1
|
Wu H, Fujioka Y, Sakaguchi S, Suzuki Y, Nakano T. Morphological analysis for two types of viral particles in vacuoles of SARS-CoV-2-infected cells. Med Mol Morphol 2024; 57:124-135. [PMID: 38393367 DOI: 10.1007/s00795-024-00381-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/17/2024] [Indexed: 02/25/2024]
Abstract
In this study, we analyzed the morphological structure of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in human cells. We identified the two types of viral particles present within the vacuoles of infected cells. Using transmission electron microscopy, we observed that SARS-CoV-2 particles exhibited both low- and high-electron-density structures, which was further confirmed through three-dimensional reconstruction using electron tomography. The budding of these particles was exclusively observed within these vacuoles. Intriguingly, viral particles with low-electron-density structures were confined to vacuoles, whereas those with high-electron-density structures were found in vacuoles and on the cell membrane surface of infected cells. Notably, high-electron-density particles found within vacuoles exhibited the same morphology as those outside the infected cells. This observation suggests that the two types of viral particles identified in this study had different maturation status. Our findings provide valuable insights into the molecular details of SARS-CoV-2 particles, contributing to our understanding of the virus.
Collapse
Affiliation(s)
- Hong Wu
- Department of Microbiology and Infection Control, Faculty of Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki, Osaka, 569-8686, Japan.
| | - Yoshihiko Fujioka
- Department of Microbiology and Infection Control, Faculty of Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki, Osaka, 569-8686, Japan
| | - Shoichi Sakaguchi
- Department of Microbiology and Infection Control, Faculty of Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki, Osaka, 569-8686, Japan
| | - Youichi Suzuki
- Department of Microbiology and Infection Control, Faculty of Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki, Osaka, 569-8686, Japan
| | - Takashi Nakano
- Department of Microbiology and Infection Control, Faculty of Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki, Osaka, 569-8686, Japan
| |
Collapse
|
2
|
Balabdaoui F, Wierzbicki T, Bao E. Reconstruction of the real 3D shape of the SARS-CoV-2 virus. Biophys J 2024; 123:1297-1310. [PMID: 38715359 PMCID: PMC11140469 DOI: 10.1016/j.bpj.2024.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 02/23/2024] [Accepted: 04/19/2024] [Indexed: 05/19/2024] Open
Abstract
The photographs of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus taken by electron transmission microscopy and cryoelectron microscopy provide only a 2D silhouette. The viruses appear to look like distorted circles. The present paper questions the real shape of the SARS-CoV-2 virus and makes an attempt to give an answer. Is this a general ellipsoid, a spheroid with rotational symmetry, a sphere, or something else? The answer requires the application of tools from three different disciplines: structural mechanics, microbiology, and statistics. A total of 590 virus photographs taken from 22 recently published papers were examined. From this experimental data pool, the histogram of diameter ratios was built from the 283 measurements where the virus images could be approximated as ellipses. The curve peaks at the diameter ratio of 1.22. The transformation equation for the spatial shape to the planar shade was derived for a fixed light source of the microscope. This equation involves an unknown orientation of the viruses with respect to the microscope. Two sets of models were developed, one with a uniform distribution of the virus orientation and the other with the orientation defined by the normalized beta distribution. In both sets of models, the unknown diameter ratio of the spheroidal virus was regarded as a random realization from translated gamma distributions. The parameters of the distribution of the kernel functions were determined by minimizing the mean square difference between the predicted and measured 2D histograms. The information included in the measured histograms was found to be insufficient to find an unknown distribution of the virus's orientation. Simply too many unknown parameters render the solution physically unrealistic. The minimization procedure with a uniform probability of virus orientation predicted the peak of the aspect ratio of the 3D spheroid at 1.32. Based on this result, models of the virus will be developed in the continuation of this research for a full dynamic analysis.
Collapse
Affiliation(s)
| | | | - Emma Bao
- Duke University, Durham, North Carolina
| |
Collapse
|
3
|
Oda JM, den Hartigh AB, Jackson SM, Tronco AR, Fink SL. The unfolded protein response components IRE1α and XBP1 promote human coronavirus infection. mBio 2023; 14:e0054023. [PMID: 37306512 PMCID: PMC10470493 DOI: 10.1128/mbio.00540-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 04/20/2023] [Indexed: 06/13/2023] Open
Abstract
The cellular processes that support human coronavirus replication and contribute to the pathogenesis of severe disease remain incompletely understood. Many viruses, including coronaviruses, cause endoplasmic reticulum (ER) stress during infection. IRE1α is a component of the cellular response to ER stress that initiates non-conventional splicing of XBP1 mRNA. Spliced XBP1 encodes a transcription factor that induces the expression of ER-related targets. Activation of the IRE1α-XBP1 pathway occurs in association with risk factors for severe human coronavirus infection. In this study, we found that the human coronaviruses HCoV-OC43 (human coronavirus OC43) and SARS-CoV-2 (severe acute respiratory syndrome coronavirus-2) both robustly activate the IRE1α-XBP1 branch of the unfolded protein response in cultured cells. Using IRE1α nuclease inhibitors and genetic knockdown of IRE1α and XBP1, we found that these host factors are required for optimal replication of both viruses. Our data suggest that IRE1α supports infection downstream of initial viral attachment and entry. In addition, we found that ER stress-inducing conditions are sufficient to enhance human coronavirus replication. Furthermore, we found markedly increased XBP1 in circulation in human patients with severe coronavirus disease 2019 (COVID-19). Together, these results demonstrate the importance of IRE1α and XBP1 for human coronavirus infection. IMPORTANCE There is a critical need to understand the cellular processes co-opted during human coronavirus replication, with an emphasis on identifying mechanisms underlying severe disease and potential therapeutic targets. Here, we demonstrate that the host proteins IRE1α and XBP1 are required for robust infection by the human coronaviruses, SARS-CoV-2 and HCoV-OC43. IRE1α and XBP1 participate in the cellular response to ER stress and are activated during conditions that predispose to severe COVID-19. We found enhanced viral replication with exogenous IRE1α activation, and evidence that this pathway is activated in humans during severe COVID-19. Together, these results demonstrate the importance of IRE1α and XBP1 for human coronavirus infection.
Collapse
Affiliation(s)
- Jessica M. Oda
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Andreas B. den Hartigh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Shoen M. Jackson
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Ana R. Tronco
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Susan L. Fink
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
4
|
McCormack CP, Yan AWC, Brown JC, Sukhova K, Peacock TP, Barclay WS, Dorigatti I. Modelling the viral dynamics of the SARS-CoV-2 Delta and Omicron variants in different cell types. J R Soc Interface 2023; 20:20230187. [PMID: 37553993 PMCID: PMC10410224 DOI: 10.1098/rsif.2023.0187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 07/18/2023] [Indexed: 08/10/2023] Open
Abstract
We use viral kinetic models fitted to viral load data from in vitro studies to explain why the SARS-CoV-2 Omicron variant replicates faster than the Delta variant in nasal cells, but slower than Delta in lung cells, which could explain Omicron's higher transmission potential and lower severity. We find that in both nasal and lung cells, viral infectivity is higher for Omicron but the virus production rate is higher for Delta, with an estimated approximately 200-fold increase in infectivity and 100-fold decrease in virus production when comparing Omicron with Delta in nasal cells. However, the differences are unequal between cell types, and ultimately lead to the basic reproduction number and growth rate being higher for Omicron in nasal cells, and higher for Delta in lung cells. In nasal cells, Omicron alone can enter via a TMPRSS2-independent pathway, but it is primarily increased efficiency of TMPRSS2-dependent entry which accounts for Omicron's increased activity. This work paves the way for using within-host mathematical models to understand the transmission potential and severity of future variants.
Collapse
Affiliation(s)
- Clare P. McCormack
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, UK
| | - Ada W. C. Yan
- Department of Infectious Disease, Imperial College London, London, UK
| | - Jonathan C. Brown
- Department of Infectious Disease, Imperial College London, London, UK
| | - Ksenia Sukhova
- Department of Infectious Disease, Imperial College London, London, UK
| | - Thomas P. Peacock
- Department of Infectious Disease, Imperial College London, London, UK
| | - Wendy S. Barclay
- Department of Infectious Disease, Imperial College London, London, UK
| | - Ilaria Dorigatti
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, UK
| |
Collapse
|
5
|
Hussain F, Mazumder N, Roy S. Possibilities of simulation of coronavirus SARS-CoV-2 by using light scattering approach. Lasers Med Sci 2023; 38:107. [PMID: 37076645 PMCID: PMC10115601 DOI: 10.1007/s10103-023-03768-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/04/2023] [Indexed: 04/21/2023]
Abstract
Issues related to human coronavirus (SARS CoV-2) are a burning topic of research in present times. Due to its easily contagious nature, real experimentation under laboratory conditions requires a high level of biosafety. A powerful algorithm serves as a potential tool for the analysis of these particles. We attempted to simulate the light scattering from coronavirus (SARS CoV-2) model. Different images were modelled using a modified version of a Monte Carlo code. The results indicate that spikes on the viruses exhibit a significant scattering profile and that the presence of spikes during modelling contributes to the distinctiveness of the scattering profiles.
Collapse
Affiliation(s)
- Farhana Hussain
- Department of Physics, School of Applied Sciences, University of Science & Technology, Meghalaya, District- Ri Bhoi, Baridua, Meghalaya, 793101, India
| | - Nirmal Mazumder
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Sanchita Roy
- Department of Physics, Royal School of Applied and Pure Sciences, Royal Global University, Guwahati, Assam, 781035, India.
| |
Collapse
|
6
|
Abdaljaleel M, Tawalbeh I, Sallam M, Hani AB, Al-Abdallat IM, Omari BA, Al-Mustafa S, Abder-Rahman H, Abbas AS, Zureigat M, Al-Abbadi MA. Postmortem lung and heart examination of COVID-19 patients in a case series from Jordan. J Pathol Transl Med 2023; 57:102-112. [PMID: 36950812 PMCID: PMC10028009 DOI: 10.4132/jptm.2023.01.30] [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: 11/27/2022] [Accepted: 01/30/2023] [Indexed: 03/24/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) has emerged as a pandemic for more than 2 years. Autopsy examination is an invaluable tool to understand the pathogenesis of emerging infections and their consequent mortalities. The aim of the current study was to present the lung and heart pathological findings of COVID-19-positive autopsies performed in Jordan. METHODS The study involved medicolegal cases, where the cause of death was unclear and autopsy examination was mandated by law. We included the clinical and pathologic findings of routine gross and microscopic examination of cases that were positive for COVID-19 at time of death. Testing for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was confirmed through molecular detection by real-time polymerase chain reaction, serologic testing for IgM and electron microscope examination of lung samples. RESULTS Seventeen autopsies were included, with male predominance (76.5%), Jordanians (70.6%), and 50 years as the mean age at time of death. Nine out of 16 cases (56.3%) had co-morbidities, with one case lacking such data. Histologic examination of lung tissue revealed diffuse alveolar damage in 13/17 cases (76.5%), and pulmonary microthrombi in 8/17 cases (47.1%). Microscopic cardiac findings were scarcely detected. Two patients died as a direct result of acute cardiac disease with limited pulmonary findings. CONCLUSIONS The detection of SARS-CoV-2 in postmortem examination can be an incidental or contributory finding which highlights the value of autopsy examination to determine the exact cause of death in controversial cases.
