1
|
Smail SW, Hirmiz SM, Ahmed AA, Albarzinji N, Awla HK, Amin K, Janson C. Decoding the intricacies: a comprehensive analysis of microRNAs in the pathogenesis, diagnosis, prognosis and therapeutic strategies for COVID-19. Front Med (Lausanne) 2024; 11:1430974. [PMID: 39434774 PMCID: PMC11492531 DOI: 10.3389/fmed.2024.1430974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
The pandemic of coronavirus disease-19 (COVID-19), provoked by the appearance of a novel coronavirus named severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), required a worldwide healthcare emergency. This has elicited an immediate need for accelerated research into its mechanisms of disease, criteria for diagnosis, methods for forecasting outcomes, and treatment approaches. microRNAs (miRNAs), are diminutive RNA molecules, that are non-coding and participate in gene expression regulation post-transcriptionally, having an important participation in regulating immune processes. miRNAs have granted substantial interest in their impact on viral replication, cell proliferation, and modulation of how the host's immune system responds. This narrative review delves into host miRNAs' multifaceted roles within the COVID-19 context, highlighting their involvement in disease progression, diagnostics, and prognostics aspects, given their stability in biological fluids and varied expression profiles when responding to an infection. Additionally, we discuss complicated interactions between SARS-CoV-2 and host cellular machinery facilitated by host miRNAs revealing how dysregulation of host miRNA expression profiles advances viral replication, immune evasion, and inflammatory responses. Furthermore, it investigates the potential of host miRNAs as therapeutic agents, whether synthetic or naturally occurring, which could be harnessed to either mitigate harmful inflammation or enhance antiviral responses. However, searching more deeply is needed to clarify how host's miRNAs are involved in pathogenesis of COVID-19, its diagnosis processes, prognostic assessments, and treatment approaches for patients.
Collapse
Affiliation(s)
- Shukur Wasman Smail
- College of Pharmacy, Cihan University-Erbil, Kurdistan Region, Erbil, Iraq
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Iraq
| | - Sarah Mousa Hirmiz
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Iraq
| | - Akhter Ahmed Ahmed
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Iraq
| | - Niaz Albarzinji
- Department of Medicine, Hawler Medical University, Erbil, Iraq
| | - Harem Khdir Awla
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Iraq
| | - Kawa Amin
- College of Medicine, University of Sulaimani, Sulaymaniyah, Iraq
- Department of Medical Sciences: Respiratory, Allergy and Sleep Research, Uppsala University, Uppsala, Sweden
| | - Christer Janson
- Department of Medical Sciences: Respiratory, Allergy and Sleep Research, Uppsala University, Uppsala, Sweden
| |
Collapse
|
2
|
Macharia Z, Ogoti B, Otieno M, Gitonga P, Bosco-Lauth A, Maritim M, Lemarkoko E, Keya A, Sankok J, Gitao G, Onono J, Oyugi J, Bowen RA. Transmission of SARS-CoV-2 among underserved pastoralist communities in Kajiado County, Kenya: 2020-2022. PLoS One 2024; 19:e0308318. [PMID: 39116080 PMCID: PMC11309416 DOI: 10.1371/journal.pone.0308318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 07/22/2024] [Indexed: 08/10/2024] Open
Abstract
Initial transmission of severe acute respiratory syndrome virus-2 (SARS-CoV-2) was highest in densely populated regions of Kenya. Transmission gradually trickled down to the less densely populated, remote and underserved regions such as the pastoral regions of Kajiado County which are characterized by poor healthcare systems. Molecular assays that were pivotal for COVID-19 diagnosis were not available in these regions. Serology is an alternative method for retrospectively tracking the transmission of SARS-CoV-2 in such populations. Dry blood spots (DBS) were prepared from consenting patients attending six health facilities in Kajiado County from March 2020 to March 2022. Upon elution, we conducted an enzyme-linked immunosorbent assay (ELISA) for the detection of SARS-Cov-2 IgG antibodies. Of the 908 DBSs we analyzed, 706 (78%) were from female participants. The overall seropositivity to SARS-Cov-2 antibodies was 7.3% (95% CI 5.7-9.1). The elderly (over 60 years) and male participants had a high likelihood of testing positive for SAR-CoV-2 infections. Mashuru (15.6%, 14/90) and Meto (15%, 19/127) health facilities registered the highest proportion of seropositive participants. Evidence of SARS-CoV-2 transmission among pastoralists in the remote and underserved regions of Kajiado County was established by DBS sampling and serologic testing.
Collapse
Affiliation(s)
- Zipporah Macharia
- Institute of Tropical and Infectious Diseases (UNITID), University of Nairobi, Nairobi, Kenya
- Department of Medical Microbiology and Immunology, University of Nairobi, Nairobi, Kenya
| | - Brian Ogoti
- Department of Medical Microbiology and Immunology, University of Nairobi, Nairobi, Kenya
- Center of Epidemiological Modelling and Analysis, University of Nairobi, Nairobi, Kenya
| | - Magdaline Otieno
- Institute of Tropical and Infectious Diseases (UNITID), University of Nairobi, Nairobi, Kenya
| | - Pauline Gitonga
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Angela Bosco-Lauth
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Marybeth Maritim
- Department of Clinical Medicine and Therapeutics, University of Nairobi, Nairobi, Kenya
| | | | - Aggrey Keya
- Kajiado County Referral Hospital, Kajiado Town, Kajiado, Kenya
| | - Joseph Sankok
- Kajiado County Referral Hospital, Kajiado Town, Kajiado, Kenya
| | - George Gitao
- Department of Pathology, Microbiology and Parasitology, University of Nairobi, Nairobi, Kenya
| | - Joshua Onono
- Department of Public Health Pharmacology and Toxicology, University of Nairobi, Nairobi, Kenya
| | - Julius Oyugi
- Institute of Tropical and Infectious Diseases (UNITID), University of Nairobi, Nairobi, Kenya
- Department of Medical Microbiology and Immunology, University of Nairobi, Nairobi, Kenya
| | - Richard A. Bowen
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| |
Collapse
|
3
|
Silva PV, Nobre CN. Computational methods in the analysis of SARS-CoV-2 in mammals: A systematic review of the literature. Comput Biol Med 2024; 173:108264. [PMID: 38564853 DOI: 10.1016/j.compbiomed.2024.108264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 02/15/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024]
Abstract
SARS-CoV-2 is an enveloped RNA virus that causes severe respiratory illness in humans and animals. It infects cells by binding the Spike protein to the host's angiotensin-converting enzyme 2 (ACE2). The bat is considered the natural host of the virus, and zoonotic transmission is a significant risk and can happen when humans come into close contact with infected animals. Therefore, understanding the interconnection between human, animal, and environmental health is important to prevent and control future coronavirus outbreaks. This work aimed to systematically review the literature to identify characteristics that make mammals suitable virus transmitters and raise the main computational methods used to evaluate SARS-CoV-2 in mammals. Based on this review, it was possible to identify the main factors related to transmissions mentioned in the literature, such as the expression of ACE2 and proximity to humans, in addition to identifying the computational methods used for its study, such as Machine Learning, Molecular Modeling, Computational Simulation, between others. The findings of the work contribute to the prevention and control of future outbreaks, provide information on transmission factors, and highlight the importance of advanced computational methods in the study of infectious diseases that allow a deeper understanding of transmission patterns and can help in the development of more effective control and intervention strategies.
Collapse
Affiliation(s)
- Paula Vitória Silva
- Pontifical Catholic University of Minas Gerais - PUC Minas, 500 Dom José Gaspar Street, Building 41, Coração Eucarístico, Belo Horizonte, MG 30535-901, Brazil.
| | - Cristiane N Nobre
- Pontifical Catholic University of Minas Gerais - PUC Minas, 500 Dom José Gaspar Street, Building 41, Coração Eucarístico, Belo Horizonte, MG 30535-901, Brazil.
| |
Collapse
|
4
|
Giarola JF, Soler M, Estevez MC, Tarasova A, Le Poder S, Wasniewski M, Decaro N, Lechuga LM. Validation of a plasmonic-based serology biosensor for veterinary diagnosis of COVID-19 in domestic animals. Talanta 2024; 271:125685. [PMID: 38262129 DOI: 10.1016/j.talanta.2024.125685] [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: 12/05/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 01/25/2024]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic recently demonstrated the devastating impact on public health, economy, and social development of zoonotic infectious diseases, whereby viruses jump from animals to infect humans. Due to this potential of viruses to cross the species barrier, the surveillance of infectious pathogens circulation in domestic and close-to-human animals is indispensable, as they could be potential reservoirs. Optical biosensors, mainly those based on Surface Plasmon Resonance (SPR), have widely demonstrated its ability for providing direct, label-free, and quantitative bioanalysis with excellent sensitivity and reliability. This biosensor technology can provide a powerful tool to the veterinary field, potentially being helpful for the monitoring of the infection spread. We have implemented a multi-target COVID-19 serology plasmonic biosensor for the rapid testing and screening of common European domestic animals. The multi-target serological biosensor assay enables the detection of total SARS-CoV-2 antibodies (IgG + IgM) generated towards both S and N viral antigens. The analysis is performed in less than 15 min with a low-volume serum sample (<20 μL, 1:10 dilution), reaching a limit of detection of 49.6 ng mL-1. A complete validation has been carried out with hamster, dog, and cat sera samples (N = 75, including 37 COVID-19-positive and 38 negative samples). The biosensor exhibits an excellent diagnostic sensitivity (100 %) and good specificity (71.4 %) for future application in veterinary settings. Furthermore, the biosensor technology is integrated into a compact, portable, and user-friendly device, well-suited for point-of-care testing. This study positions our plasmonic biosensor as an alternative and reliable diagnostic tool for COVID-19 serology in animal samples, expanding the applicability of plasmonic technologies for decentralized analysis in veterinary healthcare and animal research.
Collapse
Affiliation(s)
- Juliana Fátima Giarola
- Nanobiosensors and Bioanalytical Applications Group (NanoB2A), Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC, CIBER-BBN and BIST, 08193, Bellaterra, Barcelona, Spain
| | - Maria Soler
- Nanobiosensors and Bioanalytical Applications Group (NanoB2A), Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC, CIBER-BBN and BIST, 08193, Bellaterra, Barcelona, Spain.
| | - M-Carmen Estevez
- Nanobiosensors and Bioanalytical Applications Group (NanoB2A), Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC, CIBER-BBN and BIST, 08193, Bellaterra, Barcelona, Spain
| | - Anna Tarasova
- Nanobiosensors and Bioanalytical Applications Group (NanoB2A), Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC, CIBER-BBN and BIST, 08193, Bellaterra, Barcelona, Spain
| | - Sophie Le Poder
- UMR Virologie, INRAE, ANSES, École Nationale Vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Marine Wasniewski
- Interfas Unit, Nancy Laboratory for Rabies and Wildlife, ANSES, 54220, Malzéville, France
| | - Nicola Decaro
- Department of Veterinary Medicine, University of Bari Aldo Moro, 70121, Bari, Italy
| | - Laura M Lechuga
- Nanobiosensors and Bioanalytical Applications Group (NanoB2A), Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC, CIBER-BBN and BIST, 08193, Bellaterra, Barcelona, Spain
| |
Collapse
|
5
|
Rasheed WS, Sarkees AN. Immediate Adverse Reaction and SARS-CoV-2 Anti-Spike Receptor Binding Domain IgG of COVID-19 Vaccines Among Health Staffs. Disaster Med Public Health Prep 2024; 18:e66. [PMID: 38618867 DOI: 10.1017/dmp.2024.60] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
OBJECTIVE To contain the spread of coronavirus disease 2019 (COVID-19), several vaccines have been developed. This study is intended to elucidate the level of anti-severe acute respiratory syndrome coronavirus 2 immunoglobulin G (anti-SARS-CoV-2-IgG) antibodies for COVID-19 vaccines (Pfizer BioNTech [BNT162b2], Oxford/AstraZeneca [ChAdOx1], and Sinopharm [BBIBP-CorV]) among health staff from health facilities in Duhok province, and it explored the immediate adverse reactions of COVID-19 vaccines among participants. METHODS A longitudinal study was conducted from June 1, 2021, to June 30, 2022, and 300 participants were included through simple random sampling. RESULTS The immune response 1 mo after the second dose was significantly higher than the sustained immune after 5 and 9 mo as results revealed that, in 100% of study samples who had (ChAdOx1) vaccine, their antibody titers exceeded the positivity threshold of 1 AU/m, while 96% for (BNT162b2) and 90% for (BBIBP-CorV) for the first test after 1 mo from the second dose of the COVID-19 vaccine, and these rates were reduced to 94.6% for (ChAdOx1), 97.8% for (BNT162b2), and 81.9% for (BBIBP-CorV) at 5 mo after the second dose, while simultaneously the seropositivity rates were more reduced at 9 mo to 46.5% for (ChAdOx1), 67.5% for (BNT162b2), and 9.20% for (BBIBP-CorV). In terms of adverse reactionsss, fever was reported as the most prevalent after the first dose in 58% for ChAdOx1, 43% for BNT162b2, and 23% for BBIBP-CorV, followed by muscle pain, joint pain, and shoulder pain for both doses. CONCLUSIONS The implications of the findings from this study are that higher and potentially longer antibody responses can be obtained if the BNT162b2 is given as compared with the other 2 vaccines. Moreover, the booster doses of the COVID-19 vaccine are highly recommended because more than 50% of the participants either have become anti-spike protein negative or have a deficient level of anti-spike protein against COVD-19 vaccines.
Collapse
Affiliation(s)
- Waleed S Rasheed
- Department of Public Health, College of health and Medical Technology, Duhok Polytechnic University, Kurdistan Region-Iraq
| | - Alaa Noori Sarkees
- Department of Nursing, College of health and Medical Technology, Duhok Polytechnic University, Kurdistan Region-Iraq
| |
Collapse
|
6
|
Xie X, Lan Q, Zhao J, Zhang S, Liu L, Zhang Y, Xu W, Shao M, Peng J, Xia S, Zhu Y, Zhang K, Zhang X, Zhang R, Li J, Dai W, Ge Z, Hu S, Yu C, Wang J, Ma D, Zheng M, Yang H, Xiao G, Rao Z, Lu L, Zhang L, Bai F, Zhao Y, Jiang S, Liu H. Structure-based design of pan-coronavirus inhibitors targeting host cathepsin L and calpain-1. Signal Transduct Target Ther 2024; 9:54. [PMID: 38443334 PMCID: PMC10914734 DOI: 10.1038/s41392-024-01758-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 01/19/2024] [Accepted: 01/25/2024] [Indexed: 03/07/2024] Open
Abstract
Respiratory disease caused by coronavirus infection remains a global health crisis. Although several SARS-CoV-2-specific vaccines and direct-acting antivirals are available, their efficacy on emerging coronaviruses in the future, including SARS-CoV-2 variants, might be compromised. Host-targeting antivirals provide preventive and therapeutic strategies to overcome resistance and manage future outbreak of emerging coronaviruses. Cathepsin L (CTSL) and calpain-1 (CAPN1) are host cysteine proteases which play crucial roles in coronaviral entrance into cells and infection-related immune response. Here, two peptidomimetic α-ketoamide compounds, 14a and 14b, were identified as potent dual target inhibitors against CTSL and CAPN1. The X-ray crystal structures of human CTSL and CAPN1 in complex with 14a and 14b revealed the covalent binding of α-ketoamide groups of 14a and 14b to C25 of CTSL and C115 of CAPN1. Both showed potent and broad-spectrum anticoronaviral activities in vitro, and it is worth noting that they exhibited low nanomolar potency against SARS-CoV-2 and its variants of concern (VOCs) with EC50 values ranging from 0.80 to 161.7 nM in various cells. Preliminary mechanistic exploration indicated that they exhibited anticoronaviral activity through blocking viral entrance. Moreover, 14a and 14b exhibited good oral pharmacokinetic properties in mice, rats and dogs, and favorable safety in mice. In addition, both 14a and 14b treatments demonstrated potent antiviral potency against SARS-CoV-2 XBB 1.16 variant infection in a K18-hACE2 transgenic mouse model. And 14b also showed effective antiviral activity against HCoV-OC43 infection in a mouse model with a final survival rate of 60%. Further evaluation showed that 14a and 14b exhibited excellent anti-inflammatory effects in Raw 264.7 mouse macrophages and in mice with acute pneumonia. Taken together, these results suggested that 14a and 14b are promising drug candidates, providing novel insight into developing pan-coronavirus inhibitors with antiviral and anti-inflammatory properties.
Collapse
Affiliation(s)
- Xiong Xie
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiaoshuai Lan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
| | - Jinyi Zhao
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Sulin Zhang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lu Liu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yumin Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Wei Xu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
| | - Maolin Shao
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Jingjing Peng
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shuai Xia
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
| | - Yan Zhu
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Keke Zhang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xian Lin Road, Jiangsu, 210023, Nanjing, China
| | - Xianglei Zhang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Ruxue Zhang
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jian Li
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xian Lin Road, Jiangsu, 210023, Nanjing, China
| | - Wenhao Dai
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhen Ge
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xian Lin Road, Jiangsu, 210023, Nanjing, China
| | - Shulei Hu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Changyue Yu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiang Wang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Dakota Ma
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Mingyue Zheng
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xian Lin Road, Jiangsu, 210023, Nanjing, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310024, China
| | - Haitao Yang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Gengfu Xiao
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Zihe Rao
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
| | - Leike Zhang
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Fang Bai
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Yao Zhao
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Shenzhen, 518112, China.
