1
|
de Morais Gomes V, Santos DM, Macedo-da-Silva J, Lazari LC, Machado RRG, Dos Santos AF, Araujo DB, Coutinho JVP, Arini GS, Angeli CB, de Souza EE, Marques RF, Boscardin SB, Wrenger C, Marinho CRF, Oliveira DBL, Durigon EL, Labriola L, Rosa-Fernandes L, Palmisano G. P.1 and P.2 SARS-CoV-2 Brazilian variants activate the unfolded protein response with a time and pathway specificity. J Proteomics 2025; 315:105397. [PMID: 39909104 DOI: 10.1016/j.jprot.2025.105397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/07/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
COVID-19 is a human respiratory syndrome caused by the infection of the SARS-CoV-2 virus that has a high rate of infection and mortality. Viruses modulate the host machinery by altering cellular mechanisms that favor their replication. One of the mechanisms that viruses exploit is the protein folding and processing of post-translational modifications that occur in the endoplasmic reticulum (ER). When ER function is impaired, there is an accumulation of misfolded proteins leading to endoplasmic reticulum stress (ER stress). To maintain homeostasis, cells trigger an adaptive signaling mechanism called the Unfolded Protein Response (UPR) which helps cells deal with stress, but under severe conditions, can activate the apoptotic cell death mechanism. This study elucidated an activation of a diversity of molecular mechanisms by Brazilian variants of SARS-CoV-2 by a time-resolved and large-scale characterization of SARS-CoV-2-infected cells proteomics and immunoblotting. Furthermore, it was shown that pharmacological UPR modulation could reduce viral release by counteracting the different viral activations of its cellular response. Analysis of human clinical specimens and disease outcomes focusing on ER stress reinforces the importance of UPR modulation as a host regulatory mechanism during viral infection and could point to novel therapeutic targets. SIGNIFICANCE: Since the emergence of SARS-CoV-2 and the consequent COVID-19 pandemic, the rapid emergence of variants of this new coronavirus has been a cause for concern since many of them have significantly higher rates of transmissibility and virulence, being called Variants of Concern (VOC). In this work, we studied the VOCs Gamma (P.1) and Zeta (P.2), also known as Brazilian variants. Constant evidence has reported that there are particularities related to each variant of SARS-CoV-2, with different rates of transmissibility, replication and modulation of host biological processes being observed, in addition to the mutations present in the variants. For this reason, this work focused on infections caused by the Brazilian variants of SARS-CoV-2 in different cell lines, in which we were able to observe that the infections caused by the variants induced endoplasmic reticulum stress in the infected cells and activated the UPR pathways, presenting specific modulations of each variant in this pathway. Furthermore, transcriptome analysis of patients revealed a correlation between ER-related genes and COVID-19 progression. Finally, we observed that the use of UPR modulators in host cells decreased viral release of all variants without affecting cell viability. The data presented in this work complement the observations of other studies that aim to understand the pathogenicity of SARS-CoV-2 VOCs and possible new therapeutic strategies, mainly targeting biological processes related to the endoplasmic reticulum.
Collapse
Affiliation(s)
| | - Deivid Martins Santos
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Janaina Macedo-da-Silva
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Lucas C Lazari
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil
| | | | | | - Danielle Bastos Araujo
- Laboratory of Clinical and Molecular Virology, Department of Microbiology, ICB, University of São Paulo, Brazil
| | | | - Gabriel Santos Arini
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Brazil
| | - Claudia B Angeli
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Edmarcia E de Souza
- Unit for Drug Discovery, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Rodolfo F Marques
- Laboratory of Antigen Targeting for Dendritic Cells, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Silvia Beatriz Boscardin
- Laboratory of Antigen Targeting for Dendritic Cells, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Carsten Wrenger
- Unit for Drug Discovery, Department of Parasitology, ICB, University of São Paulo, Brazil
| | | | - Danielle B L Oliveira
- Laboratory of Clinical and Molecular Virology, Department of Microbiology, ICB, University of São Paulo, Brazil
| | - Edison L Durigon
- Laboratory of Clinical and Molecular Virology, Department of Microbiology, ICB, University of São Paulo, Brazil; Scientific Platform Pasteur USP, Sao Paulo, Brazil
| | - Leticia Labriola
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Brazil
| | - Livia Rosa-Fernandes
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil; Laboratory of Experimental Immunoparasitology, Department of Parasitology, ICB, University of São Paulo, Brazil; Centre for Motor Neuron Disease Research, Faculty of Medicine, Health & Human Sciences, Macquarie Medical School, Sydney, Australia
| | - Giuseppe Palmisano
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil; School of Natural Sciences, Macquarie University, Sydney, Australia.
| |
Collapse
|
2
|
Zhang L, Li F, Liu X, Liu XA, Lu D, Luo Q, Liu Q, Jiang G. Long-term effects of SARS-CoV-2 infection on metal homeostasis. J Trace Elem Med Biol 2025; 88:127625. [PMID: 40023939 DOI: 10.1016/j.jtemb.2025.127625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/10/2025] [Accepted: 02/17/2025] [Indexed: 03/04/2025]
Abstract
The outbreak of COVID-19 pandemic has caused substantial health loss worldwide, and the long-term sequelae of COVID, resulting from repeated coronavirus infection, have emerged as a new public health concern. We report the widespread presence of abnormal metallomic profiles in the sera of patients who have recovered from SARS-CoV-2 coronavirus infection, even after 6 months post-discharge from hospital. We measured the concentrations of Fe, Cu, Zn, Se, Cr, Mn, Ba, Ni, Pb, Ag, As, Cd, Co, and V in the sera of 25 recovered participants and 38 healthy controls in the cross-sectional study. Higher concentrations of Cu, Ag, As, Ba, Cd, Ni, Pb, Cr and V were observed in the recovered participants, whereas lower concentrations of Fe and Se were obtained in these participants. Except for Zn, Mn, and Co, all other elements showed significant differences (p < 0.05) between the two groups, with variations dependent on age and gender. Further correlation analysis between metallome and metabolome indicated that SARS-CoV-2 infection continues to disrupt metallic homeostasis and affect metabolic processes, such as lipid metabolism and cell respiration, as well as the functions of certain organs (e.g., liver, kidney, and heart), even after 6 months recovery. Our findings provide novel insights into the potential long-term effect of COVID-19 on the human body from a new perspective of metallomics.
Collapse
Affiliation(s)
- Luyao Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Fang Li
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Xiaoxiong Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Xin-An Liu
- Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Dawei Lu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Qian Luo
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Qian Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
3
|
Schmidt KG, Geißler P, Schuster EM, Schülein C, Harrer EG, Schönau V, Luber M, Spriewald B, Steininger P, Bergmann S, Ensser A, Schober K, Nganou-Makamdop K, Harrer T. Coronavirus replicase epitopes induce cross-reactive CD8 T cell responses in SARS-CoV-2-naive people with HIV-1. iScience 2025; 28:111949. [PMID: 40034846 PMCID: PMC11872457 DOI: 10.1016/j.isci.2025.111949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/12/2024] [Accepted: 01/30/2025] [Indexed: 03/05/2025] Open
Abstract
Cross-reactive T cell immunity between common cold coronaviruses and SARS-CoV-2 may influence COVID-19 susceptibility. To identify cross-reactive CD8 T cell epitopes, we analyzed responses to 21 homologous SARS-CoV-2 replicase peptides in 177 people living with HIV (PLWH) on antiretroviral therapy, of which 133 did not have prior SARS-CoV-2 infection. Replicase peptides induced IFN-γ responses in 63% of the SARS-CoV-2-naïve individuals and in 73% of individuals with prior SARS-CoV-2-infection. We could define several cross-reactive epitopes, including the HLA-B∗35:03 restricted CoV-YL8, and characterized a CoV-YL8-specific T cell receptor cloned from a SARS-CoV-2 seronegative individual. Analysis of the association between HLA-I alleles and SARS-CoV-2 infections over a 16-months period revealed that in a cohort of 452 PLWH, HLA-B∗35:03 and C∗07 were underrepresented in the 55 persons with a history of SARS-CoV-2 infection while HLA-B∗35:01 and HLA-C∗04 were associated with a higher infection rate. Taken together, our study suggests an HLA-I-mediated effect of common cold coronaviruses on SARS-CoV-2 immunity.