Collapse
Affiliation(s)
- Maram Abdaljaleel
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
- Department of Clinical Laboratories and Forensic Medicine, Jordan University Hospital, Amman, Jordan
| | - Isra Tawalbeh
- Department of Forensic Pathology, Ministry of Health, Amman, Jordan
| | - Malik Sallam
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
- Department of Clinical Laboratories and Forensic Medicine, Jordan University Hospital, Amman, Jordan
- Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Amjad Bani Hani
- Department of General Surgery, School of Medicine, The University of Jordan, Amman, Jordan
| | - Imad M Al-Abdallat
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
- Department of Clinical Laboratories and Forensic Medicine, Jordan University Hospital, Amman, Jordan
| | - Baheth Al Omari
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
- Department of Clinical Laboratories and Forensic Medicine, Jordan University Hospital, Amman, Jordan
| | - Sahar Al-Mustafa
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
- Department of Clinical Laboratories and Forensic Medicine, Jordan University Hospital, Amman, Jordan
| | - Hasan Abder-Rahman
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
- Department of Clinical Laboratories and Forensic Medicine, Jordan University Hospital, Amman, Jordan
| | - Adnan Said Abbas
- Department of Forensic Pathology, Ministry of Health, Amman, Jordan
| | - Mahmoud Zureigat
- Department of Forensic Pathology, Ministry of Health, Amman, Jordan
| | - Mousa A Al-Abbadi
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
- Department of Clinical Laboratories and Forensic Medicine, Jordan University Hospital, Amman, Jordan
| |
Collapse
|
7
|
Yin L, Li Y, Zhang W, Han X, Wu Q, Xie Y, Fan J, Ma L. Detection Methods for Foodborne Viruses: Current State-of-Art and Future Perspectives. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:3551-3563. [PMID: 36657010 DOI: 10.1021/acs.jafc.2c06537] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Foodborne viruses have been recognized as important threats to food safety and human health. Rapid and accurate detection is one of the most crucial measures for food safety control. With the development of biology, chemistry, nanoscience, and related interdisciplines, detection strategies have been devised and advanced continuously. This review mainly focuses on the progress of detection methods for foodborne viruses. The current detection methods for foodborne viruses are summarized, including traditional electron microscopy and cultural isolation, immunoassay, molecular technology, biosensors, and newly emerging CRISPR/Cas-based detection technology. Furthermore, a comparison of the detection methods was objectively discussed. This review provides a comprehensive account of foodborne virus detection methods from fundamentals to state-of-the-art and illustrates the advantages and disadvantages of the current methods and proposes the future trends and directions for foodborne virus detection. It is hoped that this review can update current knowledge and present blueprints in order to accelerate futuristic development.
Collapse
Affiliation(s)
- Lijuan Yin
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Yaru Li
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Wenlu Zhang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Xiao Han
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Qiankun Wu
- Academy of National Food and Strategic Reserves Administration, Beijing, 100037, China
| | - Yanyan Xie
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Jingjing Fan
- Beijing Kwinbon Biotechnology Co., Ltd, Beijing, 102200, China
| | - Long Ma
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| |
Collapse
|
8
|
Identifying Active Progeny Virus Particles in Formalin-Fixed, Paraffin-Embedded Sections Using Correlative Light and Scanning Electron Microscopy. J Transl Med 2023; 103:100020. [PMID: 36748195 DOI: 10.1016/j.labinv.2022.100020] [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: 04/07/2022] [Revised: 10/06/2022] [Accepted: 10/10/2022] [Indexed: 01/18/2023] Open
Abstract
Immunohistochemical analysis of formalin-fixed paraffin-embedded (FFPE) tissue blocks is routinely used to identify virus-infected cells. However, detecting virus particles in FFPE sections using light microscopy is difficult because of the light diffraction resolution limitations of an optical microscope. In this study, light microscopy and field emission scanning electron microscopy were performed to observe 3-dimensional virus particles in FFPE sections in a nondestructive manner using NanoSuit or osmium conductive treatment methods. The virus particles in FFPE sections were immunostained with specific antibodies against the surface antigens of the viral particles and stained with 3,3'-diaminobenzidine. A metal solution (0.2% gold chloride or 2% osmium tetroxide) was applied to enhance the 3,3'-diaminobenzidine-stained area. This procedure is nondestructive for FFPE sections and is a simpler method than transmission electron microscopy. To validate the applicability of this technique, we performed 3-dimensional imaging of the virus particles of different sizes, such as human papillomavirus, cytomegalovirus, and varicella-zoster virus. Furthermore, ultrathin sections from the FFPE sections that were observed to harbor viral particles using field emission scanning electron microscopy were prepared and assessed using transmission electron microscopy. In the correlative areas, transmission electron microscopy confirmed the presence of large numbers of virus particles. These results indicated that the combination of marking viral particles with 3,3'-diaminobenzidine/metal staining and conductive treatment can identify active progeny virus particles in FFPE sections using scanning electron microscopy. This easy correlative imaging of field emission scanning electron microscopy of the identical area of FFPE in light microscopy may help elucidate new pathological mechanisms of virus-related diseases.
Collapse
|
9
|
Bajrai LH, El-Kafrawy SA, Hassan AM, Tolah AM, Alnahas RS, Sohrab SS, Rehan M, Azhar EI. In vitro screening of anti-viral and virucidal effects against SARS-CoV-2 by Hypericum perforatum and Echinacea. Sci Rep 2022; 12:21723. [PMID: 36522420 PMCID: PMC9754313 DOI: 10.1038/s41598-022-26157-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Hypericum perforatum and Echinacea are reported to have antiviral activities against several viral infections. In this study, H. perforatum (St. John's Wort) and Echinacea were tested in vitro using Vero E6 cells for their anti-viral effects against the newly identified Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) through its infectious cycle from 0 to 48 h post infection. The hypericin of H. perforatum and the different parts (roots, seeds, aerial) of two types of Echinacea species (Echinacea purpurea and Echinacea angustifolia) were tested for their anti-viral activities to measure the inhibition of viral load using quantitative real-time polymerase chain reaction (qRT-PCR) on cell culture assay. Interestingly, the H. perforatum-Echinacea mixture (1:1 ratio) of H. perforatum and Echinacea was tested as well on SARS-CoV-2 and showed crucial anti-viral activity competing H. perforatum then Echinacea effects as anti-viral treatment. Therefore, the results H. perforatum and Echinacea species, applied in this study showed significant anti-viral and virucidal effects in the following order of potency: H. perforatum, H. perforatum-Echinacea mixture, and Echinacea on SARS-CoV-2 infectious cycle. Additionally, molecular simulation analysis of the compounds with essential proteins (Mpro and RdRp) of the SARS-CoV-2 revealed the most potent bioactive compounds such as Echinacin, Echinacoside, Cyanin, Cyanidin 3-(6''-alonylglucoside, Quercetin-3-O-glucuronide, Proanthocyanidins, Rutin, Kaempferol-3-O-rutinoside, and Quercetin-3-O-xyloside. Thus, based on the outcome of this study, it is demanding the setup of clinical trial with specific therapeutic protocol.
Collapse
Affiliation(s)
- Leena Hussein Bajrai
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.
- Biochemistry Department, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Sherif Ali El-Kafrawy
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmed Mohamed Hassan
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmed Majdi Tolah
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Rabig, Saudi Arabia
| | - Rabie Saleh Alnahas
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sayed Sartaj Sohrab
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohd Rehan
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Esam Ibraheem Azhar
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
10
|
Bergner T, Zech F, Hirschenberger M, Stenger S, Sparrer KMJ, Kirchhoff F, Read C. Near-Native Visualization of SARS-CoV-2 Induced Membrane Remodeling and Virion Morphogenesis. Viruses 2022; 14:v14122786. [PMID: 36560790 PMCID: PMC9784144 DOI: 10.3390/v14122786] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/02/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Infection with the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the causative agent of the COVID-19 pandemic, leads to profound remodeling of cellular membranes, promoting viral replication and virion assembly. A full understanding of this drastic remodeling and the process of virion morphogenesis remains lacking. In this study, we applied room temperature transmission electron microscopy (TEM) and scanning transmission electron microscopy (STEM) tomography to visualize the SARS-CoV-2 replication factory in Vero cells, and present our results in comparison with published cryo-EM studies. We obtained cryo-EM-like clarity of the ultrastructure by employing high-pressure freezing, freeze substitution (HPF-FS) and embedding, allowing room temperature visualization of double-membrane vesicles (DMVs) in a near-native state. In addition, our data illustrate the consecutive stages of virion morphogenesis and reveal that SARS-CoV-2 ribonucleoprotein assembly and membrane curvature occur simultaneously. Finally, we show the tethering of virions to the plasma membrane in 3D, and that accumulations of virus particles lacking spike protein in large vesicles are most likely not a result of defective virion assembly at their membrane. In conclusion, this study puts forward a room-temperature EM technique providing near-native ultrastructural information about SARS-CoV-2 replication, adding to our understanding of the interaction of this pandemic virus with its host cell.
Collapse
Affiliation(s)
- Tim Bergner
- Central Facility for Electron Microscopy, Ulm University, 89081 Ulm, Germany
| | - Fabian Zech
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Steffen Stenger
- Institute for Microbiology and Hygiene, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Clarissa Read
- Central Facility for Electron Microscopy, Ulm University, 89081 Ulm, Germany
- Institute of Virology, Ulm University Medical Center, 89081 Ulm, Germany
- Correspondence:
| |
Collapse
|
11
|
Brahim Belhaouari D, Baudoin JP, Lagier JC, Monnet-Corti V, La Scola B, Antezack A. Microscopic observations of SARS-CoV-2 like particles in different oral samples. Eur J Oral Sci 2022; 130:e12903. [PMID: 36404273 PMCID: PMC10099536 DOI: 10.1111/eos.12903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 10/21/2022] [Indexed: 11/22/2022]
Abstract
The emerging coronavirus pneumonia epidemic caused by the SARS-CoV-2 infection has spread rapidly around the world. The main routes of transmission of SARS-CoV-2 are currently recognised as aerosol/droplet inhalation. However, the involvement of the oral cavity in coronavirus disease 2019 (COVID-19) is poorly known. The current data indicates the presence of viral RNA in oral samples, suggesting the implication of saliva in SARS-CoV-2 transmission, however, no direct observation of SARS-CoV-2 particles in different oral samples has been reported. In this study, we investigated whether particles of SARS-CoV-2 were present in oral samples collected from three symptomatic COVID-19 patients. Using scanning electron microscopy (SEM), the correlative strategy of light microscopy and electron microscopy and immunofluorescence staining, we showed the presence of SARS-like particles in RT-qPCR SARS-CoV-2-positive saliva, dental plaque and gingival crevicular fluid (GCF) samples. In the saliva samples, we demonstrated the presence of epithelial oral cells with morphogenetic features of SARS-CoV-2 infected cells. Inside those cells, vacuoles filled with nascent particles were observed, suggesting the potential infection and replication of SARS-CoV-2 in oral tissues. Our results corroborate previous studies and confirm that the oral cavity may be a potential niche for SARS-CoV-2 infection and a potential source of transmission.