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China.
| | - Hong Liu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xian Lin Road, Jiangsu, 210023, Nanjing, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310024, China.
| |
Collapse
|
7
|
Zyoud S. Global Mapping and Visualization Analysis of One Health Knowledge in the COVID-19 Context. ENVIRONMENTAL HEALTH INSIGHTS 2024; 18:11786302241236017. [PMID: 38449589 PMCID: PMC10916474 DOI: 10.1177/11786302241236017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 02/13/2024] [Indexed: 03/08/2024]
Abstract
Globally, the COVID-19 pandemic had a significant impact on the health, social, and economic systems, triggering lasting damage and exposing the complexity of the problem beyond just being a health emergency. This crisis has highlighted the need for a comprehensive and collaborative strategy to successfully counter infectious diseases and other global challenges. With the COVID-19 pandemic pushing One Health to the forefront of global health and sustainable development agendas, this concept has emerged as a potential approach for addressing these challenges. In the context of COVID-19, this study investigates global knowledge about One Health by examining its state, significant contributions, and future directions. It seeks to offer an integrated framework of insights guiding the development of well-informed decisions. A comprehensive search using the Scopus database was conducted, employing specific terms related to One Health and COVID-19. VOSviewer 1.6.19 software was used to generate network visualization maps. Countries' research output was adjusted based on their gross domestic product (GDP) and population size. The study identified a total of 527 publications. The United States led with 134 documents (25.4%), but India topped the adjusted ranking. One Health journal stood as the most common outlet for disseminating knowledge (49 documents; 9.3%), while Centers for Disease Control and Prevention (CDC), the United States emerged as the most prolific institution (13 documents; 2.5%). Key topics were related to the virus transmission mechanisms, climate change impacts, antimicrobial resistance, ecosystem health, preparedness, collaboration, community engagement, and developing of efficient surveillance systems. The study emphasizes how critical it is to capitalize on the present momentum of COVID-19 to advance One Health concepts. Integrating social and environmental sciences, and a variety of professions for better interaction and collaboration is crucial. Additionally, increased funding for developing countries, and legislative empowerment are vital to advance One Health and boost disease prevention.
Collapse
Affiliation(s)
- Shaher Zyoud
- Department of Building Engineering & Environment,Palestine Technical University (Kadoorie), Tulkarem, Palestine
- Department of Civil Engineering & Sustainable Structures,Palestine Technical University (Kadoorie), Tulkarem, Palestine
| |
Collapse
|
8
|
Zech F, Jung C, Jacob T, Kirchhoff F. Causes and Consequences of Coronavirus Spike Protein Variability. Viruses 2024; 16:177. [PMID: 38399953 PMCID: PMC10892391 DOI: 10.3390/v16020177] [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: 12/28/2023] [Revised: 01/20/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
Coronaviruses are a large family of enveloped RNA viruses found in numerous animal species. They are well known for their ability to cross species barriers and have been transmitted from bats or intermediate hosts to humans on several occasions. Four of the seven human coronaviruses (hCoVs) are responsible for approximately 20% of common colds (hCoV-229E, -NL63, -OC43, -HKU1). Two others (SARS-CoV-1 and MERS-CoV) cause severe and frequently lethal respiratory syndromes but have only spread to very limited extents in the human population. In contrast the most recent human hCoV, SARS-CoV-2, while exhibiting intermediate pathogenicity, has a profound impact on public health due to its enormous spread. In this review, we discuss which initial features of the SARS-CoV-2 Spike protein and subsequent adaptations to the new human host may have helped this pathogen to cause the COVID-19 pandemic. Our focus is on host forces driving changes in the Spike protein and their consequences for virus infectivity, pathogenicity, immune evasion and resistance to preventive or therapeutic agents. In addition, we briefly address the significance and perspectives of broad-spectrum therapeutics and vaccines.
Collapse
Affiliation(s)
- Fabian Zech
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Christoph Jung
- Institute of Electrochemistry, Ulm University, 89081 Ulm, Germany; (C.J.); (T.J.)
- Helmholtz-Institute Ulm (HIU) Electrochemical Energy Storage, 89081 Ulm, Germany
- Karlsruhe Institute of Technology (KIT), 76021 Karlsruhe, Germany
| | - Timo Jacob
- Institute of Electrochemistry, Ulm University, 89081 Ulm, Germany; (C.J.); (T.J.)
- Helmholtz-Institute Ulm (HIU) Electrochemical Energy Storage, 89081 Ulm, Germany
- Karlsruhe Institute of Technology (KIT), 76021 Karlsruhe, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| |
Collapse
|
9
|
Bedding MJ, Franck C, Johansen-Leete J, Aggarwal A, Maxwell JWC, Patel K, Hawkins PME, Low JKK, Siddiquee R, Sani HM, Ford DJ, Turville S, Mackay JP, Passioura T, Christie M, Payne RJ. Discovery of High Affinity Cyclic Peptide Ligands for Human ACE2 with SARS-CoV-2 Entry Inhibitory Activity. ACS Chem Biol 2024; 19:141-152. [PMID: 38085789 DOI: 10.1021/acschembio.3c00568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
The development of effective antiviral compounds is essential for mitigating the effects of the COVID-19 pandemic. Entry of SARS-CoV-2 virions into host cells is mediated by the interaction between the viral spike (S) protein and membrane-bound angiotensin-converting enzyme 2 (ACE2) on the surface of epithelial cells. Inhibition of this viral protein-host protein interaction is an attractive avenue for the development of antiviral molecules with numerous spike-binding molecules generated to date. Herein, we describe an alternative approach to inhibit the spike-ACE2 interaction by targeting the spike-binding interface of human ACE2 via mRNA display. Two consecutive display selections were performed to direct cyclic peptide ligand binding toward the spike binding interface of ACE2. Through this process, potent cyclic peptide binders of human ACE2 (with affinities in the picomolar to nanomolar range) were identified, two of which neutralized SARS-CoV-2 entry. This work demonstrates the potential of targeting ACE2 for the generation of anti-SARS-CoV-2 therapeutics as well as broad spectrum antivirals for the treatment of SARS-like betacoronavirus infection.
Collapse
Affiliation(s)
- Max J Bedding
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Charlotte Franck
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Jason Johansen-Leete
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | | | - Joshua W C Maxwell
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Karishma Patel
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Paige M E Hawkins
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Jason K K Low
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Rezwan Siddiquee
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Hakimeh Moghaddas Sani
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Daniel J Ford
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | | | - Joel P Mackay
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Toby Passioura
- Sydney Analytical Core Research Facility, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Mary Christie
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
- School of Medical Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Richard J Payne
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
10
|
Tong X, Keung W, Arnold LD, Stevens LJ, Pruijssers AJ, Kook S, Lopatin U, Denison M, Kwong AD. Evaluation of in vitro antiviral activity of SARS-CoV-2 M pro inhibitor pomotrelvir and cross-resistance to nirmatrelvir resistance substitutions. Antimicrob Agents Chemother 2023; 67:e0084023. [PMID: 37800975 PMCID: PMC10649086 DOI: 10.1128/aac.00840-23] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 07/31/2023] [Indexed: 10/07/2023] Open
Abstract
The unprecedented scale of the COVID-19 pandemic and the rapid evolution of SARS-CoV-2 variants underscore the need for broadly active inhibitors with a high barrier to resistance. The coronavirus main protease (Mpro) is an essential cysteine protease required for viral polyprotein processing and is highly conserved across human coronaviruses. Pomotrelvir is a novel Mpro inhibitor that has recently completed a phase 2 clinical trial. In this report, we demonstrated that pomotrelvir is a potent competitive inhibitor of SARS-CoV-2 Mpro with high selectivity against human proteases. In the enzyme assay, pomotrelvir is also active against Mpro proteins derived from human coronaviruses CoV-229E, CoV-OC43, CoV-HKU1, CoV-NL63, MERS, and SARS-CoV. In cell-based SARS-CoV-2 replicon and SARS-CoV-2 infection assays, pomotrelvir has shown potent inhibitory activity and is broadly active against SARS-CoV-2 clinical isolates including Omicron variants. Many resistance substitutions of the Mpro inhibitor nirmatrelvir confer cross-resistance to pomotrelvir, consistent with the finding from our enzymatic analysis that pomotrelvir and nirmatrelvir compete for the same binding site. In a SARS-CoV-2 infection assay, pomotrelvir is additive when combined with remdesivir or molnupiravir, two nucleoside analogs targeting viral RNA synthesis. In conclusion, our results from the in vitro characterization of pomotrelvir antiviral activity support its further clinical development as an alternative COVID-19 therapeutic option.
Collapse
Affiliation(s)
- Xiao Tong
- Pardes Biosciences, Inc., Carlsbad, California, USA
| | - Walter Keung
- Pardes Biosciences, Inc., Carlsbad, California, USA
| | | | | | | | - Seunghyi Kook
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Uri Lopatin
- Pardes Biosciences, Inc., Carlsbad, California, USA
| | - Mark Denison
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ann D. Kwong
- Pardes Biosciences, Inc., Carlsbad, California, USA
| |
Collapse
|
11
|
Wang Q, Noettger S, Xie Q, Pastorio C, Seidel A, Müller JA, Jung C, Jacob T, Sparrer KMJ, Zech F, Kirchhoff F. Determinants of species-specific utilization of ACE2 by human and animal coronaviruses. Commun Biol 2023; 6:1051. [PMID: 37848611 PMCID: PMC10582019 DOI: 10.1038/s42003-023-05436-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 10/09/2023] [Indexed: 10/19/2023] Open
Abstract
Utilization of human ACE2 allowed several bat coronaviruses (CoVs), including the causative agent of COVID-19, to infect humans directly or via intermediate hosts. However, the determinants of species-specific differences in ACE2 usage and the frequency of the ability of animal CoVs to use human ACE2 are poorly understood. Here we applied VSV pseudoviruses to analyze the ability of Spike proteins from 26 human or animal CoVs to use ACE2 receptors across nine reservoir, potential intermediate and human hosts. We show that SARS-CoV-2 Omicron variants evolved towards more efficient ACE2 usage but mutation of R493Q in BA.4/5 and XBB Spike proteins disrupts utilization of ACE2 from Greater horseshoe bats. Variations in ACE2 residues 31, 41 and 354 govern species-specific differences in usage by coronaviral Spike proteins. Mutation of T403R allows the RaTG13 bat CoV Spike to efficiently use all ACE2 orthologs for viral entry. Sera from COVID-19 vaccinated individuals neutralize the Spike proteins of various bat Sarbecoviruses. Our results define determinants of ACE2 receptor usage of diverse CoVs and suggest that COVID-19 vaccination may protect against future zoonoses of bat coronaviruses.
Collapse
Affiliation(s)
- Qingxing Wang
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Sabrina Noettger
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Qinya Xie
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Chiara Pastorio
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Alina Seidel
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Janis A Müller
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
- Institute of Virology, Philipps University Marburg, 35043, Marburg, Germany
| | - Christoph Jung
- Institute of Electrochemistry, Ulm University, 89081, Ulm, Germany
- Helmholtz-Institute Ulm (HIU) Electrochemical Energy Storage, 89081, Ulm, Germany
- Karlsruhe Institute of Technology (KIT), 76021, Karlsruhe, Germany
| | - Timo Jacob
- Institute of Electrochemistry, Ulm University, 89081, Ulm, Germany
- Helmholtz-Institute Ulm (HIU) Electrochemical Energy Storage, 89081, Ulm, Germany
- Karlsruhe Institute of Technology (KIT), 76021, Karlsruhe, Germany
| | | | - Fabian Zech
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany.
| |
Collapse
|
12
|
Prado NDR, Brilhante-Da-Silva N, Sousa RMO, Morais MSDS, Roberto SA, Luiz MB, Assis LCD, Marinho ACM, Araujo LFLD, Pontes RDS, Stabeli RG, Fernandes CFC, Pereira SDS. Single-domain antibodies applied as antiviral immunotherapeutics. J Virol Methods 2023; 320:114787. [PMID: 37516366 DOI: 10.1016/j.jviromet.2023.114787] [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: 12/12/2022] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023]
Abstract
Viral infections have been the cause of high mortality rates throughout different periods in history. Over the last two decades, outbreaks caused by zoonotic diseases and transmitted by arboviruses have had a significant impact on human health. The emergence of viral infections in different parts of the world encourages the search for new inputs to fight pathologies of viral origin. Antibodies represent the predominant class of new drugs developed in recent years and approved for the treatment of various human diseases, including cancer, autoimmune and infectious diseases. A promising group of antibodies are single-domain antibodies derived from camelid heavy chain immunoglobulins, or VHHs, are biomolecules with nanometric dimensions and unique pharmaceutical and biophysical properties that can be used in the diagnosis and immunotherapy of viral infections. For viral neutralization to occur, VHHs can act in different stages of the viral cycle, including the actual inhibition of infection, to hindering viral replication or assembly. This review article addresses advances involving the use of VHHs in therapeutic propositions aimed to battle different viruses that affect human health.
Collapse
Affiliation(s)
- Nidiane Dantas Reis Prado
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, FIOCRUZ, unidade Rondônia, Porto Velho, RO, Brazil
| | - Nairo Brilhante-Da-Silva
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, FIOCRUZ, unidade Rondônia, Porto Velho, RO, Brazil; Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, RJ, Brazil
| | - Rosa Maria Oliveira Sousa
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, FIOCRUZ, unidade Rondônia, Porto Velho, RO, Brazil
| | | | - Sibele Andrade Roberto
- Plataforma Bi-institucional de Medicina Translacional, Fundação Oswaldo Cruz-USP, Ribeirão Preto, SP, Brazil
| | - Marcos Barros Luiz
- Instituto Federal de Rondônia Campus Guajará-Mirim, IFRO, Guajará-Mirim, RO, Brazil
| | - Livia Coelho de Assis
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, RJ, Brazil; Laboratório Multiusuário de Pesquisa e Desenvolvimento, Fundação Oswaldo Cruz, Fiocruz unidade Ceará, Eusebio, CE, Brazil
| | - Anna Carolina M Marinho
- Laboratório Multiusuário de Pesquisa e Desenvolvimento, Fundação Oswaldo Cruz, Fiocruz unidade Ceará, Eusebio, CE, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Luiz Felipe Lemes de Araujo
- Plataforma Bi-institucional de Medicina Translacional, Fundação Oswaldo Cruz-USP, Ribeirão Preto, SP, Brazil; Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade de São Paulo, USP, Ribeirão Preto, SP, Brazil
| | - Rafael de Souza Pontes
- Plataforma Bi-institucional de Medicina Translacional, Fundação Oswaldo Cruz-USP, Ribeirão Preto, SP, Brazil; Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade de São Paulo, USP, Ribeirão Preto, SP, Brazil
| | - Rodrigo Guerino Stabeli
- Plataforma Bi-institucional de Medicina Translacional, Fundação Oswaldo Cruz-USP, Ribeirão Preto, SP, Brazil
| | - Carla Freire Celedonio Fernandes
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, RJ, Brazil; Laboratório Multiusuário de Pesquisa e Desenvolvimento, Fundação Oswaldo Cruz, Fiocruz unidade Ceará, Eusebio, CE, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Soraya Dos Santos Pereira
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, FIOCRUZ, unidade Rondônia, Porto Velho, RO, Brazil; Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, RJ, Brazil; Programa de Pós-graduação em Biologia Experimental, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil.
| |
Collapse
|
13
|
Zhao Y, Bi Q, Wei Y, Wang R, Wang G, Fu G, Ran Z, Lu J, Zhang H, Zhang L, Jin R, Nie Y. A DNA vaccine (EG95-PT1/2/3-IL2) encoding multi-epitope antigen and IL-2 provokes efficient and long-term immunity to echinococcosis. J Control Release 2023; 361:402-416. [PMID: 37527761 DOI: 10.1016/j.jconrel.2023.07.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 07/20/2023] [Accepted: 07/29/2023] [Indexed: 08/03/2023]
Abstract
Echinococcosis is a highly prevalent global zoonosis, and vaccines are required. The commercial vaccine based on a protein-based subunit (EG95), however, is limited by its insufficient cellular immunity, a short protection period, and limited prevention against novel mutant strains. Herein, we applied bioinformatics to develop a DNA vaccine (pEG95-IL2) expressing both multi-epitope-based antigens (EG95-PT1/2/3) and an IL-2 adjuvant to regulate T cell differentiation and memory cell response. EG95-PT1/2/3 was screened with hierarchical structure prediction from the epitope conformation of B cells with high confidence across various species to guarantee immunogenicity. Importantly, cationic arginine-rich lipid nanoparticles (RNP) were utilized as a delivery vehicle to form lipoplexes that had a transfection efficiency of nearly two orders of magnitude greater than that of commercial reagents (Lipofectamine 2000 and polyethyleneimine) with both immune and nonimmune cells (DC2.4 and L929 cells, respectively). RNP/pEG95-IL2 lipoplexes displayed a robust and long-term antigen expression, as well as adjuvant effects during the immunization. Consequently, intramuscular injection of RNP/pEG95-IL2 elicited similar humoral immune responses and significantly greater cellular responses in mice when compared with those of the commercial vaccine. In addition, the inoculation protocol of RNP/pEG95-IL2 with sequential booster further strengthens cellular immunity in comparison with the homologous booster. Those findings provide a promising strategy for improving plasmid vaccine efficacy.