Collapse
Affiliation(s)
- Katja G. Schmidt
- Infectious Diseases and Immunodeficiency Section, Department of Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Paulina Geißler
- Infectious Diseases and Immunodeficiency Section, Department of Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Ev-Marie Schuster
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Christine Schülein
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Ellen G. Harrer
- Infectious Diseases and Immunodeficiency Section, Department of Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Verena Schönau
- Department of Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Markus Luber
- Department of Medicine 5, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Bernd Spriewald
- Department of Medicine 5, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- FAU Profile Center Immunomedicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Philipp Steininger
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Silke Bergmann
- Infectious Diseases and Immunodeficiency Section, Department of Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Armin Ensser
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Kilian Schober
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- FAU Profile Center Immunomedicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Krystelle Nganou-Makamdop
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Thomas Harrer
- Infectious Diseases and Immunodeficiency Section, Department of Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- FAU Profile Center Immunomedicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
4
|
Wu L, Geng S, Zhou Y, Jiao R, Ouyang H, Cheng S, Shang Z, Yan W, Wang J, Jia Y, Li X, Liu W. Direct and Indirect Effects of Risk Perception and Risk Information on PTSD in Frontline Healthcare Workers. Neuropsychiatr Dis Treat 2025; 21:663-676. [PMID: 40134761 PMCID: PMC11934884 DOI: 10.2147/ndt.s491046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
Background The COVID-19 Delta variant caused an outbreak in Guangdong in mid-May 2021. The risk information and risk perception of COVID-19 have been considered factors associated with mental health statuses, especially posttraumatic stress disorder (PTSD), in frontline healthcare workers. Objective The aim of this study is to investigate the interactive relationship in risk information and risk perception of COVID-19 and PTSD in healthcare personnel from the emergency department. Design We conducted a survey one month after the outbreak. A cross-sectional survey design is adopted, and 3078 participants are enrolled. The PTSD checklist for DSM-5 (PCL-5), risk information questionnaire, and risk perception questionnaire are utilized to collect data. Methods Bivariate correlation analysis and structural equation modelling are performed to analyze the mediating role of risk perception in the relationship between risk information and PTSD. Results The estimated prevalence of PTSD among frontline healthcare workers is 28.2%. The risk information for COVID-19, risk perception, and PTSD symptoms are mutually correlated. The specific paths from risk perception to PTSD show significant effects, in which two had negative effects and one had a positive effect. The risk perception feature in 2020 differed from that in 2019, which is largely due to the risk perception of the virus. Conclusion The estimated prevalence of PTSD among frontline healthcare workers remains high. Risk information for COVID-19 has dual effects on PTSD through the mediation of risk perceptions. Unfamiliarity with the SAR-COVID-2 Delta variant increased the risk perception of COVID-19. Effective risk communication regarding COVID-19 can have a positive effect on the mental health of frontline healthcare workers.
Collapse
Affiliation(s)
- Lili Wu
- Lab for Post-Traumatic Stress Disorder, Faculty of Psychology and Mental Health, Navy Medical University, Shanghai, 200433, People’s Republic of China
- The Emotion & Cognition Lab, Faculty of Psychology and Mental Health, Navy Medical University, Shanghai, 200433, People’s Republic of China
| | - Shiyu Geng
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Yaoguang Zhou
- Lab for Post-Traumatic Stress Disorder, Faculty of Psychology and Mental Health, Navy Medical University, Shanghai, 200433, People’s Republic of China
- The Emotion & Cognition Lab, Faculty of Psychology and Mental Health, Navy Medical University, Shanghai, 200433, People’s Republic of China
| | - Runda Jiao
- Graduate School, PLA General Hospital, Beijing, 100853, People’s Republic of China
| | - Hui Ouyang
- Lab for Post-Traumatic Stress Disorder, Faculty of Psychology and Mental Health, Navy Medical University, Shanghai, 200433, People’s Republic of China
- The Emotion & Cognition Lab, Faculty of Psychology and Mental Health, Navy Medical University, Shanghai, 200433, People’s Republic of China
| | - Suhui Cheng
- Lab for Post-Traumatic Stress Disorder, Faculty of Psychology and Mental Health, Navy Medical University, Shanghai, 200433, People’s Republic of China
- The Emotion & Cognition Lab, Faculty of Psychology and Mental Health, Navy Medical University, Shanghai, 200433, People’s Republic of China
| | - Zhilei Shang
- Lab for Post-Traumatic Stress Disorder, Faculty of Psychology and Mental Health, Navy Medical University, Shanghai, 200433, People’s Republic of China
- The Emotion & Cognition Lab, Faculty of Psychology and Mental Health, Navy Medical University, Shanghai, 200433, People’s Republic of China
| | - Wenjie Yan
- Lab for Post-Traumatic Stress Disorder, Faculty of Psychology and Mental Health, Navy Medical University, Shanghai, 200433, People’s Republic of China
- The Emotion & Cognition Lab, Faculty of Psychology and Mental Health, Navy Medical University, Shanghai, 200433, People’s Republic of China
| | - Jing Wang
- Lab for Post-Traumatic Stress Disorder, Faculty of Psychology and Mental Health, Navy Medical University, Shanghai, 200433, People’s Republic of China
- The Emotion & Cognition Lab, Faculty of Psychology and Mental Health, Navy Medical University, Shanghai, 200433, People’s Republic of China
| | - Yanpu Jia
- Lab for Post-Traumatic Stress Disorder, Faculty of Psychology and Mental Health, Navy Medical University, Shanghai, 200433, People’s Republic of China
- The Emotion & Cognition Lab, Faculty of Psychology and Mental Health, Navy Medical University, Shanghai, 200433, People’s Republic of China
| | - Xu Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Weizhi Liu
- Lab for Post-Traumatic Stress Disorder, Faculty of Psychology and Mental Health, Navy Medical University, Shanghai, 200433, People’s Republic of China
- The Emotion & Cognition Lab, Faculty of Psychology and Mental Health, Navy Medical University, Shanghai, 200433, People’s Republic of China
| |
Collapse
|
5
|
Mendes S, Guimarães LC, de Oliveira LC, Costa PAC, da Silva NJA, Pereira GSAP, Fernandez CC, Figueiredo MM, Dos Santos RAS, Teixeira MM, Costa VV, Guimarães PPG, Frézard F. Intranasal liposomal remdesivir induces SARS-CoV-2 clearance in K18-hACE2 mice and ensures survival. J Control Release 2025; 379:558-573. [PMID: 39837387 DOI: 10.1016/j.jconrel.2025.01.044] [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: 07/19/2024] [Revised: 12/31/2024] [Accepted: 01/16/2025] [Indexed: 01/23/2025]
Abstract
A huge challenge after the emergence of COVID-19 has been the discovery of effective antiviral drugs. Although remdesivir (RDV) emerged as one of the most promising drugs, its pharmaceutical formulation Veklury® is limited by moderate efficacy, high toxicity and need for parenteral administration. The aim of the present work was to develop a liposomal formulation of RDV for pulmonary administration and evaluate its efficacy in models of COVID-19. Liposomal RDV nanoformulation (LRDV) was selected based on high drug encapsulation efficiency, sustained drug release property and high in vitro selectivity index. A pharmacokinetic study of intranasal LRDV in mice demonstrated effective delivery of the drug to the lungs. LRDV was then evaluated for its efficacy in SARS-CoV-2-infected K18-hACE2 mice after repeated intranasal administration at 10 mg/kg/bid for 5 days. Veklury® given intraperitoneally at 20 mg/kg/bid was used for comparison. Mice receiving LRDV remained alive up to 15 days post-infection (dpi). On the other hand, the control groups receiving PBS and empty liposomes showed 100 % death at 6 dpi and the Veklury® group had 62.5 % death at 8 dpi. Intranasal LRDV also promoted a strong reduction in viral loads in the brain and lungs of mice and prevented the inflammatory response induced by SARS-CoV-2 in the lungs. This is in contrast with Veklury®, which did not significantly reduce the viral titer in the brain and was poorly effective in preventing the inflammatory response in the lungs. Intranasal LRDV emerges as a promising therapeutic strategy for COVID-19, including "Long COVID".
