particular, correspond to the current trend of bioanalytical process acceleration and simplification. Immunosensors are based on the determination of antigen-antibody interaction, which on some occasions can be determined in a label-free mode with sufficient sensitivity.
Collapse
Affiliation(s)
- Maryia Drobysh
- State Research Institute Center for Physical and Technological Sciences, Sauletekio Ave. 3, LT-10257 Vilnius, Lithuania; (M.D.); (U.S.-B.); (V.R.)
- NanoTechnas—Center of Nanotechnology and Materials Science, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, LT-03225 Vilnius, Lithuania;
| | - Almira Ramanaviciene
- NanoTechnas—Center of Nanotechnology and Materials Science, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, LT-03225 Vilnius, Lithuania;
| | - Roman Viter
- Center for Collective Use of Scientific Equipment, Sumy State University, Sanatornaya Str. 31, 40018 Sumy, Ukraine
- Institute of Atomic Physics and Spectroscopy, University of Latvia, Jelgavas Street 3, LV-1004 Riga, Latvia
| | - Chien-Fu Chen
- Institute of Applied Mechanics, National Taiwan University 1, Sec. 4, Roosevelt Rd., Da’an Dist., Taipei 106, Taiwan;
| | - Urte Samukaite-Bubniene
- State Research Institute Center for Physical and Technological Sciences, Sauletekio Ave. 3, LT-10257 Vilnius, Lithuania; (M.D.); (U.S.-B.); (V.R.)
- NanoTechnas—Center of Nanotechnology and Materials Science, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, LT-03225 Vilnius, Lithuania;
| | - Vilma Ratautaite
- State Research Institute Center for Physical and Technological Sciences, Sauletekio Ave. 3, LT-10257 Vilnius, Lithuania; (M.D.); (U.S.-B.); (V.R.)
- NanoTechnas—Center of Nanotechnology and Materials Science, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, LT-03225 Vilnius, Lithuania;
| | - Arunas Ramanavicius
- State Research Institute Center for Physical and Technological Sciences, Sauletekio Ave. 3, LT-10257 Vilnius, Lithuania; (M.D.); (U.S.-B.); (V.R.)
- NanoTechnas—Center of Nanotechnology and Materials Science, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, LT-03225 Vilnius, Lithuania;
| |
Collapse
|
50
|
Brandner JM, Boor P, Borcherding L, Edler C, Gerber S, Heinemann A, Hilsenbeck J, Kasajima A, Lohner L, Märkl B, Pablik J, Schröder AS, Slotta-Huspenina J, Sommer L, Sperhake JP, von Stillfried S, Dintner S. Contamination of personal protective equipment during COVID-19 autopsies. Virchows Arch 2022; 480:519-528. [PMID: 34993593 PMCID: PMC8735722 DOI: 10.1007/s00428-021-03263-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/13/2021] [Accepted: 12/27/2021] [Indexed: 12/11/2022]
Abstract
Confronted with an emerging infectious disease at the beginning of the COVID-19 pandemic, the medical community faced concerns regarding the safety of autopsies on those who died of the disease. This attitude has changed, and autopsies are now recognized as indispensable tools for understanding COVID-19, but the true risk of infection to autopsy staff is nevertheless still debated. To clarify the rate of SARS-CoV-2 contamination in personal protective equipment (PPE), swabs were taken at nine points in the PPE of one physician and one assistant after each of 11 full autopsies performed at four centers. Swabs were also obtained from three minimally invasive autopsies (MIAs) conducted at a fifth center. Lung/bronchus swabs of the deceased served as positive controls, and SARS-CoV-2 RNA was detected by real-time RT-PCR. In 9 of 11 full autopsies, PPE samples tested RNA positive through PCR, accounting for 41 of the 198 PPE samples taken (21%). The main contaminated items of the PPE were gloves (64% positive), aprons (50% positive), and the tops of shoes (36% positive) while the fronts of safety goggles, for example, were positive in only 4.5% of the samples, and all the face masks were negative. In MIAs, viral RNA was observed in one sample from a glove but not in other swabs. Infectious virus isolation in cell culture was performed on RNA-positive swabs from the full autopsies. Of all the RNA-positive PPE samples, 21% of the glove samples, taken in 3 of 11 full autopsies, tested positive for infectious virus. In conclusion, PPE was contaminated with viral RNA in 82% of autopsies. In 27% of autopsies, PPE was found to be contaminated even with infectious virus, representing a potential risk of infection to autopsy staff. Adequate PPE and hygiene measures, including appropriate waste deposition, are therefore essential to ensure a safe work environment.
Collapse
Affiliation(s)
- Johanna M Brandner
- Business Division of Safety, Security, and Compliance, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Dermatology and Venerology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DEFEAT PANDEMIcs Working Group, Hamburg, Germany
| | - Peter Boor
- Institute of Pathology, Rheinisch Westfaelische Technische Hochschule, Aachen University Hospital, Aachen, Germany
| | - Lukas Borcherding
- General Pathology and Molecular Diagnostics, Medical Faculty, University of Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany
| | - Carolin Edler
- DEFEAT PANDEMIcs Working Group, Hamburg, Germany.,Department of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sven Gerber
- Business Division of Safety, Security, and Compliance, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DEFEAT PANDEMIcs Working Group, Hamburg, Germany
| | - Axel Heinemann
- DEFEAT PANDEMIcs Working Group, Hamburg, Germany.,Department of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julia Hilsenbeck
- DEFEAT PANDEMIcs Working Group, Hamburg, Germany.,Institute of Pathology, University Hospital Carl Gustav Carus Dresden, Technical University of Dresden, Dresden, Germany
| | - Atsuko Kasajima
- Institute of Pathology, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Larissa Lohner
- DEFEAT PANDEMIcs Working Group, Hamburg, Germany.,Department of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bruno Märkl
- DEFEAT PANDEMIcs Working Group, Hamburg, Germany. .,General Pathology and Molecular Diagnostics, Medical Faculty, University of Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany.
| | - Jessica Pablik
- DEFEAT PANDEMIcs Working Group, Hamburg, Germany.,Institute of Pathology, University Hospital Carl Gustav Carus Dresden, Technical University of Dresden, Dresden, Germany
| | - Ann Sophie Schröder
- DEFEAT PANDEMIcs Working Group, Hamburg, Germany.,Department of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julia Slotta-Huspenina
- DEFEAT PANDEMIcs Working Group, Hamburg, Germany.,Institute of Pathology, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Linna Sommer
- DEFEAT PANDEMIcs Working Group, Hamburg, Germany.,Institute of Pathology, University Hospital Carl Gustav Carus Dresden, Technical University of Dresden, Dresden, Germany
| | - Jan-Peter Sperhake
- DEFEAT PANDEMIcs Working Group, Hamburg, Germany.,Department of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Saskia von Stillfried
- DEFEAT PANDEMIcs Working Group, Hamburg, Germany.,Institute of Pathology, Rheinisch Westfaelische Technische Hochschule, Aachen University Hospital, Aachen, Germany
| | - Sebastian Dintner
- General Pathology and Molecular Diagnostics, Medical Faculty, University of Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany
| |
Collapse
|