Collapse
Affiliation(s)
- Yangyang Zhao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610041, China
| | - Qunjie Bi
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610041, China
| | - Yu Wei
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610041, China
| | - Ruohan Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610041, China
| | - Gang Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610041, China
| | - Gang Fu
- Chongqing Auleon Biological Co., Ltd., Chongqing 402460, China
| | - Zhiguang Ran
- Chongqing Auleon Biological Co., Ltd., Chongqing 402460, China
| | - Jiao Lu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610041, China
| | - Heyang Zhang
- Leiden Academic Center for Drug Research (LACDR), Leiden University, Leiden 2333 CC, the Netherlands
| | - Ling Zhang
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610041, China
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610041, China.
| | - Yu Nie
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
14
|
Badr EA, Elhelbawy NG, Nagy AO, Sultan AA, Elnaidany SS. Association between cyclin-dependent kinase inhibitor 2B antisense RNA 1 and zinc finger homeobox 3 gene polymorphisms and COVID-19 severity. BMC Infect Dis 2023; 23:568. [PMID: 37653506 PMCID: PMC10472581 DOI: 10.1186/s12879-023-08564-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 08/24/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND There is no doubt about the cardiovascular complications of coronavirus disease 2019 (COVID-19). Several genetic studies have demonstrated an association between genetic variants in a region on chromosome 9p21 and in a region on chromosome 16q22 with myocardial infarction (MI) and atrial fibrillation (AF) accompanied by cerebral infarction (CI), respectively. OBJECTIVES MI and CI susceptibility in patients with CDKN2B-AS1 and ZFHX3 polymorphisms, respectively, may have an effect on COVID-19 severity. We aimed to investigate whether there is an association between the cyclin-dependent kinase inhibitor 2B antisense RNA 1 (CDKN2B-AS1) rs1333049 and zinc finger homeobox 3 (ZFHX3) rs2106261 single nucleotide polymorphisms (SNPs) and the degree of COVID-19 severity. SUBJECTS AND METHODS This current work was carried out on 360 subjects. They were classified into three groups: 90 severe COVID-19 cases, 90 moderate COVID-19 cases and 180 age- and gender-matched healthy controls. All subjects underwent genotyping of CDKN2B-AS1 (rs1333049) and ZFHX3 (rs2106261) by real-time PCR. RESULTS The frequency of G/C in CDKN2B-AS1 (rs1333049) was higher in severe and moderate COVID-19 patients than in controls (71.1% and 53.3% vs. 37.8%). The frequency of the C/C of CDKN2B-AS1 (rs1333049) was higher in moderate COVID-19 patients than in controls (26.7% vs. 13.3%). There were no significant differences regarding genotype frequency and allelic distribution of ZFHX3 (rs2106261) between COVID-19 patients and healthy controls. CONCLUSION CDKN2B-AS1 (rs1333049) gene polymorphism may play a role in determining the degree of COVID-19 severity. Further studies on its effect on cyclins and cyclin-dependent kinases (CDKs) [not measured in our study] may shed light on new treatment options for COVID-19.
Collapse
Affiliation(s)
- Eman A Badr
- Medical Biochemistry & Molecular Biology, Menofia University, Shebin- El-Kom, Egypt
| | - Nesreen G Elhelbawy
- Medical Biochemistry & Molecular Biology, Menofia University, Shebin- El-Kom, Egypt
| | - Alaa Osama Nagy
- Medical Biochemistry & Molecular Biology, Menofia University, Shebin- El-Kom, Egypt.
| | - Amany A Sultan
- Anaesthesiology, Intensive Care & Pain Management Departments, Faculty of Medicine, Menofia University, Shebin- El-Kom, Egypt
| | - Shereen S Elnaidany
- Medical Biochemistry & Molecular Biology, Menofia University, Shebin- El-Kom, Egypt
| |
Collapse
|
15
|
Leonard RA, Rao VN, Bartlett A, Froggatt HM, Luftig MA, Heaton BE, Heaton NS. A low-background, fluorescent assay to evaluate inhibitors of diverse viral proteases. J Virol 2023; 97:e0059723. [PMID: 37578235 PMCID: PMC10506478 DOI: 10.1128/jvi.00597-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/11/2023] [Indexed: 08/15/2023] Open
Abstract
Multiple coronaviruses (CoVs) can cause respiratory diseases in humans. While prophylactic vaccines designed to prevent infection are available for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), incomplete vaccine efficacy, vaccine hesitancy, and the threat of other pathogenic CoVs for which vaccines do not exist have highlighted the need for effective antiviral therapies. While antiviral compounds targeting the viral polymerase and protease are already in clinical use, their sensitivity to potential resistance mutations as well as their breadth against the full range of human and preemergent CoVs remain incompletely defined. To begin to fill that gap in knowledge, we report here the development of an improved, noninfectious, cell-based fluorescent assay with high sensitivity and low background that reports on the activity of viral proteases, which are key drug targets. We demonstrate that the assay is compatible with not only the SARS-CoV-2 Mpro protein but also orthologues from a range of human and nonhuman CoVs as well as clinically reported SARS-CoV-2 drug-resistant Mpro variants. We then use this assay to define the breadth of activity of two clinically used protease inhibitors, nirmatrelvir and ensitrelvir. Continued use of this assay will help define the strengths and limitations of current therapies and may also facilitate the development of next-generation protease inhibitors that are broadly active against both currently circulating and preemergent CoVs. IMPORTANCE Coronaviruses (CoVs) are important human pathogens with the ability to cause global pandemics. Working in concert with vaccines, antivirals specifically limit viral disease in people who are actively infected. Antiviral compounds that target CoV proteases are already in clinical use; their efficacy against variant proteases and preemergent zoonotic CoVs, however, remains incompletely defined. Here, we report an improved, noninfectious, and highly sensitive fluorescent method of defining the sensitivity of CoV proteases to small molecule inhibitors. We use this approach to assay the activity of current antiviral therapies against clinically reported SARS-CoV-2 protease mutants and a panel of highly diverse CoV proteases. Additionally, we show this system is adaptable to other structurally nonrelated viral proteases. In the future, this assay can be used to not only better define the strengths and limitations of current therapies but also help develop new, broadly acting inhibitors that more broadly target viral families.
Collapse
Affiliation(s)
- Rebecca A. Leonard
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Vishwas N. Rao
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
- Medical Scientist Training Program, Duke University School of Medicine, Durham, North Carolina, USA
| | - Alexandria Bartlett
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Heather M. Froggatt
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Micah A. Luftig
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Center for Virology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Brook E. Heaton
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Nicholas S. Heaton
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Center for Virology, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
16
|
Chattopadhyay A, Jailani AAK, Mandal B. Exigency of Plant-Based Vaccine against COVID-19 Emergence as Pandemic Preparedness. Vaccines (Basel) 2023; 11:1347. [PMID: 37631915 PMCID: PMC10458178 DOI: 10.3390/vaccines11081347] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/29/2023] Open
Abstract
After two years since the declaration of COVID-19 as a pandemic by the World Health Organization (WHO), more than six million deaths have occurred due to SARS-CoV-2, leading to an unprecedented disruption of the global economy. Fortunately, within a year, a wide range of vaccines, including pathogen-based inactivated and live-attenuated vaccines, replicating and non-replicating vector-based vaccines, nucleic acid (DNA and mRNA)-based vaccines, and protein-based subunit and virus-like particle (VLP)-based vaccines, have been developed to mitigate the severe impacts of the COVID-19 pandemic. These vaccines have proven highly effective in reducing the severity of illness and preventing deaths. However, the availability and supply of COVID-19 vaccines have become an issue due to the prioritization of vaccine distribution in most countries. Additionally, as the virus continues to mutate and spread, questions have arisen regarding the effectiveness of vaccines against new strains of SARS-CoV-2 that can evade host immunity. The urgent need for booster doses to enhance immunity has been recognized. The scarcity of "safe and effective" vaccines has exacerbated global inequalities in terms of vaccine coverage. The development of COVID-19 vaccines has fallen short of the expectations set forth in 2020 and 2021. Furthermore, the equitable distribution of vaccines at the global and national levels remains a challenge, particularly in developing countries. In such circumstances, the exigency of plant virus-based vaccines has become apparent as a means to overcome supply shortages through fast manufacturing processes and to enable quick and convenient distribution to millions of people without the reliance on a cold chain system. Moreover, plant virus-based vaccines have demonstrated both safety and efficacy in eliciting robust cellular immunogenicity against COVID-19 pathogens. This review aims to shed light on the advantages and disadvantages of different types of vaccines developed against SARS-CoV-2 and provide an update on the current status of plant-based vaccines in the fight against the COVID-19 pandemic.
Collapse
Affiliation(s)
- Anirudha Chattopadhyay
- Pulses Research Station, Sardarkrushinagar Dantiwada Agricultural University, Sardarkrushinagar 385506, India;
| | - A. Abdul Kader Jailani
- Department of Plant Pathology, North Florida Research and Education Center, University of Florida, Quincy, FL 32351, USA
| | - Bikash Mandal
- Advanced Centre for Plant Virology, Division of Plant Pathology, Indian Agricultural Research Institute, New Delhi 110012, India
| |
Collapse
|
17
|
Maity S, Santra A, Vardhan Hebbani A, Pulakuntla S, Chatterjee A, Rao Badri K, Damodara Reddy V. Targeting cytokine storm as the potential anti-viral therapy: Implications in regulating SARS-CoV-2 pathogenicity. Gene 2023:147612. [PMID: 37423400 DOI: 10.1016/j.gene.2023.147612] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/18/2023] [Accepted: 07/03/2023] [Indexed: 07/11/2023]
Abstract
The latest global pandemic corona virus disease - 2019 (COVID-19) caused by the virus SARS-CoV-2 is still a matter of worrying concern both for the scientific communities and health care organizations. COVID-19 disease is proved to be a highly contagious disease transmitted through respiratory droplets and even close contact with affected individuals. COVID-19 disease is also understood to exhibit diverse symptoms of ranging severities i.e., from mild fatigue to death. Affected individuals' susceptibility to induce immunologic dysregulation phenomena termed 'cytokine storm' seems to be playing the damaging role of escalating the disease manifestation from mild to severe. Cytokine storm in patients with severe symptoms is understood to be characterized by enhanced serum levels of many cytokines including interleukin-1β, interleukin-6, IL-10, TNF, interferon-γ, MIP-1α, MIP-1β and VEGF. Since cytokine production in general is the most important antiviral defense response, understanding the COVID-19 associated cytokine storm in particular and differentiating it from the regular cytokine production response becomes crucial in developing an effective therapeutic strategy.This review focuses on the potential targeting of COVID-19 associated cytokine storm and its challenges.
Collapse
Affiliation(s)
- Subashish Maity
- Department of Biotechnology, REVA University, Bengaluru-560064, Karnataka, India
| | - Ayantika Santra
- Department of Biochemistry, Indian Academy Degree College, Bengaluru, 560 043, India
| | | | - Swetha Pulakuntla
- Department of Biotechnology, REVA University, Bengaluru-560064, Karnataka, India
| | - Ankita Chatterjee
- Department of Biotechnology, REVA University, Bengaluru-560064, Karnataka, India
| | - Kameswara Rao Badri
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Morehouse School of Medicine, GA, Atlanta-30310, USA; Clinical Analytical Chemistry Laboratory, COVID-19 Testing Laboratory, Morehouse School of Medicine, GA, Atlanta-30310, USA.
| | - Vaddi Damodara Reddy
- Department of Biotechnology, REVA University, Bengaluru-560064, Karnataka, India.
| |
Collapse
|
18
|
Nabi AHMN, Ebihara A, Shekhar HU. Impacts of SARS-CoV-2 on diabetes mellitus: A pre and post pandemic evaluation. World J Virol 2023; 12:151-171. [PMID: 37396707 PMCID: PMC10311579 DOI: 10.5501/wjv.v12.i3.151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/24/2023] [Accepted: 04/13/2023] [Indexed: 06/21/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by the novel beta coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) crippled the whole world and has resulted in large number of morbidity and mortality. The origin of the SARS-CoV-2 is still disputed. The risk of infection with SARS-CoV-2 is dependent on several risk factors as observed in many studies. The severity of the disease depends on many factors including the viral strain, host immunogenetics, environmental factors, host genetics, host nutritional status and presence of comorbidities like hypertension, diabetes, Chronic Obstructive Pulmonary Disease, cardiovascular disease, renal impairment. Diabetes is a metabolic disorder mainly characterized by hyperglycemia. Diabetic individuals are intrinsically prone to infections. SARS-CoV-2 infection in patients with diabetes result in β-cell damage and cytokine storm. Damage to the cells impairs the equilibrium of glucose, leading to hyperglycemia. The ensuing cytokine storm causes insulin resistance, especially in the muscles and liver, which also causes a hyperglycemic state. All of these increase the severity of COVID-19. Genetics also play pivotal role in disease pathogenesis. This review article focuses from the probable sources of coronaviruses and SARS-CoV-2 to its impacts on individuals with diabetes and host genetics in pre- and post-pandemic era.
Collapse
Affiliation(s)
- A H M Nurun Nabi
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka 1000, Bangladesh
| | - Akio Ebihara
- Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193, Japan
| | - Hossain Uddin Shekhar
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka 1000, Bangladesh
| |
Collapse
|
19
|
Bekheit MS, Panda SS, Kariuki BM, Mahmoud SH, Mostafa A, Girgis AS. Spiroindole-containing compounds bearing phosphonate group of potential M pro-SARS-CoV-2 inhibitory properties. Eur J Med Chem 2023; 258:115563. [PMID: 37329713 DOI: 10.1016/j.ejmech.2023.115563] [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: 04/06/2023] [Revised: 05/23/2023] [Accepted: 06/09/2023] [Indexed: 06/19/2023]
Abstract
Microwave-assisted reaction of 3,5-bis((E)-ylidene)-1-phosphonate-4-piperidones 3a‒g with azomethine ylide (produced through interaction of isatins 4 and sarcosine 5) cycloaddition afforded the corresponding (dispiro[indoline-3,2'-pyrrolidine-3',3″-piperidin]-1″-yl)phosphonates 6a‒l in excellent yields (80-95%). Structure of the synthesized agents was evidenced by single crystal X-ray studies of 6d, 6i and 6l. Some of the synthesized agents revealed promising anti-SARS-CoV-2 properties in the viral infected Vero-E6 cell technique with noticeable selectivity indices. Compounds 6g and 6b are the most promising agents synthesized (R = 4-BrC6H4, Ph; R' = H, Cl, respectively) with considerable selectivity index values. Mpro-SARS-CoV-2 inhibitory properties supported the anti-SARS-CoV-2 observations of the potent analogs synthesized. Molecular docking studies (PDB ID: 7C8U) are consistent with the Mpro inhibitory properties. The presumed mode of action was supported by both experimentally investigated Mpro-SARS-CoV-2 inhibitory properties and explained by docking observations.
Collapse
Affiliation(s)
- Mohamed S Bekheit
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Siva S Panda
- Department of Chemistry and Physics, Augusta University, Augusta, GA, 30912, USA
| | - Benson M Kariuki
- School of Chemistry, Cardiff University, Main Building, Park Place, Cardiff, CF10 3AT, UK
| | - Sara H Mahmoud
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, 12622, Egypt
| | - Ahmed Mostafa
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, 12622, Egypt
| | - Adel S Girgis
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza, 12622, Egypt.
| |
Collapse
|
20
|
Pahlow S, Richard-Lacroix M, Hornung F, Köse-Vogel N, Mayerhöfer TG, Hniopek J, Ryabchykov O, Bocklitz T, Weber K, Ehricht R, Löffler B, Deinhardt-Emmer S, Popp J. Simple, Fast and Convenient Magnetic Bead-Based Sample Preparation for Detecting Viruses via Raman-Spectroscopy. BIOSENSORS 2023; 13:594. [PMID: 37366959 DOI: 10.3390/bios13060594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/22/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023]
Abstract
We introduce a magnetic bead-based sample preparation scheme for enabling the Raman spectroscopic differentiation of severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2)-positive and -negative samples. The beads were functionalized with the angiotensin-converting enzyme 2 (ACE2) receptor protein, which is used as a recognition element to selectively enrich SARS-CoV-2 on the surface of the magnetic beads. The subsequent Raman measurements directly enable discriminating SARS-CoV-2-positive and -negative samples. The proposed approach is also applicable for other virus species when the specific recognition element is exchanged. A series of Raman spectra were measured on three types of samples, namely SARS-CoV-2, Influenza A H1N1 virus and a negative control. For each sample type, eight independent replicates were considered. All of the spectra are dominated by the magnetic bead substrate and no obvious differences between the sample types are apparent. In order to address the subtle differences in the spectra, we calculated different correlation coefficients, namely the Pearson coefficient and the Normalized cross correlation coefficient. By comparing the correlation with the negative control, differentiating between SARS-CoV-2 and Influenza A virus is possible. This study provides a first step towards the detection and potential classification of different viruses with the use of conventional Raman spectroscopy.