Collapse
Affiliation(s)
- Sabrina Mendes
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Lays Cordeiro Guimarães
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Leonardo Camilo de Oliveira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Pedro Augusto Carvalho Costa
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Natália Jordana Alves da Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Gabriel Silva Alves Pessim Pereira
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Clara Couto Fernandez
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | | | - Robson Augusto Souza Dos Santos
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Vivian Vasconcelos Costa
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Pedro Pires Goulart Guimarães
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Frédéric Frézard
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil.
| |
Collapse
|
6
|
Agudelo OL, Reyes-Loaiza V, Giraldo-Parra L, Rosales-Chilama M, Perdomo S, Gómez MA, Rodriguez JW, Ortega V, Daza Rivera CF, Galindo D, Valencia DP, Quimbaya M, Plata S, Bogdanowicz R, Rosso F, Jaramillo-Botero A. Performance assessment of disposable carbon-based immunosensors for the detection of SARS-CoV-2 infections. Sci Rep 2025; 15:7741. [PMID: 40044778 PMCID: PMC11883031 DOI: 10.1038/s41598-025-92104-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/25/2025] [Indexed: 03/09/2025] Open
Abstract
We designed, developed, and clinically tested two rapid antigen-based immunosensors for SARS-CoV-2 detection, enabling diagnosis and viral load quantification for under USD $2. In a first clinical study, a screen-printed disposable carbon-based (SPC) sensor was assessed on prospectively recruited adult participants classified into three study groups: healthy donors (n = 46); SARS-CoV-2-infected symptomatic patients (n = 58); and co-habitants of patients without prior testing (n = 38). Nasopharyngeal aspirates (NA), oropharyngeal swabs (OS), and saliva (SA) samples were obtained from all participants. Performance was measured in terms of clinical sensitivity and specificity against a reference diagnostic RT-qPCR kit and analytical sensitivity (limit of detection, LoD) and specificity using recombinant material in lab tests. A second study was performed using the same sensor design, albeit with laser-induced graphene (LIG) electrodes, using nasopharyngeal swabs (NS) on 224 patient samples obtained at different stages of the pandemic, of which 110 tested negative and 114 positive via RT-qPCR. We find OS was the most informative sample, when compared to NA and SA. The SPC-based sensors had a 93.8% sensitivity and 61.5% specificity with OS samples, while the LIG-based sensors with NS had a lower sensitivity of 68.93%, albeit a significantly higher specificity of 86.17%. We believe specificity values for the SPC sensors were driven by positive results from co-habitants and healthy donors and were affected by the low sensitivity (75.5%) and high LoD (> 20,000 viral copies/mL) of the reference RT-qPCR kit used, and the lower sensitivity of the LIG-based was due to a reduced set of effective antigen-binding sites caused by the non-covalent LIG-mAb ligands used. The immunosensor's LoD to spike protein in phosphate-buffered saline (PBS) for both types of sensors was near 1 fg/mL and showed no cross-reactivity to recombinant structural proteins of Epstein-Barr and Influenza. Performance metrics and time-to-result (5 < 12 min) provide proof-of-principle of the immunosensor's applicability as a low-cost, rapid technology for determining SARS-CoV-2 infections. Changing the working electrode material to LIG, instead of SPC, improved specificity even in the presence of pathogen variants. Discordant results between our two immunosensor versions and RT-qPCR tests are attributed not only to limited antibody effectiveness in the former but also to the quality of RT-qPCR probes used at the height of the pandemic.
Collapse
Affiliation(s)
- Olga L Agudelo
- Centro de Investigaciones Clínicas (CIC), Fundación Valle del Lili, Cra 98 No. 18 - 49, 760032, Cali, Colombia
| | | | - Lina Giraldo-Parra
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Universidad ICESI, Cali, Colombia
| | - Mariana Rosales-Chilama
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Universidad ICESI, Cali, Colombia
| | - Sammy Perdomo
- iOMICAS, Pontificia Universidad Javeriana, Calle 18 # 118-250, Cali, Colombia
| | - María Adelaida Gómez
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Universidad ICESI, Cali, Colombia
| | - John W Rodriguez
- iOMICAS, Pontificia Universidad Javeriana, Calle 18 # 118-250, Cali, Colombia
| | - Viviana Ortega
- iOMICAS, Pontificia Universidad Javeriana, Calle 18 # 118-250, Cali, Colombia
| | - Carlos F Daza Rivera
- Gobernación del Valle del Cauca, Secretaria de Salud Departamental, Laboratorio de Salud Pública Departamental del Valle, Cra. 76 #4-70, Cali, Valle del Cauca, Colombia
| | - Diana Galindo
- Gobernación del Valle del Cauca, Secretaria de Salud Departamental, Laboratorio de Salud Pública Departamental del Valle, Cra. 76 #4-70, Cali, Valle del Cauca, Colombia
| | - Drochss P Valencia
- iOMICAS, Pontificia Universidad Javeriana, Calle 18 # 118-250, Cali, Colombia
| | - Mauricio Quimbaya
- iOMICAS, Pontificia Universidad Javeriana, Calle 18 # 118-250, Cali, Colombia
| | - Simón Plata
- iOMICAS, Pontificia Universidad Javeriana, Calle 18 # 118-250, Cali, Colombia
| | - Robert Bogdanowicz
- Faculty of Electronics, Telecommunications and Informatics, Gdansk University of Technology, Narutowicza Str.11/12, 80-233, Gdansk, Poland
| | - Fernando Rosso
- Centro de Investigaciones Clínicas (CIC), Fundación Valle del Lili, Cra 98 No. 18 - 49, 760032, Cali, Colombia
- Universidad ICESI, Cali, Colombia
| | - Andres Jaramillo-Botero
- iOMICAS, Pontificia Universidad Javeriana, Calle 18 # 118-250, Cali, Colombia.
- Chemistry and Chemical Engineering, California Institute of Technology, 1200 E California Blvd, Pasadena, CA, 91125, USA.
| |
Collapse
|
7
|
Xi B, Hua Z, Jiang D, Chen Z, Wei J, Meng Y, Du H. Within-Host Fitness and Antigenicity Shift Are Key Factors Influencing the Prevalence of Within-Host Variations in the SARS-CoV-2 S Gene. Viruses 2025; 17:362. [PMID: 40143291 PMCID: PMC11945823 DOI: 10.3390/v17030362] [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: 01/27/2025] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
Within-host evolution plays a critical role in shaping the diversity of SARS-CoV-2. However, understanding the primary factors contributing to the prevalence of intra-host single nucleotide variants (iSNVs) in the viral population remains elusive. Here, we conducted a comprehensive analysis of over 556,000 SARS-CoV-2 sequencing data and prevalence data of different SARS-CoV-2 S protein amino acid mutations to elucidate key factors influencing the prevalence of iSNVs in the SARS-CoV-2 S gene. Within-host diversity analysis revealed the presence of mutational hotspots within the S gene, mainly located in NTD, RBD, TM, and CT domains. Additionally, we generated a single amino acid resolution selection status map of the S protein. We observed a significant variance in within-host fitness among iSNVs in the S protein. The majority of iSNVs exhibited low to no within-host fitness and displayed low alternate allele frequency (AAF), suggesting that they will be eliminated due to the narrow transmission bottleneck of SARS-CoV-2. Notably, iSNVs with moderate AAFs (0.06-0.12) were found to be more prevalent than those with high AAFs. Furthermore, iSNVs with the potential to alter antigenicity were more prevalent. These findings underscore the significance of within-host fitness and antigenicity shift as two key factors influencing the prevalence of iSNVs in the SARS-CoV-2 S gene.