Collapse
Affiliation(s)
- Djamal Brahim Belhaouari
- IRD, AP-HM, IHU Méditerranée Infection, MEPHI, Aix Marseille Univ, Marseille, France.,Department of Veterinary Pathobiology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Jean-Pierre Baudoin
- IRD, AP-HM, IHU Méditerranée Infection, MEPHI, Aix Marseille Univ, Marseille, France.,Assistance Publique-Hopitaux de Marseille, Hopital Timone, Marseille, France
| | - Jean-Christophe Lagier
- IRD, AP-HM, IHU Méditerranée Infection, MEPHI, Aix Marseille Univ, Marseille, France.,Assistance Publique-Hopitaux de Marseille, Hopital Timone, Marseille, France
| | - Virginie Monnet-Corti
- IRD, AP-HM, IHU Méditerranée Infection, MEPHI, Aix Marseille Univ, Marseille, France.,Assistance Publique-Hopitaux de Marseille, Hopital Timone, Marseille, France.,Faculté des Sciences Médicales et Paramédicales, Ecole de Médecine Dentaire, Aix Marseille Univ, Marseille, France
| | - Bernard La Scola
- IRD, AP-HM, IHU Méditerranée Infection, MEPHI, Aix Marseille Univ, Marseille, France.,Assistance Publique-Hopitaux de Marseille, Hopital Timone, Marseille, France
| | - Angéline Antezack
- IRD, AP-HM, IHU Méditerranée Infection, MEPHI, Aix Marseille Univ, Marseille, France.,Assistance Publique-Hopitaux de Marseille, Hopital Timone, Marseille, France.,Faculté des Sciences Médicales et Paramédicales, Ecole de Médecine Dentaire, Aix Marseille Univ, Marseille, France
| |
Collapse
|
12
|
Jalali Moghaddam M, Ghavipour M. Towards smart diagnostic methods for COVID-19: Review of deep learning for medical imaging. IPEM-TRANSLATION 2022; 3:100008. [PMID: 36312890 PMCID: PMC9597575 DOI: 10.1016/j.ipemt.2022.100008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 11/08/2022]
Abstract
The infectious disease known as COVID-19 has spread dramatically all over the world since December 2019. The fast diagnosis and isolation of infected patients are key factors in slowing down the spread of this virus and better management of the pandemic. Although the CT and X-ray modalities are commonly used for the diagnosis of COVID-19, identifying COVID-19 patients from medical images is a time-consuming and error-prone task. Artificial intelligence has shown to have great potential to speed up and optimize the prognosis and diagnosis process of COVID-19. Herein, we review publications on the application of deep learning (DL) techniques for diagnostics of patients with COVID-19 using CT and X-ray chest images for a period from January 2020 to October 2021. Our review focuses solely on peer-reviewed, well-documented articles. It provides a comprehensive summary of the technical details of models developed in these articles and discusses the challenges in the smart diagnosis of COVID-19 using DL techniques. Based on these challenges, it seems that the effectiveness of the developed models in clinical use needs to be further investigated. This review provides some recommendations to help researchers develop more accurate prediction models.
Collapse
Affiliation(s)
- Marjan Jalali Moghaddam
- Department of Computer Engineering and Information Technology, Amirkabir University of Technology, Tehran, Iran
| | - Mina Ghavipour
- Department of Computer Engineering and Information Technology, Amirkabir University of Technology, Tehran, Iran
| |
Collapse
|
13
|
Concentration of SARS-CoV-2-Infected Cell Culture Supernatants for Detection of Virus-like Particles by Scanning Electron Microscopy. Viruses 2022; 14:v14112388. [PMID: 36366486 PMCID: PMC9698492 DOI: 10.3390/v14112388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/21/2022] [Accepted: 10/27/2022] [Indexed: 02/06/2023] Open
Abstract
There is currently a need for new rapid viral diagnostic electron microscopy methods. Although the gold standard remains the transmission electron microscopy (TEM) negative staining method for electron microscopic examination of samples containing a virus, difficulties can arise when the virus particle content of the sample that has to be examined is poor. Such samples include supernatants of virus-infected cells that can be difficult to examine, as sometimes only a few virus particles are released in the culture medium upon infection. In addition to TEM, scanning electron microscopy (SEM) can also be used for visualizing virus particles. One advantage of SEM over TEM is its ability to rapidly screen several large specimens, such as microscopy slides. In this study, we investigated this possibility and tested different coating molecules as well as the effect of centrifugation for analyzing SARS-CoV-2-virus-infected cell culture supernatants deposited on microscopy glass slides by SEM. We found that centrifugation of 25XConcanavalinA-coated microscopy glass slides in shell vials provided an improved method for concentrating SARS-CoV-2-virus-infected cell supernatants for virus-like particle detection by SEM.
Collapse
|
14
|
Heinl ES, Lorenz S, Schmidt B, Nasser M Laqtom N, Mazzulli JR, Francelle L, Yu TW, Greenberg B, Storch S, Tegtmeier I, Othmen H, Maurer K, Steinfurth M, Witzgall R, Milenkovic V, Wetzel CH, Reichold M. CLN7/MFSD8 may be an important factor for SARS-CoV-2 cell entry. iScience 2022; 25:105082. [PMID: 36093380 PMCID: PMC9444308 DOI: 10.1016/j.isci.2022.105082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 05/12/2022] [Accepted: 08/31/2022] [Indexed: 11/12/2022] Open
Abstract
The SARS-CoV-2 virus has triggered a worldwide pandemic. According to the BioGrid database, CLN7 (MFSD8) is thought to interact with several viral proteins. The aim of this work was to investigate a possible involvement of CLN7 in the infection process. Experiments on a CLN7-deficient HEK293T cell line exhibited a 90% reduced viral load compared to wild-type cells. This observation may be linked to the finding that CLN7 ko cells have a significantly reduced GM1 content in their cell membrane. GM1 is found highly enriched in lipid rafts, which are thought to play an important role in SARS-CoV-2 infection. In contrast, overexpression of CLN7 led to an increase in viral load. This study provides evidence that CLN7 is involved in SARS-CoV-2 infection. This makes it a potential pharmacological target for drug development against COVID-19. Furthermore, it provides insights into the physiological function of CLN7 where still only little is known about.
Collapse
Affiliation(s)
- Elena-Sofia Heinl
- Medical Cell Biology, University Regensburg, 93053 Regensburg, Germany
| | - Sebastian Lorenz
- Medical Cell Biology, University Regensburg, 93053 Regensburg, Germany
| | - Barbara Schmidt
- Institute of Clinical Microbiology and Hygiene, University of Regensburg, 93053 Regensburg, Germany
| | - Nouf Nasser M Laqtom
- Departments of Chemical Engineering and Genetics, Stanford University, Stanford, CA 94305, USA
| | - Joseph R. Mazzulli
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Laetitia Francelle
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Timothy W. Yu
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Benjamin Greenberg
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Stephan Storch
- Children’s Hospital Biochemistry, University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany
| | - Ines Tegtmeier
- Medical Cell Biology, University Regensburg, 93053 Regensburg, Germany
| | - Helga Othmen
- Medical Cell Biology, University Regensburg, 93053 Regensburg, Germany
- Institute for Molecular and Cellular Anatomy, University Regensburg, 93053 Regensburg, Germany
| | - Katja Maurer
- Medical Cell Biology, University Regensburg, 93053 Regensburg, Germany
| | - Malin Steinfurth
- Medical Cell Biology, University Regensburg, 93053 Regensburg, Germany
| | - Ralph Witzgall
- Institute for Molecular and Cellular Anatomy, University Regensburg, 93053 Regensburg, Germany
| | - Vladimir Milenkovic
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Christian H. Wetzel
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Markus Reichold
- Medical Cell Biology, University Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
15
|
Le Bideau M, Pires de Souza GA, Boschi C, Baudoin JP, Penant G, Jardot P, Fenollar F, Colson P, Lenk M, La Scola B. Limited permissibility of ENL-R and Mv-1-Lu mink cell lines to SARS-CoV-2. Front Microbiol 2022; 13:1003824. [PMID: 36312916 PMCID: PMC9597503 DOI: 10.3389/fmicb.2022.1003824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
The SARS-CoV-2 pandemic started in the end of 2019 in Wuhan, China, which highlighted the scenario of frequent cross-species transmission events. From the outbreak possibly initiated by viral spill-over into humans from an animal reservoir, now we face the human host moving globally while interacting with domesticated and peridomestic animals. The emergence of a new virus into the ecosystem leads to selecting forces and species-specific adaptations. The adaptation of SARS-CoV-2 to other animals represents a risk to controlling the dissemination of this coronavirus and the emergence of new variants. Since 2020, several mink farms in Europe and the United States have had SARS-CoV-2 outbreaks with human-mink and mink-human transmission, where the mink-selected variants possibly hold evolutionary concerning advantages. Here we investigated the permissibility of mink lung-derived cells using two cell lines, Mv-1-Lu and ENL-R, against several lineages of SARS-CoV-2, including some classified as variants of concern. The viral release rate and the infectious titers indicate that these cells support infections by different SARS-CoV-2 lineages. The viral production occurs in the first few days after infection with the low viral release by these mink cells, which is often absent for the omicron variant for lung cells. The electron microscopy reveals that during the viral replication cycle, the endomembrane system of the mink-host cell undergoes typical changes while the viral particles are produced, especially in the first days of infection. Therefore, even if limited, mink lung cells may represent a selecting source for SARS-CoV-2 variants, impacting their transmissibility and pathogenicity and making it difficult to control this new coronavirus.
Collapse
Affiliation(s)
- Marion Le Bideau
- Microbes, Evolution, Phylogénie et Infection (MEPHI), Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Marseille, France
- Institut Hospitalo-Universitaire (IHU) Méditerranée Infection, Marseille, France
| | - Gabriel Augusto Pires de Souza
- Microbes, Evolution, Phylogénie et Infection (MEPHI), Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Marseille, France
- Institut Hospitalo-Universitaire (IHU) Méditerranée Infection, Marseille, France
| | - Celine Boschi
- Microbes, Evolution, Phylogénie et Infection (MEPHI), Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Marseille, France
- Institut Hospitalo-Universitaire (IHU) Méditerranée Infection, Marseille, France
| | - Jean-Pierre Baudoin
- Microbes, Evolution, Phylogénie et Infection (MEPHI), Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Marseille, France
- Institut Hospitalo-Universitaire (IHU) Méditerranée Infection, Marseille, France
| | - Gwilherm Penant
- Microbes, Evolution, Phylogénie et Infection (MEPHI), Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Marseille, France
- Institut Hospitalo-Universitaire (IHU) Méditerranée Infection, Marseille, France
| | - Priscilla Jardot
- Microbes, Evolution, Phylogénie et Infection (MEPHI), Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Marseille, France
- Institut Hospitalo-Universitaire (IHU) Méditerranée Infection, Marseille, France
| | - Florence Fenollar
- Institut Hospitalo-Universitaire (IHU) Méditerranée Infection, Marseille, France
- Vecteurs – Infections Tropicales et Méditerranéennes (VITROME), Aix Marseille Univ, Institut Hospitalo-Universitaire (IHU), AP-HM, Marseille, France
| | - Philippe Colson
- Microbes, Evolution, Phylogénie et Infection (MEPHI), Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Marseille, France
- Institut Hospitalo-Universitaire (IHU) Méditerranée Infection, Marseille, France
| | - Matthias Lenk
- Collection of Cell Lines in Veterinary Medicine (CCLV), Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Bernard La Scola
- Microbes, Evolution, Phylogénie et Infection (MEPHI), Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), Marseille, France
- Institut Hospitalo-Universitaire (IHU) Méditerranée Infection, Marseille, France
| |
Collapse
|
16
|
Ultrastructural Characterization of Human Bronchial Epithelial Cells during SARS-CoV-2 Infection: Morphological Comparison of Wild-Type and CFTR-Modified Cells. Int J Mol Sci 2022; 23:ijms23179724. [PMID: 36077122 PMCID: PMC9455986 DOI: 10.3390/ijms23179724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/20/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
SARS-CoV-2 replicates in host cell cytoplasm. People with cystic fibrosis, considered at risk of developing severe symptoms of COVID-19, instead, tend to show mild symptoms. We, thus, analyzed at the ultrastructural level the morphological effects of SARS-CoV-2 infection on wild-type (WT) and F508del (ΔF) CFTR-expressing CFBE41o- cells at early and late time points post infection. We also investigated ACE2 expression through immune-electron microscopy. At early times of infection, WT cells exhibited double-membrane vesicles, representing typical replicative structures, with granular and vesicular content, while at late time points, they contained vesicles with viral particles. ∆F cells exhibited double-membrane vesicles with an irregular shape and degenerative changes and at late time of infection, showed vesicles containing viruses lacking a regular structure and a well-organized distribution. ACE2 was expressed at the plasma membrane and present in the cytoplasm only at early times in WT, while it persisted even at late times of infection in ΔF cells. The autophagosome content also differed between the cells: in WT cells, it comprised vesicles associated with virus-containing structures, while in ΔF cells, it comprised ingested material for lysosomal digestion. Our data suggest that CFTR-modified cells infected with SARS-CoV-2 have impaired organization of normo-conformed replicative structures.