Collapse
Affiliation(s)
- Susanne Pahlow
- Abbe Center of Photonics, Institute of Physical Chemistry, Friedrich Schiller University Jena, Helmholtzweg 4, 07743 Jena, Germany
- Center for Applied Research, InfectoGnostics Research Campus Jena, Philosophenweg 7, 07743 Jena, Germany
- Leibniz Centre for Photonics in Infection Research (LPI), Leibniz Institute of Photonic Technology, Albert-Einstein-Straße 9, 07745 Jena, Germany
| | - Marie Richard-Lacroix
- Leibniz Centre for Photonics in Infection Research (LPI), Leibniz Institute of Photonic Technology, Albert-Einstein-Straße 9, 07745 Jena, Germany
| | - Franziska Hornung
- Leibniz Centre for Photonics in Infection Research (LPI), Institute of Medical Microbiology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Nilay Köse-Vogel
- Leibniz Centre for Photonics in Infection Research (LPI), Institute of Medical Microbiology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Thomas G Mayerhöfer
- Abbe Center of Photonics, Institute of Physical Chemistry, Friedrich Schiller University Jena, Helmholtzweg 4, 07743 Jena, Germany
- Leibniz Centre for Photonics in Infection Research (LPI), Leibniz Institute of Photonic Technology, Albert-Einstein-Straße 9, 07745 Jena, Germany
| | - Julian Hniopek
- Abbe Center of Photonics, Institute of Physical Chemistry, Friedrich Schiller University Jena, Helmholtzweg 4, 07743 Jena, Germany
- Center for Applied Research, InfectoGnostics Research Campus Jena, Philosophenweg 7, 07743 Jena, Germany
- Leibniz Centre for Photonics in Infection Research (LPI), Leibniz Institute of Photonic Technology, Albert-Einstein-Straße 9, 07745 Jena, Germany
| | - Oleg Ryabchykov
- Abbe Center of Photonics, Institute of Physical Chemistry, Friedrich Schiller University Jena, Helmholtzweg 4, 07743 Jena, Germany
- Center for Applied Research, InfectoGnostics Research Campus Jena, Philosophenweg 7, 07743 Jena, Germany
- Leibniz Centre for Photonics in Infection Research (LPI), Leibniz Institute of Photonic Technology, Albert-Einstein-Straße 9, 07745 Jena, Germany
| | - Thomas Bocklitz
- Abbe Center of Photonics, Institute of Physical Chemistry, Friedrich Schiller University Jena, Helmholtzweg 4, 07743 Jena, Germany
- Center for Applied Research, InfectoGnostics Research Campus Jena, Philosophenweg 7, 07743 Jena, Germany
- Leibniz Centre for Photonics in Infection Research (LPI), Leibniz Institute of Photonic Technology, Albert-Einstein-Straße 9, 07745 Jena, Germany
- Physics & Computer Science, Faculty of Mathematics, Institute of Computer Science, University Bayreuth, Universitätsstraße 30, 95447 Bayreuth, Germany
| | - Karina Weber
- Abbe Center of Photonics, Institute of Physical Chemistry, Friedrich Schiller University Jena, Helmholtzweg 4, 07743 Jena, Germany
- Center for Applied Research, InfectoGnostics Research Campus Jena, Philosophenweg 7, 07743 Jena, Germany
- Leibniz Centre for Photonics in Infection Research (LPI), Leibniz Institute of Photonic Technology, Albert-Einstein-Straße 9, 07745 Jena, Germany
| | - Ralf Ehricht
- Abbe Center of Photonics, Institute of Physical Chemistry, Friedrich Schiller University Jena, Helmholtzweg 4, 07743 Jena, Germany
- Center for Applied Research, InfectoGnostics Research Campus Jena, Philosophenweg 7, 07743 Jena, Germany
- Leibniz Centre for Photonics in Infection Research (LPI), Leibniz Institute of Photonic Technology, Albert-Einstein-Straße 9, 07745 Jena, Germany
| | - Bettina Löffler
- Leibniz Centre for Photonics in Infection Research (LPI), Institute of Medical Microbiology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Stefanie Deinhardt-Emmer
- Leibniz Centre for Photonics in Infection Research (LPI), Institute of Medical Microbiology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Jürgen Popp
- Abbe Center of Photonics, Institute of Physical Chemistry, Friedrich Schiller University Jena, Helmholtzweg 4, 07743 Jena, Germany
- Center for Applied Research, InfectoGnostics Research Campus Jena, Philosophenweg 7, 07743 Jena, Germany
- Leibniz Centre for Photonics in Infection Research (LPI), Leibniz Institute of Photonic Technology, Albert-Einstein-Straße 9, 07745 Jena, Germany
| |
Collapse
|
21
|
Happi AN, Ayinla AO, Ogunsanya OA, Sijuwola AE, Saibu FM, Akano K, George UE, Sopeju AE, Rabinowitz PM, Ojo KK, Barrett LK, Van Voorhis WC, Happi CT. Detection of SARS-CoV-2 in Terrestrial Animals in Southern Nigeria: Potential Cases of Reverse Zoonosis. Viruses 2023; 15:1187. [PMID: 37243273 PMCID: PMC10222409 DOI: 10.3390/v15051187] [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: 04/17/2023] [Revised: 05/13/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Since SARS-CoV-2 caused the COVID-19 pandemic, records have suggested the occurrence of reverse zoonosis of pets and farm animals in contact with SARS-CoV-2-positive humans in the Occident. However, there is little information on the spread of the virus among animals in contact with humans in Africa. Therefore, this study aimed to investigate the occurrence of SARS-CoV-2 in various animals in Nigeria. Overall, 791 animals from Ebonyi, Ogun, Ondo, and Oyo States, Nigeria were screened for SARS-CoV-2 using RT-qPCR (n = 364) and IgG ELISA (n = 654). SARS-CoV-2 positivity rates were 45.9% (RT-qPCR) and 1.4% (ELISA). SARS-CoV-2 RNA was detected in almost all animal taxa and sampling locations except Oyo State. SARS-CoV-2 IgGs were detected only in goats from Ebonyi and pigs from Ogun States. Overall, SARS-CoV-2 infectivity rates were higher in 2021 than in 2022. Our study highlights the ability of the virus to infect various animals. It presents the first report of natural SARS-CoV-2 infection in poultry, pigs, domestic ruminants, and lizards. The close human-animal interactions in these settings suggest ongoing reverse zoonosis, highlighting the role of behavioral factors of transmission and the potential for SARS-CoV-2 to spread among animals. These underscore the importance of continuous monitoring to detect and intervene in any eventual upsurge.
Collapse
Affiliation(s)
- Anise N. Happi
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer’s University, Ede 23210, Osun State, Nigeria; (A.O.A.); (O.A.O.); (A.E.S.); (F.M.S.); (K.A.); (U.E.G.); (A.E.S.); (C.T.H.)
| | - Akeemat O. Ayinla
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer’s University, Ede 23210, Osun State, Nigeria; (A.O.A.); (O.A.O.); (A.E.S.); (F.M.S.); (K.A.); (U.E.G.); (A.E.S.); (C.T.H.)
| | - Olusola A. Ogunsanya
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer’s University, Ede 23210, Osun State, Nigeria; (A.O.A.); (O.A.O.); (A.E.S.); (F.M.S.); (K.A.); (U.E.G.); (A.E.S.); (C.T.H.)
| | - Ayotunde E. Sijuwola
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer’s University, Ede 23210, Osun State, Nigeria; (A.O.A.); (O.A.O.); (A.E.S.); (F.M.S.); (K.A.); (U.E.G.); (A.E.S.); (C.T.H.)
| | - Femi M. Saibu
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer’s University, Ede 23210, Osun State, Nigeria; (A.O.A.); (O.A.O.); (A.E.S.); (F.M.S.); (K.A.); (U.E.G.); (A.E.S.); (C.T.H.)
| | - Kazeem Akano
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer’s University, Ede 23210, Osun State, Nigeria; (A.O.A.); (O.A.O.); (A.E.S.); (F.M.S.); (K.A.); (U.E.G.); (A.E.S.); (C.T.H.)
- Department of Biological Sciences, Faculty of Natural Sciences, Redeemer’s University, Ede 23210, Osun State, Nigeria
| | - Uwem E. George
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer’s University, Ede 23210, Osun State, Nigeria; (A.O.A.); (O.A.O.); (A.E.S.); (F.M.S.); (K.A.); (U.E.G.); (A.E.S.); (C.T.H.)
- Department of Biological Sciences, Faculty of Natural Sciences, Redeemer’s University, Ede 23210, Osun State, Nigeria
| | - Adebayo E. Sopeju
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer’s University, Ede 23210, Osun State, Nigeria; (A.O.A.); (O.A.O.); (A.E.S.); (F.M.S.); (K.A.); (U.E.G.); (A.E.S.); (C.T.H.)
| | - Peter M. Rabinowitz
- Center for One Health Research, Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98109, USA;
| | - Kayode K. Ojo
- Department of Medicine, Division of Allergy and Infectious Diseases, Center for Emerging and Re-Emerging Infectious Diseases (CERID), University of Washington School of Medicine, Seattle, WA 98109, USA; (K.K.O.); (L.K.B.); (W.C.V.V.)
| | - Lynn K. Barrett
- Department of Medicine, Division of Allergy and Infectious Diseases, Center for Emerging and Re-Emerging Infectious Diseases (CERID), University of Washington School of Medicine, Seattle, WA 98109, USA; (K.K.O.); (L.K.B.); (W.C.V.V.)
| | - Wesley C. Van Voorhis
- Department of Medicine, Division of Allergy and Infectious Diseases, Center for Emerging and Re-Emerging Infectious Diseases (CERID), University of Washington School of Medicine, Seattle, WA 98109, USA; (K.K.O.); (L.K.B.); (W.C.V.V.)
| | - Christian T. Happi
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer’s University, Ede 23210, Osun State, Nigeria; (A.O.A.); (O.A.O.); (A.E.S.); (F.M.S.); (K.A.); (U.E.G.); (A.E.S.); (C.T.H.)
- Department of Biological Sciences, Faculty of Natural Sciences, Redeemer’s University, Ede 23210, Osun State, Nigeria
| |
Collapse
|
22
|
Havranek B, Lindsey GW, Higuchi Y, Itoh Y, Suzuki T, Okamoto T, Hoshino A, Procko E, Islam SM. A computationally designed ACE2 decoy has broad efficacy against SARS-CoV-2 omicron variants and related viruses in vitro and in vivo. Commun Biol 2023; 6:513. [PMID: 37173421 PMCID: PMC10177734 DOI: 10.1038/s42003-023-04860-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
SARS-CoV-2, especially B.1.1.529/omicron and its sublineages, continues to mutate to evade monoclonal antibodies and antibodies elicited by vaccination. Affinity-enhanced soluble ACE2 (sACE2) is an alternative strategy that works by binding the SARS-CoV-2 S protein, acting as a 'decoy' to block the interaction between the S and human ACE2. Using a computational design strategy, we designed an affinity-enhanced ACE2 decoy, FLIF, that exhibited tight binding to SARS-CoV-2 delta and omicron variants. Our computationally calculated absolute binding free energies (ABFE) between sACE2:SARS-CoV-2 S proteins and their variants showed excellent agreement to binding experiments. FLIF displayed robust therapeutic utility against a broad range of SARS-CoV-2 variants and sarbecoviruses, and neutralized omicron BA.5 in vitro and in vivo. Furthermore, we directly compared the in vivo therapeutic efficacy of wild-type ACE2 (non-affinity enhanced ACE2) against FLIF. A few wild-type sACE2 decoys have shown to be effective against early circulating variants such as Wuhan in vivo. Our data suggest that moving forward, affinity-enhanced ACE2 decoys like FLIF may be required to combat evolving SARS-CoV-2 variants. The approach described herein emphasizes how computational methods have become sufficiently accurate for the design of therapeutics against viral protein targets. Affinity-enhanced ACE2 decoys remain highly effective at neutralizing omicron subvariants.
Collapse
Affiliation(s)
- Brandon Havranek
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, 60607, USA
- Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, 19107, USA
- ComputePharma, LLC., Chicago, IL, USA
| | | | - Yusuke Higuchi
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Yumi Itoh
- Institute for Advanced Co-Creation Studies, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Tatsuya Suzuki
- Institute for Advanced Co-Creation Studies, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Toru Okamoto
- Institute for Advanced Co-Creation Studies, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Atsushi Hoshino
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Erik Procko
- Department of Biochemistry, University of Illinois, Urbana, IL, 61801, USA
- Cyrus Biotechnology, Inc., Seattle, WA, USA
| | - Shahidul M Islam
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, 60607, USA.
- ComputePharma, LLC., Chicago, IL, USA.
- Department of Chemistry, Delaware State University, Dover, DE, 19901, USA.
| |
Collapse
|
23
|
Zvornicanin SN, Shaqra AM, Huang QJ, Ornelas E, Moghe M, Knapp M, Moquin S, Dovala D, Schiffer CA, Kurt Yilmaz N. Crystal Structures of Inhibitor-Bound Main Protease from Delta- and Gamma-Coronaviruses. Viruses 2023; 15:781. [PMID: 36992489 PMCID: PMC10059799 DOI: 10.3390/v15030781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/31/2023] Open
Abstract
With the spread of SARS-CoV-2 throughout the globe causing the COVID-19 pandemic, the threat of zoonotic transmissions of coronaviruses (CoV) has become even more evident. As human infections have been caused by alpha- and beta-CoVs, structural characterization and inhibitor design mostly focused on these two genera. However, viruses from the delta and gamma genera also infect mammals and pose a potential zoonotic transmission threat. Here, we determined the inhibitor-bound crystal structures of the main protease (Mpro) from the delta-CoV porcine HKU15 and gamma-CoV SW1 from the beluga whale. A comparison with the apo structure of SW1 Mpro, which is also presented here, enabled the identification of structural arrangements upon inhibitor binding at the active site. The cocrystal structures reveal binding modes and interactions of two covalent inhibitors, PF-00835231 (active form of lufotrelvir) bound to HKU15, and GC376 bound to SW1 Mpro. These structures may be leveraged to target diverse coronaviruses and toward the structure-based design of pan-CoV inhibitors.
Collapse
Affiliation(s)
- Sarah N. Zvornicanin
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Ala M. Shaqra
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Qiuyu J. Huang
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Elizabeth Ornelas
- Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA
| | - Mallika Moghe
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Mark Knapp
- Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA
| | - Stephanie Moquin
- Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA
| | - Dustin Dovala
- Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA
| | - Celia A. Schiffer
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Nese Kurt Yilmaz
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
24
|
Roa-Linares VC, Escudero-Flórez M, Vicente-Manzanares M, Gallego-Gómez JC. Host Cell Targets for Unconventional Antivirals against RNA Viruses. Viruses 2023; 15:v15030776. [PMID: 36992484 PMCID: PMC10058429 DOI: 10.3390/v15030776] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/12/2023] [Accepted: 02/28/2023] [Indexed: 03/31/2023] Open
Abstract
The recent COVID-19 crisis has highlighted the importance of RNA-based viruses. The most prominent members of this group are SARS-CoV-2 (coronavirus), HIV (human immunodeficiency virus), EBOV (Ebola virus), DENV (dengue virus), HCV (hepatitis C virus), ZIKV (Zika virus), CHIKV (chikungunya virus), and influenza A virus. With the exception of retroviruses which produce reverse transcriptase, the majority of RNA viruses encode RNA-dependent RNA polymerases which do not include molecular proofreading tools, underlying the high mutation capacity of these viruses as they multiply in the host cells. Together with their ability to manipulate the immune system of the host in different ways, their high mutation frequency poses a challenge to develop effective and durable vaccination and/or treatments. Consequently, the use of antiviral targeting agents, while an important part of the therapeutic strategy against infection, may lead to the selection of drug-resistant variants. The crucial role of the host cell replicative and processing machinery is essential for the replicative cycle of the viruses and has driven attention to the potential use of drugs directed to the host machinery as therapeutic alternatives to treat viral infections. In this review, we discuss small molecules with antiviral effects that target cellular factors in different steps of the infectious cycle of many RNA viruses. We emphasize the repurposing of FDA-approved drugs with broad-spectrum antiviral activity. Finally, we postulate that the ferruginol analog (18-(phthalimide-2-yl) ferruginol) is a potential host-targeted antiviral.