Collapse
Affiliation(s)
- Binbin Xi
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Zhihao Hua
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Dawei Jiang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Zixi Chen
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Jinfen Wei
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Yuhuan Meng
- Guangzhou KingMed Transformative Medicine Institute, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou 510220, China
| | - Hongli Du
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
8
|
Ge J, Far AT, Digitale JC, Pletcher MJ, Lai JC. Decreasing Case Fatality Rates for Patients With Cirrhosis Infected With SARS-CoV-2: A National COVID Cohort Collaborative Study. Clin Gastroenterol Hepatol 2025; 23:591-601.e2. [PMID: 39181420 PMCID: PMC11917370 DOI: 10.1016/j.cgh.2024.07.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND & AIMS The virulence and severity of SARS-CoV-2 infections have decreased over time in the general population due to vaccinations and improved antiviral treatments. Whether a similar trend has occurred in patients with cirrhosis is unclear. We used the National COVID Cohort Collaborative (N3C) to describe the outcomes over time. METHODS We utilized the N3C level 3 data set with uncensored dates to identify all patients with chronic liver disease (CLD) with and without cirrhosis who had SARS-CoV-2 infection as of November 2023. We described the observed 30-day case fatality rate (CFR) by month of infection. We used adjusted survival analyses to calculate relative hazard of death by month of infection compared with infection at the onset of the COVID-19 pandemic. RESULTS We identified 117,811 total patients with CLD infected with SARS-CoV-2 between March 2020 and November 2023: 27,428 (23%) with cirrhosis and 90,383 (77%) without cirrhosis. The observed 30-day CFRs during the entire study period were 1.1% (1016) for patients with CLD without cirrhosis and 6.3% (1732) with cirrhosis. Observed 30-day CFRs by month of infection varied throughout the pandemic and showed a sustained downward trend since 2022. Compared with infection in Quarter 2 of 2020 (at the beginning of the pandemic), the adjusted hazards of death at 30 days for infection in Quarter 3 of 2023 were 0.20 (95% confidence interval [CI], 0.08-0.50) for patients with CLD without cirrhosis and 0.35 (95% CI, 0.18-0.69) for patients with CLD with cirrhosis. CONCLUSIONS In this N3C study, we found that the observed 30-day CFR decreased progressively for patients with CLD both with and without cirrhosis, consistent with broader trends seen in the general population.
Collapse
Affiliation(s)
- Jin Ge
- Division of Gastroenterology and Hepatology, Department of Medicine, University of California - San Francisco, San Francisco, California.
| | - Aryana T Far
- Division of Gastroenterology and Hepatology, Department of Medicine, University of California - San Francisco, San Francisco, California
| | - Jean C Digitale
- Department of Epidemiology and Biostatistics, University of California - San Francisco, San Francisco, California
| | - Mark J Pletcher
- Department of Epidemiology and Biostatistics, University of California - San Francisco, San Francisco, California
| | - Jennifer C Lai
- Division of Gastroenterology and Hepatology, Department of Medicine, University of California - San Francisco, San Francisco, California
| |
Collapse
|
9
|
Kabir MP, Mercier É, Eid W, Plaza-Diaz J, D'Aoust PM, Landgraff C, Goodridge L, Lawal OU, Wan S, Hegazy N, Nguyen T, Wong C, Thakali O, Pisharody L, Stephenson S, Graber TE, Delatolla R. Diagnostic performance of allele-specific RT-qPCR and genomic sequencing in wastewater-based surveillance of SARS-CoV-2. ECO-ENVIRONMENT & HEALTH 2025; 4:100135. [DOI: 10.1016/j.eehl.2025.100135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/07/2025]
|
10
|
Cilloniz C, Torres A. Coronavirus Disease 2019 and Emerging Lung Infections in the Immunocompromised Patient. Clin Chest Med 2025; 46:203-216. [PMID: 39890289 DOI: 10.1016/j.ccm.2024.10.015] [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: 02/03/2025]
Abstract
The immunocompromised are at higher risk of COVID-19 and lung infections, and these are associated with more severe presentations and greater risk of complication, increasing the risks of intensive care unit admission and poor outcomes. However, only limited high-quality data are available about the diagnosis and management of lung infections in this population, with many clinical trials and other large studies excluding the immunocompromised. Well-designed studies are needed to better understand the optimal diagnostic and management options to improve outcomes in the increasingly heterogeneous group of immunocompromised patients.
Collapse
Affiliation(s)
- Catia Cilloniz
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Carrer del Rosselló, 149, Barcelona, Spain; Ciber de Enfermedades Respiratorias (Ciberes) Barcelona, Spain; Faculty of Health Sciences, Continental University, Av San Carlos 1980, Huancayo, Peru. https://twitter.com/catiacilloniz
| | - Antoni Torres
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Carrer del Rosselló, 149, Barcelona, Spain; Ciber de Enfermedades Respiratorias (Ciberes) Barcelona, Spain; Pulmonary Department, Hospital Clinic of Barcelona, Villarroel 170, Barcelona, Spain.
| |
Collapse
|
11
|
Hou X, Zhai L, Fu L, Lu J, Guo P, Zhang Y, Zheng D, Ma G. Advances in Engineered Phages for Disease Treatment. SMALL METHODS 2025:e2401611. [PMID: 39935185 DOI: 10.1002/smtd.202401611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 01/15/2025] [Indexed: 02/13/2025]
Abstract
Phage therapy presents a promising solution for combating multidrug-resistant (MDR) bacterial infections and other bacteria-related diseases, attributed to their innate ability to target and lyse bacteria. Recent clinical successes, particularly in treating MDR-related respiratory and post-surgical infections, validated the therapeutic potential of phage therapy. However, the complex microenvironment within the human body poses significant challenges to phage activity and efficacy in vivo. To overcome these barriers, recent advances in phage engineering have aimed to enhance targeting accuracy, improve stability and survivability, and explore synergistic combinations with other therapeutic modalities. This review provides a comprehensive overview of phage therapy, emphasizing the application of engineered phages in antibacterial therapy, tumor therapy, and vaccine development. Furthermore, the review highlights the current challenges and future trends for advancing phage therapy toward broader clinical applications.
Collapse
Affiliation(s)
- Xiaolin Hou
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
| | - Lin Zhai
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Laiying Fu
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Junna Lu
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
| | - Peilin Guo
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yu Zhang
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Diwei Zheng
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Guanghui Ma
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
12
|
Popović ME, Tadić V, Popović M. (R)evolution of Viruses: Introduction to biothermodynamics of viruses. Virology 2025; 603:110319. [PMID: 39642612 DOI: 10.1016/j.virol.2024.110319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/15/2024] [Accepted: 11/26/2024] [Indexed: 12/09/2024]
Abstract
As of 26 April 2024, the International Committee on Taxonomy of Viruses has registered 14690 virus species. Of these, only several dozen have been chemically and thermodynamically characterized. Every virus species is characterized by a specific empirical formula and thermodynamic properties - enthalpy, entropy and Gibbs energy. These physical properties are used in a mechanistic model of virus-host interactions at the cell membrane and in the cytoplasm. This review article presents empirical formulas and Gibbs energies for all major variants of SARS-CoV-2. This article also reports and suggests a mechanistic model of evolutionary changes, with the example of time evolution of SARS-CoV-2 from 2019 to 2024.