Collapse
|
17
|
Schwab C, Domke LM, Rose F, Hausser I, Schirmacher P, Longerich T. Cell tropism and viral clearance during SARS-CoV-2 lung infection. Pathol Res Pract 2022; 236:154000. [PMID: 35797854 PMCID: PMC9245394 DOI: 10.1016/j.prp.2022.154000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/23/2022] [Accepted: 06/29/2022] [Indexed: 11/30/2022]
Abstract
Pulmonary capillary microthrombosis has been proposed as a major pathogenetic factor driving severe COVID-19. Autopsy studies reported endothelialitis but it is under debate if it is caused by SARS-CoV-2 infection of endothelial cells. In this study, RNA in situ hybridization was used to detect viral RNA and to identify the infected cell types in lung tissue of 40 patients with fatal COVID-19. SARS-CoV-2 Spike protein-coding RNA showed a steadily decreasing signal abundance over a period of three weeks. Besides the original virus strain the variants of concern Alpha (B.1.1.7), Delta (B.1.617.2), and Omicron (B.1.1.529) could also be detected by the assay. Viral RNA was mainly detected in alveolar macrophages and pulmonary epithelial cells, while only single virus-positive endothelial cells were observed even in cases with high viral load suggesting that viral infection of endothelial cells is not a key factor for the development of pulmonary capillary microthrombosis.
Collapse
|
18
|
Deroubaix A, Kramvis A. Imaging Techniques: Essential Tools for the Study of SARS-CoV-2 Infection. Front Cell Infect Microbiol 2022; 12:794264. [PMID: 35937687 PMCID: PMC9355083 DOI: 10.3389/fcimb.2022.794264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 06/21/2022] [Indexed: 01/08/2023] Open
Abstract
The world has seen the emergence of a new virus in 2019, SARS-CoV-2, causing the COVID-19 pandemic and millions of deaths worldwide. Microscopy can be much more informative than conventional detection methods such as RT-PCR. This review aims to present the up-to-date microscopy observations in patients, the in vitro studies of the virus and viral proteins and their interaction with their host, discuss the microscopy techniques for detection and study of SARS-CoV-2, and summarize the reagents used for SARS-CoV-2 detection. From basic fluorescence microscopy to high resolution techniques and combined technologies, this article shows the power and the potential of microscopy techniques, especially in the field of virology.
Collapse
Affiliation(s)
- Aurélie Deroubaix
- Hepatitis Virus Diversity Research Unit, Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Life Sciences Imaging Facility, University of the Witwatersrand, Johannesburg, South Africa
| | - Anna Kramvis
- Hepatitis Virus Diversity Research Unit, Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
19
|
Poma AM, Proietti A, Macerola E, Bonuccelli D, Conti M, Salvetti A, Dolo V, Chillà A, Basolo A, Santini F, Toniolo A, Basolo F. Suppression of Pituitary Hormone Genes in Subjects Who Died From COVID-19 Independently of Virus Detection in the Gland. J Clin Endocrinol Metab 2022; 107:2243-2253. [PMID: 35567590 PMCID: PMC9129148 DOI: 10.1210/clinem/dgac312] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Indexed: 01/08/2023]
Abstract
CONTEXT Involvement of the pituitary gland in SARS-CoV-2 infection has been clinically suggested by pituitary hormone deficiency in severe COVID-19 cases, by altered serum adrenocorticotropic hormone (ACTH) levels in hospitalized patients, and by cases of pituitary apoplexy. However, the direct viral infection of the gland has not been investigated. OBJECTIVE To evaluate whether the SARS-CoV-2 genome and antigens could be present in pituitary glands of lethal cases of COVID-19, and to assess possible changes in the expression of immune-related and pituitary-specific genes. METHODS SARS-CoV-2 genome and antigens were searched in the pituitary gland of 23 patients who died from COVID-19 and, as controls, in 12 subjects who died from trauma or sudden cardiac death. Real-time reverse transcription polymerase chain reaction (PCR), in situ hybridization, immunohistochemistry, and transmission electron microscopy were utilized. Levels of mRNA transcripts of immune-related and pituitary-specific genes were measured by the nCounter assay. RESULTS The SARS-CoV-2 genome and antigens were detected in 14/23 (61%) pituitary glands of the COVID-19 group, not in controls. In SARS-CoV-2-positive pituitaries, the viral genome was consistently detected by PCR in the adeno- and the neurohypophysis. Immunohistochemistry, in situ hybridization, and transmission electron microscopy confirmed the presence of SARS-CoV-2 in the pituitary. Activation of type I interferon signaling and enhanced levels of neutrophil and cytotoxic cell scores were found in virus-positive glands. mRNA transcripts of pituitary hormones and pituitary developmental/regulatory genes were suppressed in all COVID-19 cases irrespective of virus positivity. CONCLUSION Our study supports the tropism of SARS-CoV-2 for human pituitary and encourages exploration of pituitary dysfunction after COVID-19.
Collapse
Affiliation(s)
- Anello Marcello Poma
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Agnese Proietti
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Elisabetta Macerola
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Diana Bonuccelli
- Department of Forensic Medicine, Azienda USL Toscana Nordovest, Lucca, Italy
| | - Marco Conti
- Department of Forensic Medicine, Azienda USL Toscana Nordovest, Lucca, Italy
| | - Alessandra Salvetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Vincenza Dolo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Andrea Chillà
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Alessio Basolo
- Obesity and Lipodystrophy Center, Endocrinology Unit, University Hospital of Pisa, Pisa, Italy
| | - Ferruccio Santini
- Obesity and Lipodystrophy Center, Endocrinology Unit, University Hospital of Pisa, Pisa, Italy
| | | | - Fulvio Basolo
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
- Corresponding author: Fulvio Basolo, MD, PhD, Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Savi, 10, 56126 Pisa, Italy,
| |
Collapse
|
20
|
Shulunov VR. Rapid Parallel Search Technology with Scanning Electron Microscope and Artificial Neural Network. SMART SCIENCE 2022. [DOI: 10.1080/23080477.2022.2092671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Vyacheslav R Shulunov
- Institute of Physical Materials Science, Siberian Branch of the Russian Academy of Sciences, Ulan-Ude, Russia
| |
Collapse
|
21
|
Septisetyani EP, Prasetyaningrum PW, Anam K, Santoso A. SARS-CoV-2 Antibody Neutralization Assay Platforms Based on Epitopes Sources: Live Virus, Pseudovirus, and Recombinant S Glycoprotein RBD. Immune Netw 2022; 21:e39. [PMID: 35036026 PMCID: PMC8733193 DOI: 10.4110/in.2021.21.e39] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 12/15/2022] Open
Abstract
The high virulent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus that emerged in China at the end of 2019 has generated novel coronavirus disease, coronavirus disease 2019 (COVID-19), causing a pandemic worldwide. Every country has made great efforts to struggle against SARS-CoV-2 infection, including massive vaccination, immunological patients’ surveillance, and the utilization of convalescence plasma for COVID-19 therapy. These efforts are associated with the attempts to increase the titers of SARS-CoV-2 neutralizing Abs (nAbs) generated either after infection or vaccination that represent the body’s immune status. As there is no standard therapy for COVID-19 yet, virus eradication will mainly depend on these nAbs contents in the body. Therefore, serological nAbs neutralization assays become a requirement for researchers and clinicians to measure nAbs titers. Different platforms have been developed to evaluate nAbs titers utilizing various epitopes sources, including neutralization assays based on the live virus, pseudovirus, and neutralization assays utilizing recombinant SARS-CoV-2 S glycoprotein receptor binding site, receptor-binding domain. As a standard neutralization assay, the plaque reduction neutralization test (PRNT) requires isolation and propagation of live pathogenic SARS-CoV-2 virus conducted in a BSL-3 containment. Hence, other surrogate neutralization assays relevant to the PRNT play important alternatives that offer better safety besides facilitating high throughput analyses. This review discusses the current neutralization assay platforms used to evaluate nAbs, their techniques, advantages, and limitations.
Collapse
Affiliation(s)
- Endah Puji Septisetyani
- Research Center for Biotechnology, National Research and Innovation Agency, Bogor, West Java, Indonesia
| | | | - Khairul Anam
- Research Center for Biotechnology, National Research and Innovation Agency, Bogor, West Java, Indonesia
| | - Adi Santoso
- Research Center for Biotechnology, National Research and Innovation Agency, Bogor, West Java, Indonesia
| |
Collapse
|
22
|
Kanji JN, Pabbaraju K, Wong A, Beitku C, Deo A, Kailey S, Ma R, Tipples G, Zelyas N. Effect of not vortexing nasopharyngeal and throat swabs on SARS-CoV-2 nucleic acid detection: a pilot study. J Virol Methods 2022; 301:114468. [PMID: 35051444 PMCID: PMC8762992 DOI: 10.1016/j.jviromet.2022.114468] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/14/2022] [Accepted: 01/15/2022] [Indexed: 11/27/2022]
Abstract
The processing of swabs for respiratory virus detection involves vortexing while still in the viral transport medium (VTM). The effect of not vortexing swabs prior to analysis has not been studied extensively for SARS-CoV-2 detection, and presents an opportunity to improve pre-analytic laboratory workflow. We aimed to assess the impact of not vortexing nasopharyngeal/throat swabs submitted in VTM for SARS-CoV-2 testing. To assess the impact of not vortexing swabs, 277 swab samples were tested for SARS-CoV-2 RNA in paired vortexed and non-vortexed aliquots using eight routine nucleic acid amplification assays. We compared the qualitative (positive/negative) and semi-quantitative (cycle threshold, Ct) results. Following discordant analysis, all but one non-vortexed sample had the same qualitative result as the vortexed sample. 27.4 % of samples were SARS-CoV-2 positive. Comparison of Ct values revealed an apparent reduction in human cellular nucleic acid in the non-vortexed samples (mean Ct values of 24.0 and 26.5 for vortexed and non-vortexed samples, respectively, p < 0.0001) and increased Ct values for non-vortexed samples using a laboratory-developed SARS-CoV-2 assay (mean Ct values of 4.1 and 4.2 for vortexed and non-vortexed samples, respectively; p < 0.0001), but this was not observed for a more automated commercial SARS-CoV-2 assay (mean Ct values of 15.2 for both vortexed and non-vortexed samples, respectively; p = 0.68). While vortexing swabs appears to improve the recovery of cellular material, it does not have an appreciable impact on the qualitative sensitivity of SARS-CoV-2 nucleic acid tests, which may support omission of this step and simplification of front-end sample processing.