Collapse
Affiliation(s)
- Vicky C Roa-Linares
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia
| | - Manuela Escudero-Flórez
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia
| | - Miguel Vicente-Manzanares
- Molecular Mechanisms Program, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, 37007 Salamanca, Spain
| | - Juan C Gallego-Gómez
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia
| |
Collapse
|
25
|
Interactions between Humans and Dogs during the COVID-19 Pandemic: Recent Updates and Future Perspectives. Animals (Basel) 2023; 13:ani13030524. [PMID: 36766413 PMCID: PMC9913536 DOI: 10.3390/ani13030524] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
COVID-19 is one of the deadliest epidemics. This pandemic is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), but the role of dogs in spreading the disease in human society is poorly understood. This review sheds light on the limited susceptibility of dogs to COVID-19 infections which is likely attributed to the relatively low levels of angiotensin-converting enzyme 2 (ACE2) in the respiratory tract and the phylogenetic distance of ACE2 in dogs from the human ACE2 receptor. The low levels of ACE2 affect the binding affinity between spike and ACE2 proteins resulting in it being uncommon for dogs to spread the disease. To demonstrate the role of dogs in spreading COVID-19, we reviewed the epidemiological studies and prevalence of SARS-CoV-2 in dogs. Additionally, we discussed the use of detection dogs as a rapid and reliable method for effectively discriminating between SARS-CoV-2 infected and non-infected individuals using different types of samples (secretions, saliva, and sweat). We considered the available information on COVID-19 in the human-dog interfaces involving the possibility of transmission of COVID-19 to dogs by infected individuals and vice versa, the human-dog behavior changes, and the importance of preventive measures because the risk of transmission by domestic dogs remains a concern.
Collapse
|
26
|
Sah R, Mohapatra RK, Mishra S, Chinnam S, Rabaan AA, Alshahrani NZ, Mohanty A, Al-Ahdal T, León-Figueroa DA, Padhi BK. Cocktail of FIFA 2022 Vis-A-Vis camel beauty pageant championship; potential health threat of MERS among players and fans - A possible global spread. Travel Med Infect Dis 2023; 52:102541. [PMID: 36623743 PMCID: PMC9817334 DOI: 10.1016/j.tmaid.2023.102541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/02/2023] [Accepted: 01/05/2023] [Indexed: 01/09/2023]
Affiliation(s)
- Ranjit Sah
- Tribhuvan University Teaching Hospital, Institute of Medicine, Kathmandu, Nepal; Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India; Department of Chemistry, Government College of Engineering, Keonjhar, 758 002, Odisha, India; School of Biotechnology, Campus-11, KIIT Deemed-to-be-University, Bhubaneswar, Odisha, 751024, India; Department of Chemistry, M.S Ramaiah Institute of Technology (Affiliated to Visvesvaraya Technological University, Belgam), Bengaluru, Karnataka, 560054, India; Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, 31311, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, 11533, Saudi Arabia; Department of Public Health and Nutrition, The University of Haripur, Haripur, 22610, Pakistan; Faculty of Medicine, University of Jeddah, Jeddah, 21589, Saudi Arabia; Department of Microbiology, All India Institute of Medical Sciences, Gorakhpur, India; Institute of Global Health, Heidelberg University, Neuenheimer feld 130/3, 69120, Heidelberg, Germany; Facultad de Medicina Humana, Universidad de San Martin de Porres, Chiclayo, Peru; Department of Community Medicine and School of Public Health, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - Ranjan K Mohapatra
- Tribhuvan University Teaching Hospital, Institute of Medicine, Kathmandu, Nepal; Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India; Department of Chemistry, Government College of Engineering, Keonjhar, 758 002, Odisha, India; School of Biotechnology, Campus-11, KIIT Deemed-to-be-University, Bhubaneswar, Odisha, 751024, India; Department of Chemistry, M.S Ramaiah Institute of Technology (Affiliated to Visvesvaraya Technological University, Belgam), Bengaluru, Karnataka, 560054, India; Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, 31311, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, 11533, Saudi Arabia; Department of Public Health and Nutrition, The University of Haripur, Haripur, 22610, Pakistan; Faculty of Medicine, University of Jeddah, Jeddah, 21589, Saudi Arabia; Department of Microbiology, All India Institute of Medical Sciences, Gorakhpur, India; Institute of Global Health, Heidelberg University, Neuenheimer feld 130/3, 69120, Heidelberg, Germany; Facultad de Medicina Humana, Universidad de San Martin de Porres, Chiclayo, Peru; Department of Community Medicine and School of Public Health, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Snehasish Mishra
- Tribhuvan University Teaching Hospital, Institute of Medicine, Kathmandu, Nepal; Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India; Department of Chemistry, Government College of Engineering, Keonjhar, 758 002, Odisha, India; School of Biotechnology, Campus-11, KIIT Deemed-to-be-University, Bhubaneswar, Odisha, 751024, India; Department of Chemistry, M.S Ramaiah Institute of Technology (Affiliated to Visvesvaraya Technological University, Belgam), Bengaluru, Karnataka, 560054, India; Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, 31311, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, 11533, Saudi Arabia; Department of Public Health and Nutrition, The University of Haripur, Haripur, 22610, Pakistan; Faculty of Medicine, University of Jeddah, Jeddah, 21589, Saudi Arabia; Department of Microbiology, All India Institute of Medical Sciences, Gorakhpur, India; Institute of Global Health, Heidelberg University, Neuenheimer feld 130/3, 69120, Heidelberg, Germany; Facultad de Medicina Humana, Universidad de San Martin de Porres, Chiclayo, Peru; Department of Community Medicine and School of Public Health, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sampath Chinnam
- Tribhuvan University Teaching Hospital, Institute of Medicine, Kathmandu, Nepal; Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India; Department of Chemistry, Government College of Engineering, Keonjhar, 758 002, Odisha, India; School of Biotechnology, Campus-11, KIIT Deemed-to-be-University, Bhubaneswar, Odisha, 751024, India; Department of Chemistry, M.S Ramaiah Institute of Technology (Affiliated to Visvesvaraya Technological University, Belgam), Bengaluru, Karnataka, 560054, India; Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, 31311, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, 11533, Saudi Arabia; Department of Public Health and Nutrition, The University of Haripur, Haripur, 22610, Pakistan; Faculty of Medicine, University of Jeddah, Jeddah, 21589, Saudi Arabia; Department of Microbiology, All India Institute of Medical Sciences, Gorakhpur, India; Institute of Global Health, Heidelberg University, Neuenheimer feld 130/3, 69120, Heidelberg, Germany; Facultad de Medicina Humana, Universidad de San Martin de Porres, Chiclayo, Peru; Department of Community Medicine and School of Public Health, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ali A Rabaan
- Tribhuvan University Teaching Hospital, Institute of Medicine, Kathmandu, Nepal; Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India; Department of Chemistry, Government College of Engineering, Keonjhar, 758 002, Odisha, India; School of Biotechnology, Campus-11, KIIT Deemed-to-be-University, Bhubaneswar, Odisha, 751024, India; Department of Chemistry, M.S Ramaiah Institute of Technology (Affiliated to Visvesvaraya Technological University, Belgam), Bengaluru, Karnataka, 560054, India; Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, 31311, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, 11533, Saudi Arabia; Department of Public Health and Nutrition, The University of Haripur, Haripur, 22610, Pakistan; Faculty of Medicine, University of Jeddah, Jeddah, 21589, Saudi Arabia; Department of Microbiology, All India Institute of Medical Sciences, Gorakhpur, India; Institute of Global Health, Heidelberg University, Neuenheimer feld 130/3, 69120, Heidelberg, Germany; Facultad de Medicina Humana, Universidad de San Martin de Porres, Chiclayo, Peru; Department of Community Medicine and School of Public Health, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Najim Z Alshahrani
- Tribhuvan University Teaching Hospital, Institute of Medicine, Kathmandu, Nepal; Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India; Department of Chemistry, Government College of Engineering, Keonjhar, 758 002, Odisha, India; School of Biotechnology, Campus-11, KIIT Deemed-to-be-University, Bhubaneswar, Odisha, 751024, India; Department of Chemistry, M.S Ramaiah Institute of Technology (Affiliated to Visvesvaraya Technological University, Belgam), Bengaluru, Karnataka, 560054, India; Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, 31311, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, 11533, Saudi Arabia; Department of Public Health and Nutrition, The University of Haripur, Haripur, 22610, Pakistan; Faculty of Medicine, University of Jeddah, Jeddah, 21589, Saudi Arabia; Department of Microbiology, All India Institute of Medical Sciences, Gorakhpur, India; Institute of Global Health, Heidelberg University, Neuenheimer feld 130/3, 69120, Heidelberg, Germany; Facultad de Medicina Humana, Universidad de San Martin de Porres, Chiclayo, Peru; Department of Community Medicine and School of Public Health, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Aroop Mohanty
- Tribhuvan University Teaching Hospital, Institute of Medicine, Kathmandu, Nepal; Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India; Department of Chemistry, Government College of Engineering, Keonjhar, 758 002, Odisha, India; School of Biotechnology, Campus-11, KIIT Deemed-to-be-University, Bhubaneswar, Odisha, 751024, India; Department of Chemistry, M.S Ramaiah Institute of Technology (Affiliated to Visvesvaraya Technological University, Belgam), Bengaluru, Karnataka, 560054, India; Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, 31311, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, 11533, Saudi Arabia; Department of Public Health and Nutrition, The University of Haripur, Haripur, 22610, Pakistan; Faculty of Medicine, University of Jeddah, Jeddah, 21589, Saudi Arabia; Department of Microbiology, All India Institute of Medical Sciences, Gorakhpur, India; Institute of Global Health, Heidelberg University, Neuenheimer feld 130/3, 69120, Heidelberg, Germany; Facultad de Medicina Humana, Universidad de San Martin de Porres, Chiclayo, Peru; Department of Community Medicine and School of Public Health, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Tareq Al-Ahdal
- Tribhuvan University Teaching Hospital, Institute of Medicine, Kathmandu, Nepal; Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India; Department of Chemistry, Government College of Engineering, Keonjhar, 758 002, Odisha, India; School of Biotechnology, Campus-11, KIIT Deemed-to-be-University, Bhubaneswar, Odisha, 751024, India; Department of Chemistry, M.S Ramaiah Institute of Technology (Affiliated to Visvesvaraya Technological University, Belgam), Bengaluru, Karnataka, 560054, India; Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, 31311, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, 11533, Saudi Arabia; Department of Public Health and Nutrition, The University of Haripur, Haripur, 22610, Pakistan; Faculty of Medicine, University of Jeddah, Jeddah, 21589, Saudi Arabia; Department of Microbiology, All India Institute of Medical Sciences, Gorakhpur, India; Institute of Global Health, Heidelberg University, Neuenheimer feld 130/3, 69120, Heidelberg, Germany; Facultad de Medicina Humana, Universidad de San Martin de Porres, Chiclayo, Peru; Department of Community Medicine and School of Public Health, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Darwin A León-Figueroa
- Tribhuvan University Teaching Hospital, Institute of Medicine, Kathmandu, Nepal; Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India; Department of Chemistry, Government College of Engineering, Keonjhar, 758 002, Odisha, India; School of Biotechnology, Campus-11, KIIT Deemed-to-be-University, Bhubaneswar, Odisha, 751024, India; Department of Chemistry, M.S Ramaiah Institute of Technology (Affiliated to Visvesvaraya Technological University, Belgam), Bengaluru, Karnataka, 560054, India; Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, 31311, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, 11533, Saudi Arabia; Department of Public Health and Nutrition, The University of Haripur, Haripur, 22610, Pakistan; Faculty of Medicine, University of Jeddah, Jeddah, 21589, Saudi Arabia; Department of Microbiology, All India Institute of Medical Sciences, Gorakhpur, India; Institute of Global Health, Heidelberg University, Neuenheimer feld 130/3, 69120, Heidelberg, Germany; Facultad de Medicina Humana, Universidad de San Martin de Porres, Chiclayo, Peru; Department of Community Medicine and School of Public Health, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Bijaya K Padhi
- Tribhuvan University Teaching Hospital, Institute of Medicine, Kathmandu, Nepal; Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India; Department of Chemistry, Government College of Engineering, Keonjhar, 758 002, Odisha, India; School of Biotechnology, Campus-11, KIIT Deemed-to-be-University, Bhubaneswar, Odisha, 751024, India; Department of Chemistry, M.S Ramaiah Institute of Technology (Affiliated to Visvesvaraya Technological University, Belgam), Bengaluru, Karnataka, 560054, India; Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, 31311, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, 11533, Saudi Arabia; Department of Public Health and Nutrition, The University of Haripur, Haripur, 22610, Pakistan; Faculty of Medicine, University of Jeddah, Jeddah, 21589, Saudi Arabia; Department of Microbiology, All India Institute of Medical Sciences, Gorakhpur, India; Institute of Global Health, Heidelberg University, Neuenheimer feld 130/3, 69120, Heidelberg, Germany; Facultad de Medicina Humana, Universidad de San Martin de Porres, Chiclayo, Peru; Department of Community Medicine and School of Public Health, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
27
|
Mubareka S, Amuasi J, Banerjee A, Carabin H, Copper Jack J, Jardine C, Jaroszewicz B, Keefe G, Kotwa J, Kutz S, McGregor D, Mease A, Nicholson L, Nowak K, Pickering B, Reed MG, Saint-Charles J, Simonienko K, Smith T, Scott Weese J, Jane Parmley E. Strengthening a One Health approach to emerging zoonoses. Facets (Ott) 2023. [DOI: 10.1139/facets-2021-0190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Given the enormous global impact of the COVID-19 pandemic, outbreaks of highly pathogenic avian influenza in Canada, and manifold other zoonotic pathogen activity, there is a pressing need for a deeper understanding of the human-animal-environment interface and the intersecting biological, ecological, and societal factors contributing to the emergence, spread, and impact of zoonotic diseases. We aim to apply a One Health approach to pressing issues related to emerging zoonoses, and propose a functional framework of interconnected but distinct groups of recommendations around strategy and governance, technical leadership (operations), equity, education and research for a One Health approach and Action Plan for Canada. Change is desperately needed, beginning by reorienting our approach to health and recalibrating our perspectives to restore balance with the natural world in a rapid and sustainable fashion. In Canada, a major paradigm shift in how we think about health is required. All of society must recognize the intrinsic value of all living species and the importance of the health of humans, other animals, and ecosystems to health for all.
Collapse
Affiliation(s)
| | - John Amuasi
- Kwame Nkrumah University of Science and Technology, Kumasi, Ashanti Region, Ghana
| | | | | | - Joe Copper Jack
- Indigenous Knowledge Holder, Whitehorse, Yukon Territory, Canada
| | | | | | - Greg Keefe
- University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada
| | | | - Susan Kutz
- University of Calgary, Calgary, Alberta, Canada
| | | | - Anne Mease
- Selkirk First Nation Citizen, Selkirk First Nation, Yukon Territory, Canada
| | | | | | - Brad Pickering
- Canadian Food Inspection Agency, Winnipeg, Manitoba, Canada
| | | | | | | | | | | | | |
Collapse
|
28
|
Kumar M, Roy A, Rawat RS, Alok A, Tetala KKR, Biswas NR, Kaur P, Kumar S. Identification and structural studies of natural inhibitors against SARS-CoV-2 viral RNA methyltransferase (NSP16). J Biomol Struct Dyn 2022; 40:13965-13975. [PMID: 34766876 DOI: 10.1080/07391102.2021.1997821] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Pathogenic RNA viruses are emerging as one of the major threats and posing challenges to human community. RNA viruses have an exceptionally shorter generation time and easy to adapt in host cells. The recent emergence of SARS-CoV-2, a long RNA virus, has shown us how difficult it is to overcome this kind of pandemic without understanding the viral infection and replication mechanisms. It is essential to comprehend replications of the viral genome, including RNA polymerization and the final capping process. The mRNAs of SARS-CoV-2 coronaviruses are protected at their 5'-ends by cap structure. The cap-like system plays a significant role in viral translational process, viral RNA stability, and scatting in detecting innate immune recognition in host cells. Two coronavirus enzymes, Nsp14 and Nsp16, critically help in the formation of capping and are considered as potential drug targets for antiviral therapy. Natural and herbal medicines have a past record of treating various acute respiratory diseases. In this work, we have exploited 56000 natural compounds to screen potential inhibitors against NSP16. In silico virtual screening, docking and Molecular Dynamics (MD) simulation studies were performed to understand how these potential inhibitors are bound to NSP16. We observed that the most highly screened compound binds to protein molecules with a high dock score, primarily through hydrophobic interactions and hydrogen bonding, as previously reported for NSP16. Compound-13 (2-hydroxy-N-({1-[2-hydroxy-1-(hydroxymethyl)ethyl]piperidin-3-yl}methyl)-5-methylbenzamide) and compound-51 (N-(2-isobutoxybenzyl)-N,2-dimethyl-2,8-diazaspiro[4.5]decane-3-carboxamide) occupied in active site along with good pharmokinetices properties. In conclusion, the selected compounds could be used as a novel therapeutic against SARS-CoV-2.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mukesh Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Anik Roy
- Centre for Bioseparation Technology (CBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Ravindra Singh Rawat
- Centre for Bioseparation Technology (CBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Amit Alok
- Division of Radiation Biodosimetry, Institute of Nuclear Medicine & Allied Sciences, Timarpur, Delhi, India
| | - Kishore K R Tetala
- Centre for Bioseparation Technology (CBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Nihar Ranjan Biswas
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India.,Department of Pharmacology, Indira Gandhi Institute of Medical Science (IGIMS), Patna, India
| | - Punit Kaur
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Sanjit Kumar
- Centre for Bioseparation Technology (CBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| |
Collapse
|
29
|
Balasubramaniyam A, Ryan E, Brown D, Hamza T, Harrison W, Gan M, Sankhala RS, Chen WH, Martinez EJ, Jensen JL, Dussupt V, Mendez-Rivera L, Mayer S, King J, Michael NL, Regules J, Krebs S, Rao M, Matyas GR, Joyce MG, Batchelor AH, Gromowski GD, Dutta S. Unglycosylated Soluble SARS-CoV-2 Receptor Binding Domain (RBD) Produced in E. coli Combined with the Army Liposomal Formulation Containing QS21 (ALFQ) Elicits Neutralizing Antibodies against Mismatched Variants. Vaccines (Basel) 2022; 11:vaccines11010042. [PMID: 36679887 PMCID: PMC9864931 DOI: 10.3390/vaccines11010042] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/18/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022] Open
Abstract
The emergence of novel potentially pandemic pathogens necessitates the rapid manufacture and deployment of effective, stable, and locally manufacturable vaccines on a global scale. In this study, the ability of the Escherichia coli expression system to produce the receptor binding domain (RBD) of the SARS-CoV-2 spike protein was evaluated. The RBD of the original Wuhan-Hu1 variant and of the Alpha and Beta variants of concern (VoC) were expressed in E. coli, and their biochemical and immunological profiles were compared to RBD produced in mammalian cells. The E. coli-produced RBD variants recapitulated the structural character of mammalian-expressed RBD and bound to human angiotensin converting enzyme (ACE2) receptor and a panel of neutralizing SARS-CoV-2 monoclonal antibodies. A pilot vaccination in mice with bacterial RBDs formulated with a novel liposomal adjuvant, Army Liposomal Formulation containing QS21 (ALFQ), induced polyclonal antibodies that inhibited RBD association to ACE2 in vitro and potently neutralized homologous and heterologous SARS-CoV-2 pseudoviruses. Although all vaccines induced neutralization of the non-vaccine Delta variant, only the Beta RBD vaccine produced in E. coli and mammalian cells effectively neutralized the Omicron BA.1 pseudovirus. These outcomes warrant further exploration of E. coli as an expression platform for non-glycosylated, soluble immunogens for future rapid response to emerging pandemic pathogens.