Collapse
Affiliation(s)
- Marko E Popović
- University of Belgrade, Institute of Chemistry, Technology and Metallurgy, Njegoševa 12, 11000, Belgrade, Serbia.
| | - Vojin Tadić
- Department for Experimental Testing of Precious Metals, Mining and Metallurgy Institute, Zeleni Bulevar 35, 19210, Bor, Serbia
| | - Marta Popović
- University of Belgrade, Faculty of Biology, Studentski trg 16, 11000, Belgrade, Serbia
| |
Collapse
|
13
|
Kiran NS, Singh S, Yashaswini C, Prajapati BG. Revisiting the potential of natural antimicrobial peptides against emerging respiratory viral disease: a review. 3 Biotech 2025; 15:40. [PMID: 39816617 PMCID: PMC11729606 DOI: 10.1007/s13205-024-04184-3] [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: 07/02/2024] [Accepted: 11/21/2024] [Indexed: 01/18/2025] Open
Abstract
This review assesses the antiviral capabilities of antimicrobial peptides (AMPs) against SARS-CoV-2 and other respiratory viruses, focussing on their therapeutic potential. AMPs, derived from natural sources, exhibit promising antiviral properties by disrupting viral membranes, inhibiting viral entry, and modulating host immune responses. Preclinical studies demonstrate that peptides such as defensins, cathelicidins, and lactoferrin can effectively reduce SARS-CoV-2 replication and inhibit viral spread. In addition, AMPs have shown potential in enhancing the host's antiviral immunity. Despite these promising outcomes, several challenges require assessments before transforming into clinical translation. Several issues related to peptide stability, cytotoxicity, and efficient delivery systems pose significant limitations to their therapeutic application. Recent advancements in peptide engineering, nanotechnology-based delivery systems, and peptide conjugation strategies have improved AMPs stability and bioavailability; however, further optimization is essential. Moreover, whilst AMPs are safe, their effects on host cells and tissues need a thorough investigation to minimise potential adverse reactions. This review concludes that whilst AMPs present a promising route for antiviral therapies, particularly in targeting SARS-CoV-2, extensive clinical trials and additional studies are required to overcome current limitations. Future research should focus on developing more stable, less toxic AMPs formulations with enhanced delivery mechanisms, aiming to integrate AMPs into viable therapeutic options for respiratory viral diseases, including COVID-19 and other emerging infections.
Collapse
Affiliation(s)
| | - Sudarshan Singh
- Office of Research Administration, Chiang Mai University, Chiang Mai, 50200 Thailand
- Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Chandrashekar Yashaswini
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru, Karnataka 560064 India
| | - Bhupendra G. Prajapati
- Shree. S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva, Gujarat 384012 India
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000 Thailand
| |
Collapse
|
14
|
Alhamlan FS, Al-Qahtani AA. SARS-CoV-2 Variants: Genetic Insights, Epidemiological Tracking, and Implications for Vaccine Strategies. Int J Mol Sci 2025; 26:1263. [PMID: 39941026 PMCID: PMC11818319 DOI: 10.3390/ijms26031263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/04/2025] [Accepted: 01/09/2025] [Indexed: 02/16/2025] Open
Abstract
The emergence of SARS-CoV-2 variants has significantly impacted the global response to the COVID-19 pandemic. This review examines the genetic diversity of SARS-CoV-2 variants, their roles in epidemiological tracking, and their influence on viral fitness. Variants of concern (VOCs) such as Alpha, Beta, Gamma, Delta, and Omicron have demonstrated increased transmissibility, altered pathogenicity, and potential resistance to neutralizing antibodies. Epidemiological tracking of these variants is crucial for understanding their spread, informing public health interventions, and guiding vaccine development. The review also explores how specific mutations in the spike protein and other genomic regions contribute to viral fitness, affecting replication efficiency, immune escape, and transmission dynamics. By integrating genomic surveillance data with epidemiological and clinical findings, this review provides a comprehensive overview of the ongoing evolution of SARS-CoV-2 and its implications for public health strategies and new vaccine development.
Collapse
Affiliation(s)
- Fatimah S. Alhamlan
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, 11211 Riyadh, Saudi Arabia;
- Department of Microbiology and Immunology, College of Medicine, Alfaisal University, 11211 Riyadh, Saudi Arabia
| | - Ahmed A. Al-Qahtani
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, 11211 Riyadh, Saudi Arabia;
- Department of Microbiology and Immunology, College of Medicine, Alfaisal University, 11211 Riyadh, Saudi Arabia
| |
Collapse
|
15
|
Berleant JD, Banal JL, Rao DK, Bathe M. Enabling global-scale nucleic acid repositories through versatile, scalable biochemical selection from room-temperature archives. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.04.12.24305660. [PMID: 38699348 PMCID: PMC11064994 DOI: 10.1101/2024.04.12.24305660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Conventional collection, preservation, and retrieval of nucleic acid specimens, particularly unstable RNA, require costly cold-chain infrastructure and rely on inefficient robotic sample handling, hindering downstream analyses. These generate critical bottlenecks for global pathogen surveillance and genomic biobanking efforts, prohibiting large-scale nucleic acid sample collection and analyses that are needed to empower pathogen tracing, as well as rare disease diagnostics1. Here, we introduce a scalable nucleic acid storage system that enables rapid and precise retrieval on pooled nucleic acid samples-stored at room-temperature with minimal physical footprint2,3-using versatile database-like queries on barcoded, encapsulated samples. Queries can incorporate numerical ranges, categorical filters, and combinations thereof, which is a significant advancement beyond previous demonstrations limited to single-sample retrieval or Boolean classifiers. We apply our system to a pool of ninety-six mock SARS-CoV-2 genomic samples identified with theoretical patient data including patient age, geographic location, and diagnostic state, allowing rapid, multiplexed nucleic acid sample retrieval in a scalable manner to empower genomic analyses. By avoiding expensive and cumbersome freezer storage and retrieval systems, our approach in principle scales to millions of samples without loss of fidelity or throughput, thereby supporting the development of large-scale pathogen and genomic repositories in under-resourced or isolated regions of the US and worldwide.
Collapse
Affiliation(s)
- Joseph D. Berleant
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - James L. Banal
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Present address: Cache DNA, Inc. 733 Industrial Rd., San Carlos, CA 94070 USA
| | | | - Mark Bathe
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02139 USA
| |
Collapse
|
16
|
Montone RA, Rinaldi R, Masciocchi C, Lilli L, Damiani A, La Vecchia G, Iannaccone G, Basile M, Salzillo C, Caffè A, Bonanni A, De Pascale G, Grieco DL, Tanzarella ES, Buonsenso D, Murri R, Fantoni M, Liuzzo G, Sanna T, Richeldi L, Sanguinetti M, Massetti M, Trani C, Tshomba Y, Gasbarrini A, Valentini V, Antonelli M, Crea F. Vaccines and myocardial injury in patients hospitalized for COVID-19 infection: the CardioCOVID-Gemelli study. EUROPEAN HEART JOURNAL. QUALITY OF CARE & CLINICAL OUTCOMES 2025; 11:59-67. [PMID: 38414273 PMCID: PMC11736151 DOI: 10.1093/ehjqcco/qcae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 02/06/2024] [Accepted: 02/26/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND Myocardial injury is prevalent among patients hospitalized for COVID-19. However, the role of COVID-19 vaccines in modifying the risk of myocardial injury is unknown. AIMS To assess the role of vaccines in modifying the risk of myocardial injury in COVID-19. METHODS AND RESULTS We enrolled COVID-19 patients admitted from March 2021 to February 2022 with known vaccination status and ≥1 assessment of hs-cTnI within 30 days from the admission. The primary endpoint was the occurrence of myocardial injury (hs-cTnI levels >99th percentile upper reference limit). A total of 1019 patients were included (mean age: 67.7 ± 14.8 years, 60.8% male, and 34.5% vaccinated against COVID-19). Myocardial injury occurred in 145 (14.2%) patients. At multivariate logistic regression analysis, advanced age, chronic kidney disease, and hypertension, but not vaccination status, were independent predictors of myocardial injury. In the analysis according to age tertiles distribution, myocardial injury occurred more frequently in the III tertile (≥76 years) compared with other tertiles (I tertile: ≤60 years; II tertile: 61-75 years) (P < 0.001). Moreover, in the III tertile, vaccination was protective against myocardial injury [odds ratio (OR): 0.57, 95% confidence interval (CI): 0.34-0.94; P = 0.03], while a previous history of coronary artery disease was an independent positive predictor. In contrast, in the I tertile, chronic kidney disease (OR: 6.94, 95% CI: 1.31-36.79, P = 0.02) and vaccination (OR: 4.44, 95% CI: 1.28-15.34, P = 0.02) were independent positive predictors of myocardial injury. CONCLUSION In patients ≥76 years, COVID-19 vaccines were protective for the occurrence of myocardial injury, while in patients ≤60 years, myocardial injury was associated with previous COVID-19 vaccination. Further studies are warranted to clarify the underlying mechanisms.