Collapse
Affiliation(s)
- Jamil N Kanji
- Public Health Laboratory, Alberta Precision Laboratories, Calgary, AB, Canada; Division of Infectious Diseases, Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Pathology & Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Laboratory Medicine & Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| | - Kanti Pabbaraju
- Public Health Laboratory, Alberta Precision Laboratories, Calgary, AB, Canada
| | - Anita Wong
- Public Health Laboratory, Alberta Precision Laboratories, Calgary, AB, Canada
| | - Candace Beitku
- Public Health Laboratory, Alberta Precision Laboratories, Edmonton, AB, Canada
| | - Ashwin Deo
- Public Health Laboratory, Alberta Precision Laboratories, Edmonton, AB, Canada
| | - Seema Kailey
- Public Health Laboratory, Alberta Precision Laboratories, Edmonton, AB, Canada
| | - Raymond Ma
- Public Health Laboratory, Alberta Precision Laboratories, Edmonton, AB, Canada
| | - Graham Tipples
- Public Health Laboratory, Alberta Precision Laboratories, Edmonton, AB, Canada; Department of Medical Microbiology & Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada; Li Ka Shing Institute for Virology, University of Alberta, Edmonton, AB, Canada
| | - Nathan Zelyas
- Department of Laboratory Medicine & Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada; Public Health Laboratory, Alberta Precision Laboratories, Edmonton, AB, Canada
| |
Collapse
|
23
|
Jha P, Singh P, Arora S, Sultan A, Nayek A, Ponnusamy K, Syed MA, Dohare R, Chopra M. Integrative multiomics and in silico analysis revealed the role of ARHGEF1 and its screened antagonist in mild and severe COVID-19 patients. J Cell Biochem 2022; 123:673-690. [PMID: 35037717 PMCID: PMC9015317 DOI: 10.1002/jcb.30213] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/21/2021] [Accepted: 12/30/2021] [Indexed: 12/22/2022]
Abstract
COVID‐19 is a sneaking deadly disease caused by severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2). The rapid increase in the number of infected patients worldwide enhances the exigency for medicines. However, precise therapeutic drugs are not available for COVID‐19; thus, exhaustive research is critically required to unscramble the pathogenic tools and probable therapeutic targets for the development of effective therapy. This study utilizes a chemogenomics strategy, including computational tools for the identification of viral‐associated differentially expressed genes (DEGs), and molecular docking of potential chemical compounds available in antiviral, anticancer, and natural product‐based libraries against these DEGs. We scrutinized the messenger RNA expression profile of SARS‐CoV‐2 patients, publicly available on the National Center for Biotechnology Information–Gene Expression Omnibus database, stratified them into different groups based on the severity of infection, superseded by identification of overlapping mild and severe infectious (MSI)‐DEGs. The profoundly expressed MSI‐DEGs were then subjected to trait‐linked weighted co‐expression network construction and hub module detection. The hub module MSI‐DEGs were then exposed to enrichment (gene ontology + pathway) and protein–protein interaction network analyses where Rho guanine nucleotide exchange factor 1 (ARHGEF1) gene conjectured in all groups and could be a probable target of therapy. Finally, we used the molecular docking and molecular dynamics method to identify inherent hits against the ARHGEF1 gene from antiviral, anticancer, and natural product‐based libraries. Although the study has an identified significant association of the ARHGEF1 gene in COVID19; and probable compounds targeting it, using in silico methods, these targets need to be validated by both in vitro and in vivo methods to effectively determine their therapeutic efficacy against the devastating virus.
Collapse
Affiliation(s)
- Prakash Jha
- Laboratory of Molecular Modeling and Anticancer Drug Development, Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, India
| | - Prithvi Singh
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Shweta Arora
- Department of Biotechnology, Translational Research Lab, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi, India
| | - Armiya Sultan
- Department of Biosciences, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi, India
| | - Arnab Nayek
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Kalaiarasan Ponnusamy
- Synthetic Biology Lab, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Mansoor Ali Syed
- Department of Biotechnology, Translational Research Lab, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi, India
| | - Ravins Dohare
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Madhu Chopra
- Laboratory of Molecular Modeling and Anticancer Drug Development, Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, India
| |
Collapse
|
24
|
Tng DJH, Yin BCY, Cao J, Ko KKK, Goh KCM, Chua DXW, Zhang Y, Chua MLK, Low JGH, Ooi EE, Soo KC. Amplified parallel antigen rapid test for point-of-care salivary detection of SARS-CoV-2 with improved sensitivity. Mikrochim Acta 2022; 189:14. [PMID: 34870771 PMCID: PMC8646336 DOI: 10.1007/s00604-021-05113-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/23/2021] [Indexed: 01/11/2023]
Abstract
In the ongoing COVID-19 pandemic, simple, rapid, point-of-care tests not requiring trained personnel for primary care testing are essential. Saliva-based antigen rapid tests (ARTs) can fulfil this need, but these tests require overnight-fasted samples; without which independent studies have demonstrated sensitivities of only 11.7 to 23.1%. Herein, we report an Amplified Parallel ART (AP-ART) with sensitivity above 90%, even with non-fasted samples. The virus was captured multimodally, using both anti-spike protein antibodies and Angiotensin Converting Enzyme 2 (ACE2) protein. It also featured two parallel flow channels. The first contained spike protein binding gold nanoparticles which produced a visible red line upon encountering the virus. The second contained signal amplifying nanoparticles that complex with the former and amplify the signal without any linker. Compared to existing dual gold amplification techniques, a limit of detection of one order of magnitude lower was achieved (0.0064 ng·mL-1). AP-ART performance in detecting SARS-CoV-2 in saliva of COVID-19 patients was investigated using a case-control study (139 participants enrolled and 162 saliva samples tested). Unlike commercially available ARTs, the sensitivity of AP-ART was maintained even when non-fasting saliva was used. Compared to the gold standard reverse transcription-polymerase chain reaction testing on nasopharyngeal samples, non-fasting saliva tested on AP-ART showed a sensitivity of 97.0% (95% CI: 84.7-99.8); without amplification, the sensitivity was 72.7% (95% CI: 83.7-94.8). Thus, AP-ART has the potential to be developed for point-of-care testing, which may be particularly important in resource-limited settings, and for early diagnosis to initiate newly approved therapies to reduce COVID-19 severity.
Collapse
Affiliation(s)
- Danny Jian Hang Tng
- Department of Infectious Diseases, Singapore General Hospital, 20 College Road, Singapore, 169856 Singapore
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore, 169857 Singapore
- Department of Head and Neck and Thoracic Cancers, Division of Radiation Oncology, National Cancer Centre Singapore, 11 Hospital Crescent, Singapore, 169610 Singapore
| | - Bryan Chu Yang Yin
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore, 169857 Singapore
| | - Jing Cao
- Department of Biomedical Engineering, National University Singapore, 4 Engineering Drive 3, Engineering Block 4, Singapore, 117583 Singapore
- State Key Laboratory for Oncogenes and Related Genes, School of Biomedical Engineering and Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai, 200030 People’s Republic of China
| | - Kwan Ki Karrie Ko
- Department of Microbiology, Singapore General Hospital, 20 College Road, Singapore, 169856 Singapore
| | - Kenneth Choon Meng Goh
- Department of Microbiology, Singapore General Hospital, 20 College Road, Singapore, 169856 Singapore
| | - Delia Xue Wen Chua
- Department of Biomedical Engineering, National University Singapore, 4 Engineering Drive 3, Engineering Block 4, Singapore, 117583 Singapore
| | - Yong Zhang
- Department of Biomedical Engineering, National University Singapore, 4 Engineering Drive 3, Engineering Block 4, Singapore, 117583 Singapore
| | - Melvin Lee Kiang Chua
- Department of Head and Neck and Thoracic Cancers, Division of Radiation Oncology, National Cancer Centre Singapore, 11 Hospital Crescent, Singapore, 169610 Singapore
- Division of Medical Sciences, National Cancer Centre Singapore, 11 Hospital Crescent, Singapore, 169610 Singapore
- Oncology Academic Programme, Duke-NUS Medical School, 8 College Road, Singapore, 169857 Singapore
| | - Jenny Guek Hong Low
- Department of Infectious Diseases, Singapore General Hospital, 20 College Road, Singapore, 169856 Singapore
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore, 169857 Singapore
| | - Eng Eong Ooi
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore, 169857 Singapore
| | - Khee Chee Soo
- Division of Medical Sciences, National Cancer Centre Singapore, 11 Hospital Crescent, Singapore, 169610 Singapore
- Oncology Academic Programme, Duke-NUS Medical School, 8 College Road, Singapore, 169857 Singapore
| |
Collapse
|
25
|
A step toward better sample management of COVID-19: On-spot detection by biometric technology and artificial intelligence. COVID-19 AND THE SUSTAINABLE DEVELOPMENT GOALS 2022. [PMCID: PMC9334987 DOI: 10.1016/b978-0-323-91307-2.00017-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
26
|
Grover A, Sinha R, Jyoti D, Faggio C. Imperative role of electron microscopy in toxicity assessment: A review. Microsc Res Tech 2021; 85:1976-1989. [PMID: 34904321 DOI: 10.1002/jemt.24029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/04/2021] [Accepted: 12/06/2021] [Indexed: 02/06/2023]
Abstract
Electron microscope (EM) was developed in 1931 and since then microscopical examination of both the biological and non-biological samples has been revolutionized. Modifications in electron microscopy techniques, such as scanning EM and transmission EM, have widened their applicability in the various sectors such as understanding of drug toxicity, development of mechanism, criminal site investigation, and characterization of the nano-molecule. The present review summarizes its role in important aspects such as toxicity assessment and disease diagnosis in special reference to SARS-COV2. In the biological system, EM studies have elucidated the impact of toxicants at the ultra-structural level in various tissue in conformity to physiological alterations. Thus, EM can be concluded as an important tool in toxicity assessment and disease prognosis.
Collapse
Affiliation(s)
- Aseem Grover
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Bajhol, India
| | - Reshma Sinha
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Bajhol, India
| | - Divya Jyoti
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Bajhol, India
| | - Caterina Faggio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy
| |
Collapse
|
27
|
Leigh KE, Modis Y. Imaging and visualizing SARS-CoV-2 in a new era for structural biology. Interface Focus 2021; 11:20210019. [PMID: 34956593 PMCID: PMC8504884 DOI: 10.1098/rsfs.2021.0019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2021] [Indexed: 01/18/2023] Open
Abstract
The SARS-CoV-2 pandemic has had a global impact and has put scientific endeavour in the spotlight, perhaps more than any previous viral outbreak. Fortuitously, the pandemic came at a time when decades of research in multiple scientific fields could be rapidly brought to bear, and a new generation of vaccine platforms was on the cusp of clinical maturity. SARS-CoV-2 also emerged at the inflection point of a technological revolution in macromolecular imaging by cryo-electron microscopy, fuelled by a confluence of major technological advances in sample preparation, optics, detectors and image processing software, that complemented pre-existing techniques. Together, these advances enabled us to visualize SARS-CoV-2 and its components more rapidly, in greater detail, and in a wider variety of biologically relevant contexts than would have been possible even a few years earlier. The resulting ultrastructural information on SARS-CoV-2 and how it interacts with the host cell has played a critical role in the much-needed accelerated development of COVID-19 vaccines and therapeutics. Here, we review key imaging modalities used to visualize SARS-CoV-2 and present select example data, which have provided us with an exceptionally detailed picture of this virus.