Collapse
Affiliation(s)
- Arasu Balasubramaniyam
- Biologics Research and Development Branch, Structural Vaccinology Laboratory, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Emma Ryan
- Biologics Research and Development Branch, Structural Vaccinology Laboratory, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Dallas Brown
- Biologics Research and Development Branch, Structural Vaccinology Laboratory, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Therwa Hamza
- Biologics Research and Development Branch, Structural Vaccinology Laboratory, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - William Harrison
- Biologics Research and Development Branch, Structural Vaccinology Laboratory, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Michael Gan
- Biologics Research and Development Branch, Structural Vaccinology Laboratory, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Rajeshwer S. Sankhala
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Wei-Hung Chen
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Elizabeth J. Martinez
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Jaime L. Jensen
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Vincent Dussupt
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
- U.S. Military HIV Research Program, B-cell Biology Laboratory, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Letzibeth Mendez-Rivera
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
- U.S. Military HIV Research Program, B-cell Biology Laboratory, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Sandra Mayer
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Jocelyn King
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Nelson L. Michael
- Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Jason Regules
- Biologics Research and Development Branch, Structural Vaccinology Laboratory, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Shelly Krebs
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
- U.S. Military HIV Research Program, B-cell Biology Laboratory, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Mangala Rao
- U.S. Military HIV Research Program, Laboratory of Adjuvant and Antigen Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Gary R. Matyas
- U.S. Military HIV Research Program, Laboratory of Adjuvant and Antigen Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - M. Gordon Joyce
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Adrian H. Batchelor
- Biologics Research and Development Branch, Structural Vaccinology Laboratory, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Gregory D. Gromowski
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Sheetij Dutta
- Biologics Research and Development Branch, Structural Vaccinology Laboratory, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Correspondence: ; Tel.: +1-301-319-9154
| |
Collapse
|
30
|
Thakor JC, Dinesh M, Manikandan R, Bindu S, Sahoo M, Sahoo D, Dhawan M, Pandey MK, Tiwari R, Emran TB, Dhama K, Chaicumpa W. Swine coronaviruses (SCoVs) and their emerging threats to swine population, inter-species transmission, exploring the susceptibility of pigs for SARS-CoV-2 and zoonotic concerns. Vet Q 2022; 42:125-147. [PMID: 35584308 PMCID: PMC9225692 DOI: 10.1080/01652176.2022.2079756] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 03/23/2022] [Accepted: 05/16/2022] [Indexed: 12/23/2022] Open
Abstract
Swine coronaviruses (SCoVs) are one of the most devastating pathogens affecting the livelihoods of farmers and swine industry across the world. These include transmissible gastroenteritis virus (TGEV), porcine epidemic diarrhea virus (PEDV), porcine respiratory coronavirus (PRCV), porcine hemagglutinating encephalomyelitis virus (PHEV), swine acute diarrhea syndrome coronavirus (SADS-CoV), and porcine delta coronavirus (PDCoV). Coronaviruses infect a wide variety of animal species and humans because these are having single stranded-RNA that accounts for high mutation rates and thus could break the species barrier. The gastrointestinal, cardiovascular, and nervous systems are the primary organ systems affected by SCoVs. Infection is very common in piglets compared to adult swine causing high mortality in the former. Bat is implicated to be the origin of all CoVs affecting animals and humans. Since pig is the only domestic animal in which CoVs cause a wide range of diseases; new coronaviruses with high zoonotic potential could likely emerge in the future as observed in the past. The recently emerged severe acute respiratory syndrome coronavirus virus-2 (SARS-CoV-2), causing COVID-19 pandemic in humans, has been implicated to have animal origin, also reported from few animal species, though its zoonotic concerns are still under investigation. This review discusses SCoVs and their epidemiology, virology, evolution, pathology, wildlife reservoirs, interspecies transmission, spill-over events and highlighting their emerging threats to swine population. The role of pigs amid ongoing SARS-CoV-2 pandemic will also be discussed. A thorough investigation should be conducted to rule out zoonotic potential of SCoVs and to design appropriate strategies for their prevention and control.
Collapse
Affiliation(s)
- Jigarji C. Thakor
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Murali Dinesh
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Rajendran Manikandan
- Immunology Section, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Suresh Bindu
- Immunology Section, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Monalisa Sahoo
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Diptimayee Sahoo
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana, India
- The Trafford Group of Colleges, Manchester, United Kingdom
| | - Megha Katare Pandey
- Department of Translational Medicine Center, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, Uttar Pradesh Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan (DUVASU), Mathura, India
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Wanpen Chaicumpa
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
31
|
Molecular Dynamics Simulations to Decipher the Role of Phosphorylation of SARS-CoV-2 Nonstructural Proteins (nsps) in Viral Replication. Viruses 2022; 14:v14112436. [PMID: 36366534 PMCID: PMC9693435 DOI: 10.3390/v14112436] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/19/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
Protein phosphorylation is a post-translational modification that enables various cellular activities and plays essential roles in protein interactions. Phosphorylation is an important process for the replication of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). To shed more light on the effects of phosphorylation, we used an ensemble of neural networks to predict potential kinases that might phosphorylate SARS-CoV-2 nonstructural proteins (nsps) and molecular dynamics (MD) simulations to investigate the effects of phosphorylation on nsps structure, which could be a potential inhibitory target to attenuate viral replication. Eight target candidate sites were found as top-ranked phosphorylation sites of SARS-CoV-2. During the process of molecular dynamics (MD) simulation, the root-mean-square deviation (RMSD) analysis was used to measure conformational changes in each nsps. Root-mean-square fluctuation (RMSF) was employed to measure the fluctuation in each residue of 36 systems considered, allowing us to evaluate the most flexible regions. These analysis shows that there are significant structural deviations in the residues namely nsp1 THR 72, nsp2 THR 73, nsp3 SER 64, nsp4 SER 81, nsp4 SER 455, nsp5 SER284, nsp6 THR 238, and nsp16 SER 132. The identified list of residues suggests how phosphorylation affects SARS-CoV-2 nsps function and stability. This research also suggests that kinase inhibitors could be a possible component for evaluating drug binding studies, which are crucial in therapeutic discovery research.
Collapse
|
32
|
García-Machorro J, Ramírez-Salinas GL, Martinez-Archundia M, Correa-Basurto J. The Advantage of Using Immunoinformatic Tools on Vaccine Design and Development for Coronavirus. Vaccines (Basel) 2022; 10:1844. [PMID: 36366353 PMCID: PMC9693616 DOI: 10.3390/vaccines10111844] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 10/21/2022] [Accepted: 10/27/2022] [Indexed: 10/28/2023] Open
Abstract
After the outbreak of SARS-CoV-2 by the end of 2019, the vaccine development strategies became a worldwide priority. Furthermore, the appearances of novel SARS-CoV-2 variants challenge researchers to develop new pharmacological or preventive strategies. However, vaccines still represent an efficient way to control the SARS-CoV-2 pandemic worldwide. This review describes the importance of bioinformatic and immunoinformatic tools (in silico) for guide vaccine design. In silico strategies permit the identification of epitopes (immunogenic peptides) which could be used as potential vaccines, as well as nonacarriers such as: vector viral based vaccines, RNA-based vaccines and dendrimers through immunoinformatics. Currently, nucleic acid and protein sequential as well structural analyses through bioinformatic tools allow us to get immunogenic epitopes which can induce immune response alone or in complex with nanocarriers. One of the advantages of in silico techniques is that they facilitate the identification of epitopes, while accelerating the process and helping to economize some stages of the development of safe vaccines.
Collapse
Affiliation(s)
- Jazmín García-Machorro
- Laboratorio de Medicina de Conservación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Gema Lizbeth Ramírez-Salinas
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotécnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, México City 11340, Mexico
| | - Marlet Martinez-Archundia
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotécnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, México City 11340, Mexico
| | - José Correa-Basurto
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotécnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, México City 11340, Mexico
| |
Collapse
|
33
|
Islam A, Haider F, Ahmmed Aoni R, Ahmed R. Plasmonic photonic biosensor: in situ detection and quantification of SARS-CoV-2 particles. OPTICS EXPRESS 2022; 30:40277-40291. [PMID: 36298963 DOI: 10.1364/oe.469937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/03/2022] [Indexed: 06/16/2023]
Abstract
We conceptualized and numerically investigated a photonic crystal fiber (PCF)-based surface plasmon resonance (SPR) sensor for rapid detection and quantification of novel coronavirus. The plasmonic gold-based optical sensor permits three different ways to quantify the virus concentrations inside patient's body based on different ligand-analyte conjugate pairs. This photonic biosensor demonstrates viable detections of SARS-CoV-2 spike receptor-binding-domain (RBD), mutated viral single-stranded ribonucleic acid (RNA) and human monoclonal antibody immunoglobulin G (IgG). A marquise-shaped core is introduced to facilitate efficient light-tailoring. Analytes are dissolved in sterile phosphate buffered saline (PBS) and surfaced on the plasmonic metal layer for realizing detection. The 1-pyrene butyric acid n-hydroxy-succinimide ester is numerically used to immobilize the analytes on the sensing interface. Using the finite element method (FEM), the proposed sensor is studied critically and optimized for the refractive index (RI) range from 1.3348-1.3576, since the target analytes RIs fluctuate within this range depending on the severity of the viral infection. The polarization-dependent sensor exhibits dominant sensing attributes for x-polarized mode, where it shows the average wavelength sensitivities of 2,009 nm/RIU, 2,745 nm/RIU and 1,984 nm/RIU for analytes: spike RBD, extracted coronavirus RNA and antibody IgG, respectively. The corresponding median amplitude sensitivities are 135 RIU-1, 196 RIU-1 and 140 RIU-1, respectively. The maximum sensor resolution and figure of merit are found 2.53 × 10-5 RIU and 101 RIU-1, respectively for viral RNA detection. Also, a significant limit of detection (LOD) of 6.42 × 10-9 RIU2/nm is obtained. Considering modern bioassays, the proposed compact photonic sensor will be well-suited for rapid point-of-care COVID testing.
Collapse
|
34
|
Singh A, Singh P, Kumar R, Kaushik A. Exploring nanoselenium to tackle mutated SARS-CoV-2 for efficient COVID-19 management. FRONTIERS IN NANOTECHNOLOGY 2022. [DOI: 10.3389/fnano.2022.1004729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Despite ongoing public health measures and increasing vaccination rates, deaths and disease severity caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its new emergent variants continue to threaten the health of people around the world. Therefore, there is an urgent need to develop novel strategies for research, diagnosis, treatment, and government policies to combat the variant strains of SARS-CoV-2. Since the state-of-the-art COVID-19 pandemic, the role of selenium in dealing with COVID-19 disease has been widely discussed due to its importance as an essential micronutrient. This review aims at providing all antiviral activities of nanoselenium (Nano-Se) ever explored using different methods in the literature. We systematically summarize the studied antiviral activities of Nano-Se required to project it as an efficient antiviral system as a function of shape, size, and synthesis method. The outcomes of this article not only introduce Nano-Se to the scientific community but also motivate scholars to adopt Nano-Se to tackle any serious virus such as mutated SARS-CoV-2 to achieve an effective antiviral activity in a desired manner.
Collapse
|
35
|
Sah R, Hada V, Mohanty A, Alshahrani NZ, Chakraborty S, Bhattacharya M, Chakraborty C, Dhama K. Recent first report of human-to-dog transmission of Monkeypox virus emphasizes an urgent need of enhancing surveillance and strengthen further explorative research to reveal its real magnitude of reverse zoonosis from other animals including pets as like that happened with SARS-CoV-2 / COVID-19 pandemic - Correspondence. Int J Surg 2022; 106:106949. [PMID: 36174830 PMCID: PMC9534089 DOI: 10.1016/j.ijsu.2022.106949] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 09/22/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Ranjit Sah
- Department of Microbiology, Tribhuvan University Teaching Hospital, Institute of Medicine, Kathmandu, 44600, Nepal; Research Scholar, Harvard Medical School, Boston, MA, 02115, USA.
| | - Vivek Hada
- All India Institute of Medical Sciences, Gorakhpur, Uttar Pradesh, 273008, India.
| | - Aroop Mohanty
- All India Institute of Medical Sciences, Gorakhpur, Uttar Pradesh, 273008, India.
| | | | - Sandip Chakraborty
- Department of Veterinary Microbiology, College of Veterinary Sciences and Animal Husbandry, R.K. Nagar, West Tripura, Tripura, Pin, 799008, India.
| | - Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore, 756020, Odisha, India.
| | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, 700126, West Bengal, India.
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Izatnagar, Uttar Pradesh, 243122, India.
| |
Collapse
|
36
|
Mohapatra RK, Sarangi AK, Chakraborty S, Tuli HS, Bhattacharya M, Chakraborty C, Chaicumpa W, Dhama K. NeoCoronavirus (NeoCoV) and its possible future global health threats - Current knowledge and counteracting prospects - Correspondence. Int J Surg 2022; 106:106922. [PMID: 36150652 DOI: 10.1016/j.ijsu.2022.106922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/12/2022] [Indexed: 10/14/2022]
Affiliation(s)
- Ranjan K Mohapatra
- Department of Chemistry, Government College of Engineering, Keonjhar, 758002, Odisha, India.
| | - Ashish K Sarangi
- Department of Chemistry, School of Applied Sciences, Centurion University of Technology and Management, Odisha, India
| | - Sandip Chakraborty
- Department of Veterinary Microbiology, College of Veterinary Sciences and Animal Husbandry, R.K. Nagar, West Tripura, Tripura, Pin-799008, India
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, 133207, Haryana, India
| | - Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, VyasaVihar, Balasore, 756020, Odisha, India
| | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, 700126, West Bengal, India
| | - Wanpen Chaicumpa
- Center of Research Excellence in Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India.
| |
Collapse
|
37
|
de Souza A, de Freitas Amorim VM, Guardia GDA, dos Santos FRC, dos Santos FF, de Souza RF, de Araujo Juvenal G, Huang Y, Ge P, Jiang Y, Li C, Paudel P, Ulrich H, Galante PAF, Guzzo CR. Molecular Dynamics Analysis of Fast-Spreading Severe Acute Respiratory Syndrome Coronavirus 2 Variants and Their Effects on the Interaction with Human Angiotensin-Converting Enzyme 2. ACS OMEGA 2022; 7:30700-30709. [PMID: 36068861 PMCID: PMC9437663 DOI: 10.1021/acsomega.1c07240] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is evolving with mutations in the spike protein, especially in the receptor-binding domain (RBD). The failure of public health measures in some countries to contain the spread of the disease has given rise to novel viral variants with increased transmissibility. However, key questions about how quickly the variants can spread remain unclear. Herein, we performed a structural investigation using molecular dynamics simulations and determined dissociation constant (K D) values using surface plasmon resonance assays of three fast-spreading SARS-CoV-2 variants, alpha, beta, and gamma, as well as genetic factors in host cells that may be related to the viral infection. Our results suggest that the SARS-CoV-2 variants facilitate their entry into the host cell by moderately increased binding affinities to the human ACE2 receptor, different torsions in hACE2 mediated by RBD variants, and an increased spike exposure time to proteolytic enzymes. We also found that other host cell aspects, such as gene and isoform expression of key genes for the infection (ACE2, FURIN, and TMPRSS2), may have few contributions to the SARS-CoV-2 variant infectivity. In summary, we concluded that a combination of viral and host cell factors allows SARS-CoV-2 variants to increase their abilities to spread faster than the wild type.