Collapse
Affiliation(s)
- Rocco Antonio Montone
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Riccardo Rinaldi
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Carlotta Masciocchi
- Real World Data Facility, Gemelli Generator, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Livia Lilli
- Real World Data Facility, Gemelli Generator, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Andrea Damiani
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
| | - Giulia La Vecchia
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Giulia Iannaccone
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Mattia Basile
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Carmine Salzillo
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Andrea Caffè
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Alice Bonanni
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Gennaro De Pascale
- Department of Emergency, Intensive Care Medicine and Anaesthesia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Istituto di Anestesiologia e Rianimazione, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Domenico Luca Grieco
- Department of Emergency, Intensive Care Medicine and Anaesthesia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Eloisa Sofia Tanzarella
- Department of Emergency, Intensive Care Medicine and Anaesthesia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Danilo Buonsenso
- Department of Women's Health, Child Health and Public Health Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Rita Murri
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
- Clinic of Infectious Diseases, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Massimo Fantoni
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
- Clinic of Infectious Diseases, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Giovanna Liuzzo
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Tommaso Sanna
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Luca Richeldi
- Division of Pulmonary Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Maurizio Sanguinetti
- Department of Basic Biotechnological Sciences, Intensive and Perioperative Clinics, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Massimo Massetti
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Carlo Trani
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Yamume Tshomba
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Vincenzo Valentini
- Department of Diagnostic Imaging, Radiotherapy, Oncology and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Radiological and Hematological Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Massimo Antonelli
- Department of Emergency, Intensive Care Medicine and Anaesthesia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Istituto di Anestesiologia e Rianimazione, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Filippo Crea
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy
| |
Collapse
|
17
|
Okwumabua O, Bradley-Siemens N, Cruz C, Chittick L, Thompson M. Detection of SARS-CoV-2 and a possible variant in shelter cats. PLoS One 2025; 20:e0317104. [PMID: 39804893 PMCID: PMC11730420 DOI: 10.1371/journal.pone.0317104] [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: 07/22/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
SARS-CoV-2 is the cause of mild to severe acute respiratory disease that led to significant loss of human lives worldwide between 2019 and 2022. The virus has been detected in various animals including cats and dogs making it a major public health concern and a One Health issue. In this study, conjunctival and pharyngeal swabs (n = 350) and serum samples (n = 350) were collected between July and December 2020 from cats that were housed in an animal shelter and tested for the infection of SARS-CoV-2 using real time reverse-transcription polymerase chain reaction (rRT-PCR) that targeted the N1 and N2 genes, and a SARS-CoV-2 surrogate virus neutralization Test (sVNT), respectively. 203 (58%) swab samples were negative (N1 and N2 not detected), 2 (0.6%) were positive (N1 and N2 detected) and 145 (41%) were inconclusive (only N1 detected). Analysis of the N2 region and multiple sequence alignment revealed base-pair deletions and substitutions in the N2 probe binding region of the feline samples RNA extracts in comparison with the positive control and human SARS-CoV-2 sequences in the GenBank database. Substituting the N2 probe with a probe derived from the cat sample amplicon sequences, 123 of 127 (96.9%) of the N2 negative samples returned positive. All but one of the 350 serum samples were negative for SARS-CoV-2 antibody. These observations indicated that although detection of SARS-CoV-2 infection was low in the samples tested, pet cats can harbor the virus and serve as potential source for virus spread that may lead to human infections. Additionally, cats may harbor a yet-to-be described virus that is somewhat related to SARS-CoV-2.
Collapse
Affiliation(s)
- Ogi Okwumabua
- Department of Pathology and Population Medicine, College of Veterinary Medicine, Midwestern University, Glendale, Arizona, United States of America
| | - Nancy Bradley-Siemens
- Department of Small Animal Shelter and Community Medicine, College of Veterinary Medicine, Midwestern University, Glendale, Arizona, United States of America
| | - Catherine Cruz
- Department of Pathology and Population Medicine, College of Veterinary Medicine, Midwestern University, Glendale, Arizona, United States of America
| | - Lauren Chittick
- Department of Pathology and Population Medicine, College of Veterinary Medicine, Midwestern University, Glendale, Arizona, United States of America
| | - Melissa Thompson
- Arizona Humane Society, Phoenix, Arizona, United States of America
| |
Collapse
|
18
|
Tiniakou E, Casciola‐Rosen L, Thomas MA, Manabe Y, Antar AAR, Damarla M, Hassoun PM, Gao L, Wang Z, Zeger S, Rosen A. Autoantibodies in hospitalised patients with COVID-19. Clin Transl Immunology 2024; 13:e70019. [PMID: 39734590 PMCID: PMC11671454 DOI: 10.1002/cti2.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 10/07/2024] [Accepted: 11/13/2024] [Indexed: 12/31/2024] Open
Abstract
Objectives CD209L and its homologous protein CD209 act as alternative entry receptors for the SARS-CoV-2 virus and are highly expressed in the virally targeted tissues. We tested for the presence and clinical features of autoantibodies targeting these receptors and compared these with autoantibodies known to be associated with COVID-19. Methods Using banked samples (n = 118) from Johns Hopkins patients hospitalised with COVID-19, we defined autoantibodies against CD209 and CD209L by enzyme-linked immunosorbent assay (ELISA). Clinical associations of these antibodies were compared with those of patients with anti-interferon (IFN) and anti-angiotensin-converting enzyme-2 (ACE2) autoantibodies. Results Amongst patients hospitalised with COVID-19, 19.5% (23/118) had IgM autoantibodies against CD209L and were more likely to have coronary artery disease (44% vs 19%, P = 0.03). Antibodies against CD209 were present in 5.9% (7/118); interestingly, all 7 were male (P = 0.02). In our study, the presence of either antibody was positively associated with disease severity [OR 95% confidence interval (95% CI): 1.80 (0.69-5.03)], but the association did not reach statistical significance. In contrast, 10/118 (8.5%) had IgG autoantibodies against IFNα, and 21 (17.8%) had IgM antibodies against ACE2. These patients had significantly worse prognosis (intubation or death) and prolonged hospital stays. However, when adjusting for patient characteristics on admission, only the presence of anti-ACE2 IgM remained significant [pooled common OR (95% CI), 4.14 (1.37, 12.54)]. Conclusion We describe IgM autoantibodies against CD209 and CD209L amongst patients hospitalised with COVID-19. These were not associated with disease severity. Conversely, patients with either anti-ACE2 IgM or anti-IFNα IgG antibodies had worse outcomes. Due to the small size of the study cohort, conclusions drawn should be considered cautiously.