Collapse
Affiliation(s)
- Kendra E. Leigh
- Molecular Immunity Unit, Department of Medicine, MRC Laboratory of Molecular Biology, University of Cambridge, Francis Crick Avenue, Cambridge CB2 0QH, UK
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge School of Clinical Medicine, Cambridge CB2 0AW, UK
| | - Yorgo Modis
- Molecular Immunity Unit, Department of Medicine, MRC Laboratory of Molecular Biology, University of Cambridge, Francis Crick Avenue, Cambridge CB2 0QH, UK
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge School of Clinical Medicine, Cambridge CB2 0AW, UK
| |
Collapse
|
28
|
Three-dimensional reconstruction by electron tomography for the application to ultrastructural analysis of SARS-CoV-2 particles. Med Mol Morphol 2021; 55:60-67. [PMID: 34825978 PMCID: PMC8617558 DOI: 10.1007/s00795-021-00309-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 11/11/2021] [Indexed: 12/02/2022]
Abstract
SARS-CoV-2 is the cause of COVID-19. The three-dimensional morphology of viral particles existing and multiplying in infected cells has not been established by electron tomography, which is different from cryo-electron tomography using frozen samples. In this study, we establish the morphological structure of SARS-CoV-2 particles by three-dimensional reconstruction of images obtained by electron tomography and transmission electron microscopy of biological samples embedded in epoxy resin. The characteristic roots of spike structures were found to be arranged at the surface of a virion covered with an envelope. A high-electron-density structure that appears to be a nucleocapsid was observed inside the envelope of the virion on three-dimensional images reconstructed by electron tomography. The SARS-CoV-2 particles that budded in the vacuoles in the cytoplasm were morphologically identical to those found outside the cells, suggesting that mature and infectious SARS-CoV-2 particles were already produced in the vacuoles. Here, we show the three-dimensional morphological structure of SARS-CoV-2 particles reconstructed by electron tomography. To control infection, inhibition of viral release from vacuoles would be a new target in the development of prophylactic agents against SARS-CoV-2.
Collapse
|
29
|
A spatial multi-scale fluorescence microscopy toolbox discloses entry checkpoints of SARS-CoV-2 variants in Vero E6 cells. Comput Struct Biotechnol J 2021; 19:6140-6156. [PMID: 34745450 PMCID: PMC8562013 DOI: 10.1016/j.csbj.2021.10.038] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 01/12/2023] Open
Abstract
We exploited a multi-scale microscopy imaging toolbox to address some major issues related to SARS-CoV-2 interactions with host cells. Our approach harnesses both conventional and super-resolution fluorescence microscopy and easily matches the spatial scale of single-virus/cell checkpoints. After its validation through the characterization of infected cells and virus morphology, we leveraged this toolbox to reveal subtle issues related to the entry phase of SARS-CoV-2 variants in Vero E6 cells. Our results show that in Vero E6 cells the B.1.1.7 strain (aka Alpha Variant of Concern) is associated with much faster kinetics of endocytic uptake compared to its ancestor B.1.177. Given the cell-entry scenario dominated by the endosomal “late pathway”, the faster internalization of B.1.1.7 could be directly related to the N501Y mutation in the S protein, which is known to strengthen the binding of Spike receptor binding domain with ACE2. Remarkably, we also directly observed the central role of clathrin as a mediator of endocytosis in the late pathway of entry. In keeping with the clathrin-mediated endocytosis, we highlighted the non-raft membrane localization of ACE2. Overall, we believe that our fluorescence microscopy-based approach represents a fertile strategy to investigate the molecular features of SARS-CoV-2 interactions with cells.
Collapse
|
30
|
Radney JG, Weaver JL, Vicenzi EP, Staymates ME, Zangmeister CD. Filter Inserts Impact Cloth Mask Performance against Nano- to Micro-Sized Particles. ACS NANO 2021; 15:12860-12868. [PMID: 34251793 DOI: 10.1021/acsnano.1c05182] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
The United States Centers for Disease Control and Prevention and World Health Organization recognize that wearing cloth face coverings can slow the transmission of respiratory diseases via source control. Adding a partial layer of material with a high filtration efficiency (FE, e.g., polypropylene sheets that meet the HEPA standard) as an insert can potentially provide additional personal protection; however, data on the necessary areal coverage are sparse. The relationship between insert area ratio (IAR) relative to fabric area, FE, differential pressure (ΔP, a surrogate for breathability), and quality factor (QF, a ratio including FE and ΔP) utilizing two fabrics (rayon and 100% cotton lightweight flannel) and three insert materials (HEPA vacuum bag, sterilization wrap and paper coffee filter) was investigated. The effect of inserts on particle flows mimicking human exhalation is semiquantitatively and qualitatively examined using flow visualization techniques. The following was found: (1) The relationship between FE, ΔP, and QF is complex, and a trade-off exists between personal protection from filtration during inhalation and source control from leakage during exhalation; (2) FE and ΔP of the composite covering increase with IAR, and the rate is dependent upon insert type; (3) improvements (decrements) in the QF of the composite assemblage require inserts with a higher (lower) QF than the fabric and larger differences yield greater gains (losses); (4) the increased ΔP from an insert results in increased leakage during exhalation; (5) to minimize leaks, ΔP must be as low as possible; and (6) small relative areas not covered by an insert (i.e., IAR slightly smaller than 1) strongly deteriorate the benefits of an insert similar to small leaks in a covering.
Collapse
Affiliation(s)
- James G Radney
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Jamie L Weaver
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
- Museum Conservation Institute, Smithsonian Institution, Suitland, Maryland 20746, United States
| | - Edward P Vicenzi
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
- Museum Conservation Institute, Smithsonian Institution, Suitland, Maryland 20746, United States
| | - Matthew E Staymates
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Christopher D Zangmeister
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| |
Collapse
|
31
|
Secretory Vesicles Are the Principal Means of SARS-CoV-2 Egress. Cells 2021; 10:cells10082047. [PMID: 34440816 PMCID: PMC8393858 DOI: 10.3390/cells10082047] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/06/2021] [Accepted: 08/08/2021] [Indexed: 01/01/2023] Open
Abstract
The mechanisms of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) egress, similar to those of other coronaviruses, remain poorly understood. The virus buds in intracellular compartments and is therefore thought to be released by the biosynthetic secretory pathway. However, several studies have recently challenged this hypothesis. It has been suggested that coronaviruses, including SARS-CoV-2, use lysosomes for egress. In addition, a focused ion-beam scanning electron microscope (FIB/SEM) study suggested the existence of exit tunnels linking cellular compartments rich in viral particles to the extracellular space resembling those observed for the human immunodeficiency (HIV) in macrophages. Here, we analysed serial sections of Vero cells infected with SARS-CoV-2 by transmission electron microscopy (TEM). We found that SARS-CoV-2 was more likely to exit the cell in small secretory vesicles. Virus trafficking within the cells involves small vesicles, with each generally containing a single virus particle. These vesicles then fuse with the plasma membrane to release the virus into the extracellular space. This work sheds new light on the late stages of the SARS-CoV-2 infectious cycle of potential value for guiding the development of new antiviral strategies.
Collapse
|
32
|
Millette K, Cuala J, Wang P, Marks C, Woo V, Hayun M, Kang H, Martin M, Dhawan S, Chao L, Fraser S, Junge J, Lewis M, Georgia S. SARS-CoV2 infects pancreatic beta cells in vivo and induces cellular and subcellular disruptions that reflect beta cell dysfunction. RESEARCH SQUARE 2021:rs.3.rs-592374. [PMID: 34312617 PMCID: PMC8312902 DOI: 10.21203/rs.3.rs-592374/v1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Increasing evidence of new-onset diabetes during the COVID19 pandemic indicates that the SARS-CoV2 virus may drive beta-cell dysfunction leading to diabetes, but it is unclear if it is a primary or secondary effect. Here, we present evidence of SARS-CoV-2 infection of pancreatic beta cells in vivo using a robust and reproducible non-human primates model of mild to moderate COVID19 pathogenesis. Pancreas from SARS-CoV-2 infected subjects were positive for the SARS-CoV2 spike protein by immunohistochemistry and structures indicative of viral replication were evident by electron microscopy. Total beta cell area was decreased in SARS-CoV-2-infected pancreas, attributable to beta cell atrophy. Beta cell granularity was decreased. These histologic phenotypes persisted beyond the duration of the clinical disease course. Detailed electron microscopy of SARS-CoV-2 infected beta-cells revealed ultrastructural hallmarks of beta cell stress that are seen in islets of patients with Type 2 diabetes, including disrupted mitochondria and dilated endoplasmic reticulum. To assess the metabolic status of beta cells from SARS-CoV-2-infected subjects, we used fluorescence life-time imaging to measure the ratio of free and bound NADH as a surrogate of glycolytic and oxidative metabolism. We report an increase in free NADH levels, suggesting that beta cells from SARS-CoV-2-infected subjects adopt a more glycolytic metabolic profile. Taken together, we conclude that SARS-CoV-2 infection induces beta cell stress that may compromise beta-cell function beyond the duration of the disease course. This raises the possibility that the beta cell stress and injury may have clinical implications of the long-term future health of patients that have recovered from COVID19.
Collapse
|
33
|
Brouqui P, Drancourt M, Raoult D. COVID-19 Management at IHU Méditerranée Infection: A One-Year Experience. J Clin Med 2021; 10:2881. [PMID: 34209634 PMCID: PMC8268723 DOI: 10.3390/jcm10132881] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/11/2021] [Accepted: 06/25/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The Hospital-University Institute (IHU) Méditerranée Infection features a 27,000 square meter building hosting 700 employees and 75 hospitalized patients in the center of Marseille, France. METHOD Previous preparedness in contagious disease management allowed the IHU to manage the COVID-19 outbreak by continuing adaptation for optimal diagnosis, care and outcome. We report here the output of this management. RESULTS From 5 March 2020, and 26 April 2021, 608,313 PCR tests were provided for 424,919 patients and 44,089 returned positive. A total of 23,390 patients with COVID-19 were followed at IHU with an overall case fatality ratio of 1.7%. Of them 20,270 were followed as outpatients with an overall CFR of 0.17%. We performed 24,807 EKG, 5759 low dose CT Scanner, and 18,344 serology. Of the 7643 nasopharyngeal samples inoculated in cell cultures 3317 (43.3%) yielded SARS-Cov-2 isolates. Finally, 7370 SARS-Cov-2 genomes were analyzed, allowing description of the first genetic variants and their implication in the epidemiologic curves. Continuous clinical care quality evaluation provided the opportunity for 155 publications allowing a better understanding of the disease and improvement of care and 132 videos posted on the IHU Facebook network, totaling 60 million views and 390,000 followers, and dealing with COVID-19, outbreaks, epistemology, and ethics in medicine. CONCLUSIONS During this epidemic, IHU Méditerranée Infection played the role for which it has been created; useful clinical research to guarantee a high-quality diagnostic and care for patient and a recognized expertise.