Collapse
Affiliation(s)
- Anacleto
Silva de Souza
- Department
of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, Brazil
| | | | | | - Felipe R. C. dos Santos
- Molecular
Oncology Center, Hospital Sírio Libanes, São Paulo 01308-050, Brazil
- Programa
Interunidades Em Bioinformática, University of São Paulo, São Paulo 05508-900, Brazil
| | - Filipe F. dos Santos
- Molecular
Oncology Center, Hospital Sírio Libanes, São Paulo 01308-050, Brazil
- Department
of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-900, Brazil
| | - Robson Francisco de Souza
- Department
of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, Brazil
| | | | - Yihua Huang
- ACROBiosystems
Inc., 8 N. Hongda Rd.,
Beijing Economic-Technological Development Area, Beijing 100176, China
| | - Pingju Ge
- ACROBiosystems
Inc., 8 N. Hongda Rd.,
Beijing Economic-Technological Development Area, Beijing 100176, China
| | - Yinan Jiang
- ACROBiosystems
Inc., 8 N. Hongda Rd.,
Beijing Economic-Technological Development Area, Beijing 100176, China
| | - Coco Li
- ACROBiosystems
Inc., 8 N. Hongda Rd.,
Beijing Economic-Technological Development Area, Beijing 100176, China
| | - Prajwal Paudel
- ACROBiosystems
Inc., 8 N. Hongda Rd.,
Beijing Economic-Technological Development Area, Beijing 100176, China
| | - Henning Ulrich
- Department
of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-900, Brazil
| | - Pedro A. F. Galante
- Molecular
Oncology Center, Hospital Sírio Libanes, São Paulo 01308-050, Brazil
| | - Cristiane Rodrigues Guzzo
- Department
of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, Brazil
| |
Collapse
|
38
|
Alshakka M, Hatem NAH, Badullah W, Alsakaf R, Rageh A, Yousef SA, Mohamed Ibrahim MI. Detection of Short-Term Side Effects of ChAdOx1 nCoV-19 Vaccine: A Cross-Sectional Study in a War-Torn Country. Pragmat Obs Res 2022; 13:85-91. [PMID: 36046709 PMCID: PMC9423103 DOI: 10.2147/por.s381836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose The chAdOx1 nCoV-19 vaccine is the first COVID-19 vaccine available in Yemen. Hence, this local-based study was used to identify the type and frequency of short-term side effects following 48 hours of the first shot of the vaccine. Methods A cross-section of vaccinated participants in Aden were surveyed by telephone. Descriptive statistics were used for statistical analysis. Results A total of 500 participants were included through convenient sampling. 27% of them were health care providers. Nearly 70% of the respondent experienced side effects. The top three side effects reported were fever (n=276, 55.2%), myalgia (n=270, 54%) and fatigue (n=247, 49.4%). Generally, most participants stated that they experienced the side effects after the first 24 hours of vaccination. Conclusion Side effects that participants experienced were not different from the literature, indicating a safe profile for the vaccine. Further studies are needed to identify the side effects after the second and third dose of the vaccine. In addition, more studies are required to assess the efficacy of the existing vaccines against new variants.
Collapse
Affiliation(s)
- Mohammed Alshakka
- Section of Clinical Pharmacy, Faculty of Pharmacy, Aden University, Aden, Yemen
| | - Najmaddin A H Hatem
- Department of Pharmacy Practice, College of Clinical Pharmacy, Hodeidah University, Alhodeidah, Yemen
| | - Wafa Badullah
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Aden University, Aden, Yemen
| | - Rabab Alsakaf
- Department of Post-Marketing Surveillance, National Pharmacovigilance Center, Supreme Board of Drugs and Medical Appliances, Aden, Yemen
| | - Ali Rageh
- Section of Clinical Pharmacy, Faculty of Pharmacy, Aden University, Aden, Yemen
| | - Seena Abdulla Yousef
- Department of Community Medicine and Public Health, Faculty of Medicine and Health Sciences, Aden University, Aden, Yemen
| | | |
Collapse
|
39
|
Charitos IA, Ballini A, Lovero R, Castellaneta F, Colella M, Scacco S, Cantore S, Arrigoni R, Mastrangelo F, Dioguardi M. Update on COVID-19 and Effectiveness of a Vaccination Campaign in a Global Context. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:10712. [PMID: 36078427 PMCID: PMC9518080 DOI: 10.3390/ijerph191710712] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 06/15/2023]
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 remains a significant issue for global health, the economy, and society. When SARS-CoV-2 began to spread, the most recent serious infectious disease of this century around the world, with its high morbidity and mortality rates, it is understandable why such infections have generally been spread in the past, mainly from international travel movements. This perspective review aimed to provide an update for clinicians on the recent developments related to the microbiological perspectives in pandemics, diagnostics, prevention (such as the spread of a virus), vaccination campaigns, treatment options, and health consequences for COVID-19 based on the current literature. In this way, the authors attempt to raise awareness on the transversal nature of these challenges by identifying the main risk/vulnerability factors that the scientific community must face including our current knowledge on the virus capacity of the mechanism of entry into the cells, the current classifications of viral variants, the knowledge of the mathematical model on the spread of viruses (the possible routes of transmission), and the effectiveness of vaccination campaigns in a global context of pandemic, particularly from COVID-19, with a look at new or future vaccines.
Collapse
Affiliation(s)
- Ioannis Alexandros Charitos
- Department of Emergency and Urgency, National Poisoning Center, Riuniti University Hospital of Foggia, 71122 Foggia, Italy
| | - Andrea Ballini
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Roberto Lovero
- AOU Policlinico Consorziale di Bari-Ospedale Giovanni XXIII, Clinical Pathology Unit, Policlinico University Hospital of Bari, 70124 Bari, Italy
| | - Francesca Castellaneta
- AOU Policlinico Consorziale di Bari-Ospedale Giovanni XXIII, Clinical Pathology Unit, Policlinico University Hospital of Bari, 70124 Bari, Italy
| | - Marica Colella
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Salvatore Scacco
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari “Aldo Moro”, Piazza G. Cesare 11, 70124 Bari, Italy
| | - Stefania Cantore
- Independent Researcher, Sorriso & Benessere-Ricerca e Clinica, 70129 Bari, Italy
| | - Roberto Arrigoni
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), 70125 Bari, Italy
| | - Filiberto Mastrangelo
- Department of Clinical and Experimental Medicine, Università degli Studi di Foggia, 71122 Foggia, Italy
| | - Mario Dioguardi
- Department of Clinical and Experimental Medicine, Università degli Studi di Foggia, 71122 Foggia, Italy
| |
Collapse
|
40
|
Fawazy NG, Panda SS, Mostafa A, Kariuki BM, Bekheit MS, Moatasim Y, Kutkat O, Fayad W, El-Manawaty MA, Soliman AAF, El-Shiekh RA, Srour AM, Barghash RF, Girgis AS. Development of spiro-3-indolin-2-one containing compounds of antiproliferative and anti-SARS-CoV-2 properties. Sci Rep 2022; 12:13880. [PMID: 35974029 PMCID: PMC9380671 DOI: 10.1038/s41598-022-17883-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/02/2022] [Indexed: 11/09/2022] Open
Abstract
A series of 1″-(alkylsulfonyl)-dispiro[indoline-3,2'-pyrrolidine-3',3″-piperidine]-2,4″-diones 6a‒o has been synthesized through regioselective multi-component azomethine dipolar cycloaddition reaction of 1-(alkylsulfonyl)-3,5-bis(ylidene)-piperidin-4-ones 3a‒h. X-ray diffraction studies (6b‒d,h) confirmed the structures. The majority of the synthesized analogs reveal promising antiproliferation properties against a variety of human cancer cell lines (MCF7, HCT116, A431 and PaCa2) with good selectivity index towards normal cell (RPE1). Some of the synthesized agents exhibit potent inhibitory properties against the tested cell lines with higher efficacies than the standard references (sunitinib and 5-fluorouracil). Compound 6m is the most potent. Multi-targeted inhibitory properties against EGFR and VEGFR-2 have been observed for the synthesized agents. Flow cytometry supports the antiproliferation properties and shows the tested agents as apoptosis and necrosis forming. Vero cell viral infection model demonstrates the anti-SARS-CoV-2 properties of the synthesized agents. Compound 6f is the most promising (about 3.3 and 4.8 times the potency of the standard references, chloroquine and hydroxychloroquine). QSAR models explain and support the observed biological properties.
Collapse
Affiliation(s)
- Nehmedo G Fawazy
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Siva S Panda
- Department of Chemistry and Physics, Augusta University, Augusta, GA, 30912, USA
| | - Ahmed Mostafa
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, 12622, Egypt
| | - Benson M Kariuki
- School of Chemistry, Cardiff University, Main Building, Park Place, Cardiff, CF10 3AT, UK
| | - Mohamed S Bekheit
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Yassmin Moatasim
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, 12622, Egypt
| | - Omnia Kutkat
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, 12622, Egypt
| | - Walid Fayad
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, National Research Centre, Dokki, Giza, 12622, Egypt
| | - May A El-Manawaty
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Ahmed A F Soliman
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Riham A El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Aladdin M Srour
- Department of Therapeutic Chemistry, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Reham F Barghash
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Adel S Girgis
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza, 12622, Egypt.
| |
Collapse
|
41
|
Moreno A, Lelli D, Trogu T, Lavazza A, Barbieri I, Boniotti M, Pezzoni G, Salogni C, Giovannini S, Alborali G, Bellini S, Boldini M, Farioli M, Ruocco L, Bessi O, Maroni Ponti A, Di Bartolo I, De Sabato L, Vaccari G, Belli G, Margutti A, Giorgi M. SARS-CoV-2 in a Mink Farm in Italy: Case Description, Molecular and Serological Diagnosis by Comparing Different Tests. Viruses 2022; 14:v14081738. [PMID: 36016360 PMCID: PMC9415545 DOI: 10.3390/v14081738] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/01/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
This study described a SARS-CoV-2 infection in minks on an Italian farm. Surveillance was performed based on clinical examination and a collection of 1879 swabs and 74 sera from dead and live animals. The farm was placed under surveillance for 4.5 months, from the end of July 2020, when a man working on the farm tested positive by RT-PCR, till mid-December 2020 when all the animals were sacrificed. Clinical examination revealed no clinical signs or increased mortality rates attributable to SARS-CoV-2, while diagnostic tests detected only four weak PCR-positive samples, but 100% of sera were positive for SARS-CoV-2 anti-S antibodies. The phylogenetic analysis of two SARS-CoV-2 sequences from two minks and the sequence of the worker showed that they belonged to different clades. It could be therefore assumed that two distinct introductions of the virus occurred on the farm, and that the first introduction probably occurred before the start of the surveillance period. From the data collected, and especially from the detection of specific antibodies through the combination of different tests, it can be postulated that syndromic surveillance combined with genome detection by PCR may not be sufficient to achieve a diagnosis in asymptomatic animals. In particular, the serological approach, especially when using tests directed towards the S protein, may be useful for improving the traceability of virus circulation in similar environments.
Collapse
Affiliation(s)
- Ana Moreno
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna, IZSLER, Via Bianchi, 9, 25124 Brescia, Italy
- Correspondence:
| | - Davide Lelli
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna, IZSLER, Via Bianchi, 9, 25124 Brescia, Italy
| | - Tiziana Trogu
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna, IZSLER, Via Bianchi, 9, 25124 Brescia, Italy
| | - Antonio Lavazza
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna, IZSLER, Via Bianchi, 9, 25124 Brescia, Italy
| | - Ilaria Barbieri
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna, IZSLER, Via Bianchi, 9, 25124 Brescia, Italy
| | - MariaBeatrice Boniotti
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna, IZSLER, Via Bianchi, 9, 25124 Brescia, Italy
| | - Giulia Pezzoni
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna, IZSLER, Via Bianchi, 9, 25124 Brescia, Italy
| | - Cristian Salogni
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna, IZSLER, Via Bianchi, 9, 25124 Brescia, Italy
| | - Stefano Giovannini
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna, IZSLER, Via Bianchi, 9, 25124 Brescia, Italy
| | - Giovanni Alborali
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna, IZSLER, Via Bianchi, 9, 25124 Brescia, Italy
| | - Silvia Bellini
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna, IZSLER, Via Bianchi, 9, 25124 Brescia, Italy
| | - Massimo Boldini
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna, IZSLER, Via Bianchi, 9, 25124 Brescia, Italy
| | - Marco Farioli
- Direzione Generale Welfare, Regione Lombardia, Piazza Città di Lombardia 1, 20124 Milano, Italy
| | - Luigi Ruocco
- Direzione Generale Sanità Animale e Farmaci Veterinari, Ministero della Salute, Via Giorgio Ribotta, 5-00144 Roma, Italy
| | - Olivia Bessi
- Direzione Generale Sanità Animale e Farmaci Veterinari, Ministero della Salute, Via Giorgio Ribotta, 5-00144 Roma, Italy
| | - Andrea Maroni Ponti
- Direzione Generale Sanità Animale e Farmaci Veterinari, Ministero della Salute, Via Giorgio Ribotta, 5-00144 Roma, Italy
| | - Ilaria Di Bartolo
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Luca De Sabato
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Gabriele Vaccari
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Gabriele Belli
- Dipartimento di Prevenzione Veterinario, ATS Valpadana, Via Belgiardino, 6-26100 Cremona, Italy
| | - Alberto Margutti
- Dipartimento di Prevenzione Veterinario, ATS Valpadana, Via Belgiardino, 6-26100 Cremona, Italy
| | - Maurilio Giorgi
- Dipartimento di Prevenzione Veterinario, ATS Valpadana, Via Belgiardino, 6-26100 Cremona, Italy
| |
Collapse
|
42
|
Uppal T, Tuffo K, Khaiboullina S, Reganti S, Pandori M, Verma SC. Screening of SARS-CoV-2 antivirals through a cell-based RNA-dependent RNA polymerase (RdRp) reporter assay. CELL INSIGHT 2022; 1:100046. [PMID: 37192863 PMCID: PMC9239919 DOI: 10.1016/j.cellin.2022.100046] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/24/2022] [Accepted: 06/25/2022] [Indexed: 11/29/2022]
Abstract
COVID-19 (Coronavirus Disease 2019) caused by SARS-CoV-2 (Severe Acute Respiratory Syndrome CoronaVirus-2) continues to pose an international public health threat and thus far, has resulted in greater than 6.4 million deaths worldwide. Vaccines are critical tools to limit COVID-19 spread, but antiviral drug development is an ongoing global priority due to fast-spreading COVID-19 variants that may elude vaccine efficacies. The RNA-dependent RNA polymerase (RdRp) of SARS-CoV-2 is an essential enzyme of viral replication and transcription machinery complex. Therefore, the RdRp is an attractive target for the development of effective anti-COVID-19 therapeutics. In this study, we developed a cell-based assay to determine the enzymatic activity of SARS-CoV-2 RdRp through a luciferase reporter system. The SARS-CoV-2 RdRp reporter assay was validated using known inhibitors of RdRp polymerase, remdesivir along with other anti-virals including ribavirin, penciclovir, rhoifolin, 5′CT, and dasabuvir. Dasabuvir (an FDA-approved drug) exhibited promising RdRp inhibitory activity among these inhibitors. Anti-viral activity of dasabuvir was also tested on the replication of SARS-CoV-2 through infection of Vero E6 cells. Dasabuvir inhibited the replication of SARS-CoV-2, USA-WA1/2020 as well as B.1.617.2 (delta variant) in Vero E6 cells in a dose-dependent manner with EC50 values 9.47 μM and 10.48 μM, for USA-WA1/2020 and B.1.617.2 variants, respectively. Our results suggest that dasabuvir can be further evaluated as a therapeutic drug for COVID-19. Importantly, this system provides a robust, target-specific, and high-throughput screening compatible (z- and z’-factors of >0.5) platforms that will be a valuable tool for screening SARS-CoV-2 RdRp inhibitors.