Collapse
Affiliation(s)
- Eleni Tiniakou
- Division of Rheumatology, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Livia Casciola‐Rosen
- Division of Rheumatology, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Mekha A Thomas
- Division of Rheumatology, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Yuka Manabe
- Division of Infectious Diseases, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Annukka AR Antar
- Division of Infectious Diseases, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Mahendra Damarla
- Division of Pulmonary and Critical Care, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Paul M Hassoun
- Division of Pulmonary and Critical Care, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Li Gao
- Division of Allergy and Immunology, Department of MedicineJohns Hopkins University, School of MedicineBaltimoreMDUSA
| | - Zitong Wang
- Department of BiostatisticsBloomberg School of Public HealthBaltimoreMDUSA
| | - Scott Zeger
- Department of BiostatisticsBloomberg School of Public HealthBaltimoreMDUSA
| | - Antony Rosen
- Division of Rheumatology, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| |
Collapse
|
19
|
Gevorgyan S, Khachatryan H, Shavina A, Gharaghani S, Zakaryan H. Targeting SARS-CoV-2 main protease: a comprehensive approach using advanced virtual screening, molecular dynamics, and in vitro validation. Virol J 2024; 21:330. [PMID: 39707350 DOI: 10.1186/s12985-024-02607-4] [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: 07/15/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024] Open
Abstract
The COVID-19 pandemic, driven by the SARS-CoV-2 virus, necessitates the development of effective therapeutics. The main protease of the virus, Mpro, is a key target due to its crucial role in viral replication. Our study presents a novel approach combining ligand-based pharmacophore modeling with structure-based advanced virtual screening to identify potential inhibitors of Mpro. We screened around 200 million compounds using this integrated methodology, resulting in a shortlist of promising compounds. These were further scrutinized through molecular dynamics simulations, revealing their interaction dynamics with Mpro. Subsequent in vitro assays using the Mpro enzyme identified two compounds exhibiting significant micromolar inhibitory activity. These findings provide valuable scaffolds for the development of advanced therapeutics targeting Mpro. The comprehensive nature of our approach, spanning computational predictions to experimental validations, offers a robust pathway for rapid and efficient identification of potential drug candidates against COVID-19.
Collapse
Affiliation(s)
- Smbat Gevorgyan
- Laboratory of Antiviral Drug Discovery. Institute of Molecular Biology of National Academy of Sciences, 0014, Yerevan, Armenia.
- Denovo Sciences Inc, 0060, Yerevan, Armenia.
| | - Hamlet Khachatryan
- Laboratory of Antiviral Drug Discovery. Institute of Molecular Biology of National Academy of Sciences, 0014, Yerevan, Armenia
- Denovo Sciences Inc, 0060, Yerevan, Armenia
| | - Anastasiya Shavina
- Laboratory of Antiviral Drug Discovery. Institute of Molecular Biology of National Academy of Sciences, 0014, Yerevan, Armenia
- Denovo Sciences Inc, 0060, Yerevan, Armenia
| | - Sajjad Gharaghani
- Denovo Sciences Inc, 0060, Yerevan, Armenia
- Laboratory of Bioinformatics and Drug Design (LBD), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Hovakim Zakaryan
- Laboratory of Antiviral Drug Discovery. Institute of Molecular Biology of National Academy of Sciences, 0014, Yerevan, Armenia
- Denovo Sciences Inc, 0060, Yerevan, Armenia
| |
Collapse
|
20
|
Goux H, Green J, Wilson A, Sozhamannan S, Richard SA, Colombo R, Lindholm DA, Jones MU, Agan BK, Larson D, Saunders DL, Mody R, Cox J, Deans R, Walish J, Fries A, Simons MP, Pollett SD, Smith DR. Performance of rapid antigen tests to detect SARS-CoV-2 variant diversity and correlation with viral culture positivity: implication for diagnostic development and future public health strategies. mBio 2024; 15:e0273724. [PMID: 39480114 PMCID: PMC11633148 DOI: 10.1128/mbio.02737-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/02/2024] [Indexed: 11/02/2024] Open
Abstract
Antigen-based rapid diagnostic tests (Ag-RDTs) provide timely results, are simple to use, and are less expensive than molecular assays. Recent studies suggest that antigen-based testing aligns with virus culture-based results (a proxy of contagiousness at the peak viral phase of illness); however, the performance of Ag-RDTs for newer SARS-CoV-2 variants is unclear. In this study, we (i) assessed the performance of Ag-RDTs and diagnostic antibodies to detect a range of SARS-CoV-2 variants and (ii) determined whether Ag-RDT results correlated with culture positivity. We noted only minor differences in the limit of detection by variant for all assays, and we demonstrated consistent antibody affinity to the N protein among the different variants. We observed moderate to high sensitivity (46.8%-83.9%) for Ag-RDTs when compared to PCR positivity (100%), and all variants were assessed on each assay. Ag-RDT sensitivity and PCR Ct showed an inverse correlation with the detection of viable virus. Collectively, our results demonstrate that commercially available Ag-RDTs offer variable sensitivity compared to PCR, show similar diagnostic validity across variants, and may predict the risk of transmissibility. These findings may be used to support more tailored SARS-CoV-2 isolation strategies, particularly if other studies clarify the direct association between Ag-RDT positivity and transmission risk. The apparent trade-off between sensitivity in the detection of any PCR-positive infection and concordance with infectious virus positivity may also inform new RDT diagnostic development strategies for SARS-CoV-2 and other epidemic respiratory pathogens. IMPORTANCE Despite the availability of vaccines, COVID-19 continues to be a major health concern, and antigen-based rapid diagnostic tests (Ag-RDTs) are commonly used as point-of-care or at-home diagnostic tests. In this study, we evaluated the performance of two commercially available Ag-RDTs and a research Ag-RDT to detect multiple SARS-CoV-2 variants using upper respiratory tract swab samples from clinical COVID-19 cases. Furthermore, we determined whether Ag-RDT results correlated with culture positivity, a potential proxy of viral transmissibility. Our results have important implications to inform future testing and response strategies during periods of high COVID-19 transmission with new variants.
Collapse
Affiliation(s)
- Heather Goux
- Microbiology and Immunology Department, Biological Defense Research Directorate, Naval Medical Research Command, Fort Detrick, Maryland, USA
| | - Jennetta Green
- Microbiology and Immunology Department, Biological Defense Research Directorate, Naval Medical Research Command, Fort Detrick, Maryland, USA
| | - Andrew Wilson
- Microbiology and Immunology Department, Biological Defense Research Directorate, Naval Medical Research Command, Fort Detrick, Maryland, USA
| | - Shanmuga Sozhamannan
- Joint Program Executive Office for Chemical, Biological, Radiological and Nuclear Defense (JPEO-CBRND), Joint Project Lead for CBRND Enabling Biotechnologies, Frederick, Maryland, USA
- Joint Research and Development, Inc., Stafford, Virginia, USA
| | - Stephanie A. Richard
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Rhonda Colombo
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Madigan Army Medical Center, Joint Base Lewis McChord, Washington, USA
| | - David A. Lindholm
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Brooke Army Medical Center, Joint Base San Antonio-Fort Sam Houston, San Antonio, Texas, USA
| | - Milissa U. Jones
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Brian K. Agan
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Derek Larson
- Naval Medical Center, San Diego, California, USA
| | - David L. Saunders
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Rupal Mody
- William Beaumont Army Medical Center, El Paso, Texas, USA
| | - Jason Cox
- C2Sense, Inc., Watertown, Massachusetts, USA
| | | | | | - Anthony Fries
- US Air Force School of Aerospace Medicine, Dayton, Ohio, USA
| | - Mark P. Simons
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Simon D. Pollett
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Darci R. Smith
- Microbiology and Immunology Department, Biological Defense Research Directorate, Naval Medical Research Command, Fort Detrick, Maryland, USA
| |
Collapse
|
21
|
Yu H, Chen W, Shu J, Wu X, Quan J, Cheng H, Bao X, Wu D, Wang X, Li Z. Key β1-4 galactosylated glycan receptors of SARS-CoV-2 and its inhibitor from the galactosylated glycoproteins of bovine milk. J Adv Res 2024:S2090-1232(24)00566-6. [PMID: 39667665 DOI: 10.1016/j.jare.2024.12.010] [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: 05/12/2024] [Revised: 08/22/2024] [Accepted: 12/05/2024] [Indexed: 12/14/2024] Open
Abstract
INTRODUCTION The binding of the spike (S) protein of SARS-CoV-2 to angiotensin-converting enzyme 2 (ACE2) is a critical stage in the process of infection. While previous studies indicated that the S protein and ACE2 are extensively glycosylated, the functions of glycans in their interactions remain uncertain. OBJECTIVES This study aimed to investigate the glycan receptors of SARS-CoV-2 and evaluate the inhibitory effects of galactosylated glycoproteins derived from bovine milk on the attachment of SARS-CoV-2 pseudovirus. METHODS An antibody-overlay lectin microarray was used to profile the glycopatterns of the S protein-S1 of SARS-CoV-2 and ACE2. Molecular dynamics simulation was used to mimic the interaction between the S protein and ACE2. The effects of N-glycans and β1-4 galactosylation on the interactions between SARS-CoV-2, its variations (B1.617.2 (Delta) and B1.1.529 (Omicron)), and ACE2 was assessed using molecular docking simulation and protein microarrays. The impact of glycoproteins (specifically sialylated glycoproteins or de-sialylated glycoproteins) derived from bovine milk on the interaction between S1 and ACE2, as well as on pseudoviral attachment and entry, was assessed using protein microarrays and pseudovirus-based microneutralization assays. RESULTS Our findings indicated that the galactosylated glycoforms were the most prevalent for both S1 and ACE2. Importantly, we demonstrated that the β1-4 galactosylated N-glycans of ACE2 played a crucial role in the binding of S1 of SARS-CoV-2 and its variations to ACE2. The glycoproteins derived from bovine milk had a large amount of galactosylated glycans, which are comparable to the glycoforms of ACE2. The glycoproteins effectively blocked the attachment and entry of the SARS-CoV-2 pseudovirus by competitively blocking the binding of S1 to ACE2. CONCLUSIONS Our findings demonstrated that the β1-4 galactosylated N-glycans of ACE2 play a crucial role as glycan receptors for the binding of S1 of SARS-CoV-2 and its variations. Moreover, the glycoproteins with 'receptor-like' glycoforms could be an effective inhibitor to prevent SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Hanjie Yu
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Wentian Chen
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Jian Shu
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Xin Wu
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Jia Quan
- College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Hongwei Cheng
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Xiaojuan Bao
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Di Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega- Science, CAS, Wuhan 430071, China
| | - Xilong Wang
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Zheng Li
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an 710069, China.