Collapse
Affiliation(s)
| | | | - Didier Raoult
- Aix-Marseille University, IRD, MEPHI, IHU Méditerranée Infection, 13005 Marseille, France; (P.B.); (M.D.)
| | | |
Collapse
|
34
|
Pires de Souza GA, Osman IO, Le Bideau M, Baudoin JP, Jaafar R, Devaux C, La Scola B. Angiotensin II Receptor Blockers (ARBs Antihypertensive Agents) Increase Replication of SARS-CoV-2 in Vero E6 Cells. Front Cell Infect Microbiol 2021; 11:639177. [PMID: 34178717 PMCID: PMC8231006 DOI: 10.3389/fcimb.2021.639177] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 05/21/2021] [Indexed: 01/08/2023] Open
Abstract
Several comorbidities, including hypertension, have been associated with an increased risk of developing severe disease during SARS-CoV-2 infection. Angiotensin II receptor blockers (ARBs) are currently some of the most widely-used drugs to control blood pressure by acting on the angiotensin II type 1 receptor (AT1R). ARBs have been reported to trigger the modulation of the angiotensin I converting enzyme 2 (ACE2), the receptor used by the virus to penetrate susceptible cells, raising concern that such treatments may promote virus capture and increase their viral load in patients receiving ARBs therapy. In this in vitro study, we reviewed the effect of ARBs on ACE2 and AT1R expression and investigated whether treatment of permissive ACE2+/AT1R+ Vero E6 cells with ARBs alters SARS-CoV-2 replication in vitro in an angiotensin II-free system. After treating the cells with the ARBs, we observed an approximate 50% relative increase in SARS-CoV-2 production in infected Vero E6 cells that correlates with the ARBs-induced up-regulation of ACE2 expression. From this data, we believe that the use of ARBs in hypertensive patients infected by SARS-CoV-2 should be carefully evaluated.
Collapse
Affiliation(s)
- Gabriel Augusto Pires de Souza
- Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France.,Microbes, Evolution, Phylogénie et Infection (MEPHI), IHU - Méditerranée Infection, Marseille, France
| | - Ikram Omar Osman
- Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France.,Microbes, Evolution, Phylogénie et Infection (MEPHI), IHU - Méditerranée Infection, Marseille, France
| | - Marion Le Bideau
- Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France.,Microbes, Evolution, Phylogénie et Infection (MEPHI), IHU - Méditerranée Infection, Marseille, France
| | - Jean-Pierre Baudoin
- Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France.,Microbes, Evolution, Phylogénie et Infection (MEPHI), IHU - Méditerranée Infection, Marseille, France
| | - Rita Jaafar
- Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France.,Microbes, Evolution, Phylogénie et Infection (MEPHI), IHU - Méditerranée Infection, Marseille, France
| | - Christian Devaux
- Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France.,Microbes, Evolution, Phylogénie et Infection (MEPHI), IHU - Méditerranée Infection, Marseille, France.,Centre National de La Recherche Scientifique (CNRS), Marseille, France
| | - Bernard La Scola
- Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France.,Microbes, Evolution, Phylogénie et Infection (MEPHI), IHU - Méditerranée Infection, Marseille, France
| |
Collapse
|
35
|
Le Bideau M, Wurtz N, Baudoin JP, La Scola B. Innovative Approach to Fast Electron Microscopy Using the Example of a Culture of Virus-Infected Cells: An Application to SARS-CoV-2. Microorganisms 2021; 9:microorganisms9061194. [PMID: 34073053 PMCID: PMC8228702 DOI: 10.3390/microorganisms9061194] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 12/23/2022] Open
Abstract
Despite the development of new diagnostic methods, co-culture, based on sample inoculation of cell monolayers coupled with electron microscopy (EM) observation, remains the gold standard in virology. Indeed, co-culture allows for the study of cell morphology (infected and not infected), the ultrastructure of the inoculated virus, and the different steps of the virus infectious cycle. Most EM methods for studying virus cycles are applied after infected cells are produced in large quantities and detached to obtain a pellet. Here, cell culture was performed in sterilized, collagen-coated single-break strip wells. After one day in culture, cells were infected with SARS-CoV-2. Wells of interest were fixed at different time points, from 2 to 36 h post-infection. Microwave-assisted resin embedding was accomplished directly in the wells in 4 h. Finally, ultra-thin sections were cut directly through the infected-cell monolayers. Our methodology requires, in total, less than four days for preparing and observing cells. Furthermore, by observing undetached infected cell monolayers, we were able to observe new ultrastructural findings, such as cell–cell interactions and baso-apical cellular organization related to the virus infectious cycle. Our innovative methodology thus not only saves time for preparation but also adds precision and new knowledge about viral infection, as shown here for SARS-CoV-2.
Collapse
Affiliation(s)
- Marion Le Bideau
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique—Hôpitaux de Marseille (AP-HM), Aix-Marseille University, 13005 Marseille, France; (M.L.B.); (N.W.)
| | - Nathalie Wurtz
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique—Hôpitaux de Marseille (AP-HM), Aix-Marseille University, 13005 Marseille, France; (M.L.B.); (N.W.)
- IHU Méditerranée Infection, 13005 Marseille, France
| | - Jean-Pierre Baudoin
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique—Hôpitaux de Marseille (AP-HM), Aix-Marseille University, 13005 Marseille, France; (M.L.B.); (N.W.)
- Correspondence: (J.-P.B.); (B.L.S.)
| | - Bernard La Scola
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Assistance Publique—Hôpitaux de Marseille (AP-HM), Aix-Marseille University, 13005 Marseille, France; (M.L.B.); (N.W.)
- IHU Méditerranée Infection, 13005 Marseille, France
- Correspondence: (J.-P.B.); (B.L.S.)
| |
Collapse
|
36
|
Brahim Belhaouari D, Wurtz N, Grimaldier C, Lacoste A, Pires de Souza GA, Penant G, Hannat S, Baudoin JP, La Scola B. Microscopic Observation of SARS-Like Particles in RT-qPCR SARS-CoV-2 Positive Sewage Samples. Pathogens 2021; 10:516. [PMID: 33923138 PMCID: PMC8146039 DOI: 10.3390/pathogens10050516] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 12/18/2022] Open
Abstract
The ongoing outbreak of novel coronavirus pneumonia (COVID-19) caused by SARS-CoV-2 infection has spread rapidly worldwide. The major transmission routes of SARS-CoV-2 are recognised as inhalation of aerosol/droplets and person-to-person contact. However, some studies have demonstrated that live SARS-CoV-2 can be isolated from the faeces and urine of infected patients, which can then enter the wastewater system. The currently available evidence indicates that the viral RNA present in wastewater may become a potential source of epidemiological data. However, to investigate whether wastewater may present a risk to humans such as sewage workers, we investigated whether intact particles of SARS-CoV-2 were observable and whether it was possible to isolate the virus in wastewater. Using a correlative strategy of light microscopy and electron microscopy (CLEM), we demonstrated the presence of intact and degraded SARS-like particles in RT-qPCR SARS-CoV-2-positive sewage sample collected in the city of Marseille. However, the viral infectivity assessment of SARS-CoV-2 in the wastewater was inconclusive, due to the presence of other viruses known to be highly resistant in the environment such as enteroviruses, rhinoviruses, and adenoviruses. Although the survival and the infectious risk of SARS-CoV-2 in wastewater cannot be excluded from our study, additional work may be required to investigate the stability, viability, fate, and decay mechanisms of SARS-CoV-2 thoroughly in wastewater.
Collapse
Affiliation(s)
- Djamal Brahim Belhaouari
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Aix- Marseille University, 13005 Marseille, France; (D.B.B.); (N.W.); (G.A.P.d.S.); (S.H.)
- IHU Méditerranée Infection, 13005 Marseille, France
| | - Nathalie Wurtz
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Aix- Marseille University, 13005 Marseille, France; (D.B.B.); (N.W.); (G.A.P.d.S.); (S.H.)
- IHU Méditerranée Infection, 13005 Marseille, France
| | - Clio Grimaldier
- Assistance Publique—Hôpitaux de Marseille, 13005 Marseille, France; (C.G.); (G.P.)
| | - Alexandre Lacoste
- Bataillon des Marins Pompiers de Marseille, 13003 Marseille, France;
| | - Gabriel Augusto Pires de Souza
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Aix- Marseille University, 13005 Marseille, France; (D.B.B.); (N.W.); (G.A.P.d.S.); (S.H.)
- IHU Méditerranée Infection, 13005 Marseille, France
| | - Gwilherm Penant
- Assistance Publique—Hôpitaux de Marseille, 13005 Marseille, France; (C.G.); (G.P.)
| | - Sihem Hannat
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Aix- Marseille University, 13005 Marseille, France; (D.B.B.); (N.W.); (G.A.P.d.S.); (S.H.)
- IHU Méditerranée Infection, 13005 Marseille, France
| | - Jean-Pierre Baudoin
- Assistance Publique—Hôpitaux de Marseille, 13005 Marseille, France; (C.G.); (G.P.)
| | - Bernard La Scola
- Microbes, Evolution, Phylogeny and Infection (MEPHI), UM63, Institut de Recherche pour le Développement (IRD), Aix- Marseille University, 13005 Marseille, France; (D.B.B.); (N.W.); (G.A.P.d.S.); (S.H.)
- IHU Méditerranée Infection, 13005 Marseille, France
- Assistance Publique—Hôpitaux de Marseille, 13005 Marseille, France; (C.G.); (G.P.)
| |
Collapse
|
37
|
Liu YN, Lv ZT, Yang SY, Liu XW. Optical Tracking of the Interfacial Dynamics of Single SARS-CoV-2 Pseudoviruses. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2021; 55:4115-4122. [PMID: 33566596 PMCID: PMC7885801 DOI: 10.1021/acs.est.0c06962] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 05/20/2023]
Abstract
The frequent detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA in healthcare environments, accommodations, and wastewater has attracted great attention to the risk of viral transmission by environmental fomites. However, the process of SARS-CoV-2 adsorption to exposed surfaces in high-risk environments remains unclear. In this study, we investigated the interfacial dynamics of single SARS-CoV-2 pseudoviruses with plasmonic imaging technology. Through the use of this technique, which has high spatial and temporal resolution, we tracked the collision of viruses at a surface and differentiated their stable adsorption and transient adsorption. We determined the effect of the electrostatic force on virus adhesion by correlating the solution and surface chemistry with the interfacial diffusion velocity and equilibrium position. Viral adsorption was found to be enhanced in real scenarios, such as in simulated saliva. This work not only describes a plasmonic imaging method to examine the interfacial dynamics of a single virus but also provides direct measurements of the factors that regulate the interfacial adsorption of SARS-CoV-2 pseudovirus. Such information is valuable for understanding virus transport and environmental transmission and even for designing anticontamination surfaces.
Collapse
Affiliation(s)
- Yi-Nan Liu
- Chinese Academy of Sciences Key Laboratory of Urban
Pollutant Conversion, Department of Environmental Science and Engineering,
University of Science and Technology of China, Hefei 230026,
China
| | - Zhen-Ting Lv
- Chinese Academy of Sciences Key Laboratory of Urban
Pollutant Conversion, Department of Environmental Science and Engineering,
University of Science and Technology of China, Hefei 230026,
China
| | - Si-Yu Yang
- Chinese Academy of Sciences Key Laboratory of Urban
Pollutant Conversion, Department of Environmental Science and Engineering,
University of Science and Technology of China, Hefei 230026,
China
| | - Xian-Wei Liu
- Chinese Academy of Sciences Key Laboratory of Urban
Pollutant Conversion, Department of Environmental Science and Engineering,
University of Science and Technology of China, Hefei 230026,
China
- Department of Applied Chemistry,
University of Science and Technology of China, Hefei 230026,
China
| |
Collapse
|
38
|
Haspel J, Kim M, Zee P, Schwarzmeier T, Montagnese S, Panda S, Albani A, Merrow M. A Timely Call to Arms: COVID-19, the Circadian Clock, and Critical Care. J Biol Rhythms 2021; 36:55-70. [PMID: 33573430 PMCID: PMC7882674 DOI: 10.1177/0748730421992587] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
We currently find ourselves in the midst of a global coronavirus disease 2019 (COVID-19) pandemic, caused by the highly infectious novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here, we discuss aspects of SARS-CoV-2 biology and pathology and how these might interact with the circadian clock of the host. We further focus on the severe manifestation of the illness, leading to hospitalization in an intensive care unit. The most common severe complications of COVID-19 relate to clock-regulated human physiology. We speculate on how the pandemic might be used to gain insights on the circadian clock but, more importantly, on how knowledge of the circadian clock might be used to mitigate the disease expression and the clinical course of COVID-19.