Collapse
|
43
|
Shanmugaraj B, Khorattanakulchai N, Panapitakkul C, Malla A, Im-Erbsin R, Inthawong M, Sunyakumthorn P, Hunsawong T, Klungthong C, Reed MC, Kemthong T, Suttisan N, Malaivijitnond S, Srimangkornkaew P, Klinkhamhom A, Manopwisedjaroen S, Thitithanyanont A, Taychakhoonavudh S, Phoolcharoen W. Preclinical evaluation of a plant-derived SARS-CoV-2 subunit vaccine: Protective efficacy, immunogenicity, safety, and toxicity. Vaccine 2022; 40:4440-4452. [PMID: 35697573 PMCID: PMC9167921 DOI: 10.1016/j.vaccine.2022.05.087] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/06/2022] [Accepted: 05/31/2022] [Indexed: 01/01/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is an acute respiratory illness caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The prevention of SARS-CoV-2 transmission has become a global priority. Previously, we showed that a protein subunit vaccine that was developed based on the fusion of the SARS-CoV-2 receptor-binding domain (RBD) to the Fc portion of human IgG1 (RBD-Fc), produced in Nicotiana benthamiana, and adjuvanted with alum, namely, Baiya SARS-CoV-2 Vax 1, induced potent immunological responses in both mice and cynomolgus monkeys. Hence, this study evaluated the protective efficacy, safety, and toxicity of Baiya SARS-CoV-2 Vax 1 in K18-hACE2 mice, monkeys and Wistar rats. Two doses of vaccine were administered three weeks apart on Days 0 and 21. The administration of the vaccine to K18-hACE2 mice reduced viral loads in the lungs and brains of the vaccinated animals and protected the mice against challenge with SARS-CoV-2. In monkeys, the results of safety pharmacology tests, general clinical observations, and a core battery of studies of three vital systems, namely, the central nervous, cardiovascular, and respiratory systems, did not reveal any safety concerns. The toxicology study of the vaccine in rats showed no vaccine-related pathological changes, and all the animals remained healthy under the conditions of this study. Furthermore, the vaccine did not cause any abnormal toxicity in rats and was clinically tolerated even at the highest tested concentration. In addition, general health status, body temperature, local toxicity at the administration site, hematology, and blood chemistry parameters were also monitored. Overall, this work presents the results of the first systematic study of the safety profile of a plant-derived vaccine, Baiya SARS-CoV-2 Vax 1; this approach can be considered a viable strategy for the development of vaccines against COVID-19.
Collapse
Affiliation(s)
| | - Narach Khorattanakulchai
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand; Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Chalisa Panapitakkul
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand; Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | | | - Rawiwan Im-Erbsin
- Department of Veterinary Medicine, U.S. Army Medical Directorate-Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Manutsanun Inthawong
- Department of Veterinary Medicine, U.S. Army Medical Directorate-Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Piyanate Sunyakumthorn
- Department of Veterinary Medicine, U.S. Army Medical Directorate-Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Taweewun Hunsawong
- Department of Virology, U.S. Army Medical Directorate-Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Chonticha Klungthong
- Department of Virology, U.S. Army Medical Directorate-Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Matthew C Reed
- Department of Veterinary Medicine, U.S. Army Medical Directorate-Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Taratorn Kemthong
- National Primate Research Center of Thailand-Chulalongkorn University, Saraburi 18110, Thailand
| | - Nutchanat Suttisan
- National Primate Research Center of Thailand-Chulalongkorn University, Saraburi 18110, Thailand
| | - Suchinda Malaivijitnond
- National Primate Research Center of Thailand-Chulalongkorn University, Saraburi 18110, Thailand
| | | | - Aekkarin Klinkhamhom
- National Laboratory Animal Center, Mahidol University, Nakorn Pathom 73170, Thailand
| | | | - Arunee Thitithanyanont
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Suthira Taychakhoonavudh
- Department of Social and Administrative Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Waranyoo Phoolcharoen
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand; Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
44
|
Reviewing the Past, Present, and Future Risks of Pathogens in Ghana and What This Means for Rethinking Infectious Disease Surveillance for Sub-Saharan Africa. J Trop Med 2022; 2022:4589007. [PMID: 35846072 PMCID: PMC9284326 DOI: 10.1155/2022/4589007] [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: 12/05/2021] [Revised: 04/17/2022] [Accepted: 06/15/2022] [Indexed: 11/18/2022] Open
Abstract
The current epidemiological transition makes us wonder how the parallel of infectious diseases (IDs) might be at the end of each passing year. Yet, the surveillance of these IDs continues to focus on high-profile diseases of public health importance without keeping track of the broad spectrum of the IDs we face. Here, we presented the prevalence of the broad spectrum of IDs in Ghana. Data from the annual reports on Gold Coast now Ghana, Global Infectious Diseases and Epidemiology Network (GIDEON), and the District Health Information Management System II (DHIMS2) databases were examined for records of ID prevalence in Ghana. Using the IDs from these databases, the paper assessed the epidemiological transition, pathogen-host interactions, spatiotemporal distribution, transmission routes, and their potential areas of impact in Ghana. The topmost ID recorded in health facilities in Ghana transitioned from yaws in the 1890s to malaria in the 1950s through 2020. We then presented the hosts of a pathogen and the pathogens of a host, the administrative districts where a pathogen was found, and the pathogens found in each district of Ghana. The highest modes of transmission routes were through direct contact for bacteria and airborne or droplet-borne for viral pathogens. From GIDEON, 226 IDs were identified as endemic or potentially endemic in Ghana, with 42% cited in peer-reviewed articles from 2000 to 2020. From the extent of risk of endemic or potentially endemic IDs, Ghana faces a high risk of ID burden that we should be mindful of their changing patterns and should keep track of the state of each of them.
Collapse
|
45
|
Weil T, Lawrenz J, Seidel A, Münch J, Müller JA. Immunodetection assays for the quantification of seasonal common cold coronaviruses OC43, NL63, or 229E infection confirm nirmatrelvir as broad coronavirus inhibitor. Antiviral Res 2022; 203:105343. [PMID: 35598779 PMCID: PMC9119192 DOI: 10.1016/j.antiviral.2022.105343] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/05/2022] [Accepted: 05/16/2022] [Indexed: 01/21/2023]
Abstract
Besides pandemic SARS-CoV-2, also endemic seasonal human common cold coronaviruses (hCoVs) have a significant impact on human health and economy. Studies on hCoVs and the identification of antivirals are therefore crucial to improve human well-being. However, hCoVs have long been neglected and the methodology to study virus infection, replication and inhibition warrants being updated. We here evaluated the established plaque-based assays to determine viral titers and cell-to-cell spread and developed protocols for the immunodetection of the viral nucleocapsid protein by flow cytometry and in-cell ELISA to study infection rates at early time points. The developed protocols allow detection of hCoV-229E infection after 2, and hCoV-NL63 and -OC43 infection after 3 days at a single cell level or in a 96 well microtiter format, in large sample numbers without being laborious or expensive. Both assays can be applied to assess the susceptibility of cells to hCoV infection and replication, and to determine the efficacy of antiviral compounds as well as neutralizing antibodies in a sensitive and quantitative manner. Application revealed that clinically applied SARS-CoV-2 targeting monoclonal antibodies are inactive against hCoVs, but that the viral polymerase targeting antivirals remdesivir and molnupiravir are broadly active also against all three hCoVs. Further, the in-cell ELISA provided evidence that nirmatrelvir, previously shown to broadly inhibit coronavirus proteases, also prevents replication of authentic hCoVs. Importantly, the protocols described here can be easily adapted to other coronavirus strains and species as well as viruses of other families within a short time. This will facilitate future research on known and emerging (corona)viruses, support the identification of antivirals and increase the preparedness for future virus outbreaks.
Collapse
Affiliation(s)
- Tatjana Weil
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Jan Lawrenz
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Alina Seidel
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany; Core Facility Functional Peptidomics, Ulm University Medical Center, 89081, Ulm, Germany
| | - Janis A Müller
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany; Institute of Virology, Philipps University of Marburg, 35043, Marburg, Germany.
| |
Collapse
|
46
|
Ahmad S, Bano N, Qazi S, Yadav MK, Ahmad N, Raza K. Multitargeted Molecular Dynamic Understanding of Butoxypheser Against SARS-CoV-2: An in Silico Study. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221115499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) was initially reported in the Wuhan province of China, spread throughout the world, and caused massive destruction in the form of a global pandemic that started back in 2020. SARS-CoV-2 is an RNA virus with various proteins like main protease, spike protein, NSP15 endoribonuclease, RNA-dependent RNA polymerase, and papain-like protease targeted to screen and find the novel drug candidate that can potentially work against the virus. Previous studies have reported multiple drugs after screening and validation against a single target and reported multiple medications. Nevertheless, many drugs are being used to date but do not have enough potential to work against SARS-CoV-2 and curb the spread and death rate. In this study, with the hypothesis of 1 drug and multiple targets, we have taken 5 main target proteins and screened the Asinex's complete BioDesign library (1,70,269 compounds) and identified N-{2-[(2S)-2-Amino-3-methylbutoxy]-6-propylbenzoyl}-L-phenylalanyl-L-serine (Butoxypheser) as multitarget inhibitor against SARS-CoV-2. Also, Butoxypheser has shown excellent docking scores, hydrogen bonding, and other bonding configurations like van der Waals force and water bridges. The stability and interaction pattern of the compound was validated with structural interaction fingerprints (SIFts) and molecular dynamics (MD) simulation. The Butoxypheser has performed flawlessly throughout the study, and the same results were used to compare the compound's activity against multiple targets. After a thorough theoretical comparative analysis, Butoxypheser can be treated as a multitargeted inhibitor candidate against SARS-CoV-2. Further, this study needs to be validated experimentally before human use.
Collapse
Affiliation(s)
- Shaban Ahmad
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| | - Nagmi Bano
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| | - Sahar Qazi
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| | - Manoj Kumar Yadav
- Department of Biomedical Engineering, SRM University, Delhi-NCR, Sonepat, India
| | - Nadeem Ahmad
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Khalid Raza
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
47
|
Lin Q, Lu C, Hong Y, Li R, Chen J, Chen W, Chen J. Animal models for studying coronavirus infections and developing antiviral agents and vaccines. Antiviral Res 2022; 203:105345. [PMID: 35605699 PMCID: PMC9122840 DOI: 10.1016/j.antiviral.2022.105345] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 04/30/2022] [Accepted: 05/17/2022] [Indexed: 01/17/2023]
Abstract
In addition to severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV), SARS-CoV-2 has become the third deadly coronavirus that infects humans and causes the new coronavirus disease (COVID-19). COVID-19 has already caused more than six million deaths worldwide and it is likely the biggest pandemic of this century faced by mankind. Although many studies on SARS-CoV-2 have been conducted, a detailed understanding of SARS-CoV-2 and COVID-19 is still lacking. Animal models are indispensable for studying its pathogenesis and developing vaccines and antivirals. In this review, we analyze animal models of coronavirus infections and explore their applications on antivirals and vaccines.
Collapse
Affiliation(s)
- Qisheng Lin
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Chunni Lu
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria 3168, Australia
| | - Yuqi Hong
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Runfeng Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Jinding Chen
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Weisan Chen
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia.
| | - Jianxin Chen
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
48
|
Wassenaar TM, Wanchai V, Buzard G, Ussery DW. The first three waves of the Covid-19 pandemic hint at a limited genetic repertoire for SARS-CoV-2. FEMS Microbiol Rev 2022; 46:fuac003. [PMID: 35076068 PMCID: PMC9075578 DOI: 10.1093/femsre/fuac003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 12/17/2021] [Accepted: 01/13/2022] [Indexed: 11/22/2022] Open
Abstract
The genomic diversity of SARS-CoV-2 is the result of a relatively low level of spontaneous mutations introduced during viral replication. With millions of SARS-CoV-2 genome sequences now available, we can begin to assess the overall genetic repertoire of this virus. We find that during 2020, there was a global wave of one variant that went largely unnoticed, possibly because its members were divided over several sublineages (B.1.177 and sublineages B.1.177.XX). We collectively call this Janus, and it was eventually replaced by the Alpha (B.1.1.7) variant of concern (VoC), next replaced by Delta (B.1.617.2), which itself might soon be replaced by a fourth pandemic wave consisting of Omicron (B.1.1.529). We observe that splitting up and redefining variant lineages over time, as was the case with Janus and is now happening with Alpha, Delta and Omicron, is not helpful to describe the epidemic waves spreading globally. Only ∼5% of the 30 000 nucleotides of the SARS-CoV-2 genome are found to be variable. We conclude that a fourth wave of the pandemic with the Omicron variant might not be that different from other VoCs, and that we may already have the tools in hand to effectively deal with this new VoC.
Collapse
Affiliation(s)
- Trudy M Wassenaar
- Molecular Microbiology and Genomics Consultants, Tannenstrasse 7, 55576 Zotzenheim, Germany
| | - Visanu Wanchai
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR 772205, USA
| | | | - David W Ussery
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR 772205, USA
| |
Collapse
|
49
|
Cameron A, Esiovwa R, Connolly J, Hursthouse A, Henriquez F. Antimicrobial Resistance as a Global Health Threat: The Need to Learn Lessons from the COVID-19 Pandemic. GLOBAL POLICY 2022; 13:179-192. [PMID: 35601654 PMCID: PMC9111155 DOI: 10.1111/1758-5899.13049] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 06/10/2023]
Abstract
The global COVID-19 pandemic has exacerbated existing health, social, and economic challenges and threatened progress towards achieving the UN sustainable development goals. We discuss lessons learned from the COVID-19 pandemic for global policymaking for health security governance, with a particular focus on antimicrobial resistance. We identify One Health as the primary foundation of public health risk management owing to the collaborative, multidisciplinary, and multisectoral efforts that underpin the One Health approach and that enhance understanding of the complex interactions at the human-animal-environment interface. We discuss the narrow human-centric focus of the One Health approach, highlight the underrepresentation of the environmental sector in One Health networks, and encourage greater representation from the environmental sector. Furthermore, we highlight the importance of the social sciences for health security research and the need for effective communication and trust. Finally, we underscore the importance of strengthened and collaborative health, social care, and disaster management systems. The application of these lessons will facilitate holistic, multisectoral, collaborative, and ethical actions on antimicrobial resistance.
Collapse
|
50
|
Chaintoutis SC, Siarkou VI, Mylonakis ME, Kazakos GM, Skeva P, Bampali M, Dimitriou M, Dovrolis N, Polizopoulou ZS, Karakasiliotis I, Dovas CI. Limited cross-species transmission and absence of mutations associated with SARS-CoV-2 adaptation in cats: A case study of infection in a small household setting. Transbound Emerg Dis 2022; 69:1606-1616. [PMID: 33908152 PMCID: PMC8242912 DOI: 10.1111/tbed.14132] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/23/2021] [Indexed: 12/20/2022]
Abstract
In the present study, the course of SARS-CoV-2 natural infection in two asymptomatic cats, which were negative for immunosuppressive retroviral infections, is investigated. The source of the virus for the cats was their COVID-19-affected owner, with whom they were in continuous proximity in a small household setting. The owner's signs included fatigue, sneezing, anosmia and loss of taste, and diagnosis was confirmed 4 days after symptom onset. Oropharyngeal and faecal swabs were collected from the cats, to investigate the course of SARS-CoV-2 RNA concentrations, as well as the directionality of the chain of virus transmission. Both infected cats were real-time RT-PCR-positive on various time-points. Pharyngeal shedding of at least 6 days was observed in them, with high SARS-CoV-2 titres (> 7 Log10 copies/swab) on the first sampling time-point, that is, 7 days after the onset of owner's clinical signs. In one cat, after the initial decline, slightly increasing virus titres were measured 3 to 6 days after the first real-time RT-PCR-positive swab. Serological testing of this cat revealed absence of seroconversion. The course of viral RNA concentrations in the faecal swabs of the other cat was similar to that in its pharynx. The detected SARS-CoV-2 strains, from both infected cats and their owner, underwent whole-genome sequencing, revealing the absence of emergence of cross-species adaptive mutations in cats. The results support the notion that human SARS-CoV-2 strains are relatively well-adapted to cats. It is still unclear whether asymptomatic animals could play a role in COVID-19 epidemiology, in case of interaction with naïve animals and/or people. Our findings highlight difficulties in SARS-CoV-2 transmission to cats, as neither the two infected cats nor their owner was able to transmit the virus to a third cat living in the same small flat, despite their very close contact during the days corresponding to high virus shedding.
Collapse
Affiliation(s)
- Serafeim C. Chaintoutis
- Diagnostic LaboratorySchool of Veterinary MedicineFaculty of Health SciencesAristotle University of ThessalonikiThessalonikiGreece
| | - Victoria I. Siarkou
- Laboratory of Microbiology and Infectious DiseasesSchool of Veterinary MedicineFaculty of Health SciencesAristotle University of ThessalonikiThessalonikiGreece
| | - Mathios E. Mylonakis
- Companion Animal ClinicSchool of Veterinary MedicineFaculty of Health SciencesAristotle University of ThessalonikiThessalonikiGreece
| | - George M. Kazakos
- Companion Animal ClinicSchool of Veterinary MedicineFaculty of Health SciencesAristotle University of ThessalonikiThessalonikiGreece
| | - Panagiota‐Nefeli Skeva
- School of Veterinary MedicineFaculty of Health SciencesAristotle University of ThessalonikiThessalonikiGreece
| | - Maria Bampali
- Laboratory of BiologyDepartment of MedicineDemocritus University of ThraceXanthiGreece
| | - Marios Dimitriou
- Laboratory of BiologyDepartment of MedicineDemocritus University of ThraceXanthiGreece
| | - Nikolaos Dovrolis
- Laboratory of BiologyDepartment of MedicineDemocritus University of ThraceXanthiGreece
| | - Zoe S. Polizopoulou
- Diagnostic LaboratorySchool of Veterinary MedicineFaculty of Health SciencesAristotle University of ThessalonikiThessalonikiGreece
| | | | - Chrysostomos I. Dovas
- Diagnostic LaboratorySchool of Veterinary MedicineFaculty of Health SciencesAristotle University of ThessalonikiThessalonikiGreece
| |
Collapse
|