Collapse
Affiliation(s)
- Jeffrey Haspel
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Minjee Kim
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Phyllis Zee
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Tanja Schwarzmeier
- Institute of Medical Psychology, Faculty of Medicine, LMU Munich, Munich, Germany
| | | | | | - Adriana Albani
- Institute of Medical Psychology, Faculty of Medicine, LMU Munich, Munich, Germany
- Department of Medicine IV, LMU Munich, Munich, Germany
| | - Martha Merrow
- Institute of Medical Psychology, Faculty of Medicine, LMU Munich, Munich, Germany
| |
Collapse
|
39
|
Müller JA, Groß R, Conzelmann C, Krüger J, Merle U, Steinhart J, Weil T, Koepke L, Bozzo CP, Read C, Fois G, Eiseler T, Gehrmann J, van Vuuren J, Wessbecher IM, Frick M, Costa IG, Breunig M, Grüner B, Peters L, Schuster M, Liebau S, Seufferlein T, Stenger S, Stenzinger A, MacDonald PE, Kirchhoff F, Sparrer KMJ, Walther P, Lickert H, Barth TFE, Wagner M, Münch J, Heller S, Kleger A. SARS-CoV-2 infects and replicates in cells of the human endocrine and exocrine pancreas. Nat Metab 2021; 3:149-165. [PMID: 33536639 DOI: 10.1038/s42255-021-00347-1] [Citation(s) in RCA: 337] [Impact Index Per Article: 112.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 01/14/2021] [Indexed: 02/07/2023]
Abstract
Infection-related diabetes can arise as a result of virus-associated β-cell destruction. Clinical data suggest that the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), causing the coronavirus disease 2019 (COVID-19), impairs glucose homoeostasis, but experimental evidence that SARS-CoV-2 can infect pancreatic tissue has been lacking. In the present study, we show that SARS-CoV-2 infects cells of the human exocrine and endocrine pancreas ex vivo and in vivo. We demonstrate that human β-cells express viral entry proteins, and SARS-CoV-2 infects and replicates in cultured human islets. Infection is associated with morphological, transcriptional and functional changes, including reduced numbers of insulin-secretory granules in β-cells and impaired glucose-stimulated insulin secretion. In COVID-19 full-body postmortem examinations, we detected SARS-CoV-2 nucleocapsid protein in pancreatic exocrine cells, and in cells that stain positive for the β-cell marker NKX6.1 and are in close proximity to the islets of Langerhans in all four patients investigated. Our data identify the human pancreas as a target of SARS-CoV-2 infection and suggest that β-cell infection could contribute to the metabolic dysregulation observed in patients with COVID-19.
Collapse
Affiliation(s)
- Janis A Müller
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Rüdiger Groß
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Carina Conzelmann
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Jana Krüger
- Department of Internal Medicine 1, Ulm University Hospital, Ulm, Germany
| | - Uta Merle
- Department of Internal Medicine 4, University of Heidelberg, Heidelberg, Germany
| | | | - Tatjana Weil
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Lennart Koepke
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | | | - Clarissa Read
- Central Facility for Electron Microscopy, Ulm University, Ulm, Germany
- Institute of Virology, Ulm University Medical Center, Ulm, Germany
| | - Giorgio Fois
- Institute of General Physiology, Ulm University, Ulm, Germany
| | - Tim Eiseler
- Department of Internal Medicine 1, Ulm University Hospital, Ulm, Germany
| | - Julia Gehrmann
- Institute for Computational Genomics, RWTH Aachen University, Aachen, Germany
| | - Joanne van Vuuren
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- School of Medicine, Technical University of Munich, Munich, Germany
| | - Isabel M Wessbecher
- Tissue Bank of the German Center for Infection Research, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Manfred Frick
- Institute of General Physiology, Ulm University, Ulm, Germany
| | - Ivan G Costa
- Institute for Computational Genomics, RWTH Aachen University, Aachen, Germany
| | - Markus Breunig
- Department of Internal Medicine 1, Ulm University Hospital, Ulm, Germany
| | - Beate Grüner
- Department of Internal Medicine 3, Ulm University Hospital, Ulm, Germany
| | - Lynn Peters
- Department of Internal Medicine 3, Ulm University Hospital, Ulm, Germany
| | - Michael Schuster
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Stefan Liebau
- Institute of Neuroanatomy & Developmental Biology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Thomas Seufferlein
- Department of Internal Medicine 1, Ulm University Hospital, Ulm, Germany
| | - Steffen Stenger
- Institute for Microbiology and Hygiene, Ulm University Medical Center, Ulm, Germany
| | | | - Patrick E MacDonald
- Alberta Diabetes Institute and Department of Pharmacology, University of Alberta, Edmonton, Canada
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | | | - Paul Walther
- Central Facility for Electron Microscopy, Ulm University, Ulm, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- School of Medicine, Technical University of Munich, Munich, Germany
| | | | - Martin Wagner
- Department of Internal Medicine 1, Ulm University Hospital, Ulm, Germany.
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.
| | - Sandra Heller
- Department of Internal Medicine 1, Ulm University Hospital, Ulm, Germany.
| | - Alexander Kleger
- Department of Internal Medicine 1, Ulm University Hospital, Ulm, Germany.
| |
Collapse
|
40
|
Eymieux S, Rouillé Y, Terrier O, Seron K, Blanchard E, Rosa-Calatrava M, Dubuisson J, Belouzard S, Roingeard P. Ultrastructural modifications induced by SARS-CoV-2 in Vero cells: a kinetic analysis of viral factory formation, viral particle morphogenesis and virion release. Cell Mol Life Sci 2021; 78:3565-3576. [PMID: 33449149 PMCID: PMC7809227 DOI: 10.1007/s00018-020-03745-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/08/2020] [Accepted: 12/16/2020] [Indexed: 12/17/2022]
Abstract
Many studies on SARS-CoV-2 have been performed over short-time scale, but few have focused on the ultrastructural characteristics of infected cells. We used TEM to perform kinetic analysis of the ultrastructure of SARS-CoV-2-infected cells. Early infection events were characterized by the presence of clusters of single-membrane vesicles and stacks of membrane containing nuclear pores called annulate lamellae (AL). A large network of host cell-derived organelles transformed into virus factories was subsequently observed in the cells. As previously described for other RNA viruses, these replication factories consisted of double-membrane vesicles (DMVs) located close to the nucleus. Viruses released at the cell surface by exocytosis harbored the typical crown of spike proteins, but viral particles without spikes were also observed in intracellular compartments, possibly reflecting incorrect assembly or a cell degradation process.
Collapse
Affiliation(s)
- Sébastien Eymieux
- Inserm U1259 MAVIVH, Université de Tours and CHRU de Tours, Tours, France
- Plate-Forme IBiSA de Microscopie Electronique, Université de Tours and CHRU de Tours, Tours, France
| | - Yves Rouillé
- Université Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, Lille, France
| | - Olivier Terrier
- Virologie Et Pathologie Humaine-VirPath Team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Karin Seron
- Université Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, Lille, France
| | - Emmanuelle Blanchard
- Inserm U1259 MAVIVH, Université de Tours and CHRU de Tours, Tours, France
- Plate-Forme IBiSA de Microscopie Electronique, Université de Tours and CHRU de Tours, Tours, France
| | - Manuel Rosa-Calatrava
- Virologie Et Pathologie Humaine-VirPath Team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Jean Dubuisson
- Université Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, Lille, France
| | - Sandrine Belouzard
- Université Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, Lille, France
| | - Philippe Roingeard
- Inserm U1259 MAVIVH, Université de Tours and CHRU de Tours, Tours, France.
- Plate-Forme IBiSA de Microscopie Electronique, Université de Tours and CHRU de Tours, Tours, France.
| |
Collapse
|
41
|
Al-Ahmady ZS, Ali-Boucetta H. Nanomedicine & Nanotoxicology Future Could Be Reshaped Post-COVID-19 Pandemic. FRONTIERS IN NANOTECHNOLOGY 2020. [DOI: 10.3389/fnano.2020.610465] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Since its first emergence in December 2019, the coronavirus-2 infection has quickly spread around the world and the severity of the pandemic has already re-shaped our lives. This review highlights the role of nanotechnology in the fight against this pandemic with a focus on the design of effective nano-based prevention and treatment options that overcome the limitations associated with conventional vaccines and other therapies. How nanotechnology could be utilized to understand the pathology of the ongoing pandemic is also discussed as well as how our knowledge about SARS-CoV-2 cellular uptake and toxicity could influence future nanotoxicological considerations and nanomedicine design of safe yet effective nanomaterials.
Collapse
|
42
|
Caldas LA, Carneiro FA, Higa LM, Monteiro FL, da Silva GP, da Costa LJ, Durigon EL, Tanuri A, de Souza W. Ultrastructural analysis of SARS-CoV-2 interactions with the host cell via high resolution scanning electron microscopy. Sci Rep 2020; 10:16099. [PMID: 32999356 PMCID: PMC7528159 DOI: 10.1038/s41598-020-73162-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/24/2020] [Indexed: 12/24/2022] Open
Abstract
SARS-CoV-2 is the cause of the ongoing COVID-19 pandemic. Here, we investigated the interaction of this new coronavirus with Vero cells using high resolution scanning electron microscopy. Surface morphology, the interior of infected cells and the distribution of viral particles in both environments were observed 2 and 48 h after infection. We showed areas of viral processing, details of vacuole contents, and viral interactions with the cell surface. Intercellular connections were also approached, and viral particles were adhered to these extensions suggesting direct cell-to-cell transmission of SARS-CoV-2.
Collapse
Affiliation(s)
- Lucio Ayres Caldas
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Cidade Universitária. Av., Carlos Chagas Filho 373, Prédio CCS, Bloco C, subsolo, CEP: 21941902, Rio de Janeiro, RJ, Brazil. .,Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Rio de Janeiro, Brazil. .,Núcleo Multidisciplinar de Pesquisa UFRJ-Xerém em Biologia - NUMPEX-BIO, Universidade Federal Do Rio de Janeiro, Campus Duque de Caxias Geraldo Cidade. CEP: 25265-970, Rio de Janeiro, RJ, Brazil.
| | - Fabiana Avila Carneiro
- Núcleo Multidisciplinar de Pesquisa UFRJ-Xerém em Biologia - NUMPEX-BIO, Universidade Federal Do Rio de Janeiro, Campus Duque de Caxias Geraldo Cidade. CEP: 25265-970, Rio de Janeiro, RJ, Brazil
| | - Luiza Mendonça Higa
- Departamento de Genética, Instituto de Biologia, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fábio Luiz Monteiro
- Departamento de Genética, Instituto de Biologia, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gustavo Peixoto da Silva
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana Jesus da Costa
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Edison Luiz Durigon
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Amilcar Tanuri
- Departamento de Genética, Instituto de Biologia, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Wanderley de Souza
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Cidade Universitária. Av., Carlos Chagas Filho 373, Prédio CCS, Bloco C, subsolo, CEP: 21941902, Rio de Janeiro, RJ, Brazil.,Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Rio de Janeiro, Brazil
| |
Collapse
|