| |
Collapse
|
22
|
Dhamotharan K, Korn SM, Wacker A, Becker MA, Günther S, Schwalbe H, Schlundt A. A core network in the SARS-CoV-2 nucleocapsid NTD mediates structural integrity and selective RNA-binding. Nat Commun 2024; 15:10656. [PMID: 39653699 PMCID: PMC11628620 DOI: 10.1038/s41467-024-55024-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024] Open
Abstract
The SARS-CoV-2 nucleocapsid protein is indispensable for viral RNA genome processing. Although the N-terminal domain (NTD) is suggested to mediate specific RNA-interactions, high-resolution structures with viral RNA are still lacking. Available hybrid structures of the NTD with ssRNA and dsRNA provide valuable insights; however, the precise mechanism of complex formation remains elusive. Similarly, the molecular impact of nucleocapsid NTD mutations that have emerged since 2019 has not yet been fully explored. Using crystallography and solution NMR, we investigate how NTD mutations influence structural integrity and RNA-binding. We find that both features rely on a core network of residues conserved in Betacoronaviruses, crucial for protein stability and communication among flexible loop-regions that facilitate RNA-recognition. Our comprehensive structural analysis demonstrates that contacts within this network guide selective RNA-interactions. We propose that the core network renders the NTD evolutionarily robust in stability and plasticity for its versatile RNA processing roles.
Collapse
Affiliation(s)
- Karthikeyan Dhamotharan
- Institute for Molecular Biosciences, Goethe University, Frankfurt, Germany
- Center for Biomolecular Magnetic Resonance (BMRZ), Goethe University, Frankfurt, Germany
| | - Sophie M Korn
- Institute for Molecular Biosciences, Goethe University, Frankfurt, Germany.
- Center for Biomolecular Magnetic Resonance (BMRZ), Goethe University, Frankfurt, Germany.
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA.
| | - Anna Wacker
- Center for Biomolecular Magnetic Resonance (BMRZ), Goethe University, Frankfurt, Germany
- Institute for Organic Chemistry and Chemical Biology, Goethe University, Frankfurt, Germany
| | - Matthias A Becker
- Center for Biomolecular Magnetic Resonance (BMRZ), Goethe University, Frankfurt, Germany
- Institute for Organic Chemistry and Chemical Biology, Goethe University, Frankfurt, Germany
| | - Sebastian Günther
- Center for Free-Electron Laser Science CFEL, Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, Hamburg, Germany
| | - Harald Schwalbe
- Center for Biomolecular Magnetic Resonance (BMRZ), Goethe University, Frankfurt, Germany
- Institute for Organic Chemistry and Chemical Biology, Goethe University, Frankfurt, Germany
| | - Andreas Schlundt
- Institute for Molecular Biosciences, Goethe University, Frankfurt, Germany.
- Center for Biomolecular Magnetic Resonance (BMRZ), Goethe University, Frankfurt, Germany.
- Institute of Biochemistry, University of Greifswald, Greifswald, Germany.
| |
Collapse
|
23
|
Jones CH, Beitelshees M, Williams BA, Hill AB, Welch VL, True JM. In silico prediction of pathogen's pandemic potential using the viral trait assessment for pandemics (ViTAP) model. PNAS NEXUS 2024; 3:pgae558. [PMID: 39703231 PMCID: PMC11658415 DOI: 10.1093/pnasnexus/pgae558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024]
Abstract
Our world is ever evolving and interconnected, creating constant opportunities for disease outbreaks and pandemics to occur, making pandemic preparedness and pathogen management crucial for global health security. Early pathogen identification and intervention play a key role in mitigating the impacts of disease outbreaks. In this perspective, we present the Viral Trait Assessment for Pandemics (ViTAP) model to aid in the early identification of high-risk viruses that have pandemic potential, which incorporates lessons from past pandemics, including which key viral characteristics are important such as genetic makeup, transmission modes, mutation rates, and symptom severity. This model serves as the foundation for the development of powerful, quantitative tools for the early prediction of pandemic pathogens. The use of such a tool, in conjunction with other pandemic preparedness measures, can allow for early intervention and containment of the virus. This proactive approach could enable timely interventions, guiding public health responses, and resource allocation to prevent widespread outbreaks and mitigate the impact of emerging pathogens.
Collapse
Affiliation(s)
| | | | | | | | - Verna L Welch
- Pfizer, 66 Hudson Boulevard, New York, NY 10018, USA
| | - Jane M True
- Pfizer, 66 Hudson Boulevard, New York, NY 10018, USA
| |
Collapse
|
24
|
Hasan M, He Z, Jia M, Leung ACF, Natarajan K, Xu W, Yap S, Zhou F, Chen S, Su H, Zhu K, Su H. Dynamic expedition of leading mutations in SARS-CoV-2 spike glycoproteins. Comput Struct Biotechnol J 2024; 23:2407-2417. [PMID: 38882678 PMCID: PMC11176665 DOI: 10.1016/j.csbj.2024.05.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 06/18/2024] Open
Abstract
The continuous evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which caused the recent pandemic, has generated countless new variants with varying fitness. Mutations of the spike glycoprotein play a particularly vital role in shaping its evolutionary trajectory, as they have the capability to alter its infectivity and antigenicity. We present a time-resolved statistical method, Dynamic Expedition of Leading Mutations (deLemus), to analyze the evolutionary dynamics of the SARS-CoV-2 spike glycoprotein. The proposed L -index of the deLemus method is effective in quantifying the mutation strength of each amino acid site and outlining evolutionarily significant sites, allowing the comprehensive characterization of the evolutionary mutation pattern of the spike glycoprotein.
Collapse
Affiliation(s)
- Muhammad Hasan
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| | - Zhouyi He
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| | - Mengqi Jia
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Alvin C F Leung
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | | | - Wentao Xu
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Shanqi Yap
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Feng Zhou
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Shihong Chen
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Hailei Su
- Bengbu Hospital of Traditional Chinese Medicine, 4339 Huai-shang Road, Anhui 233080, China
| | - Kaicheng Zhu
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Haibin Su
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| |
Collapse
|
25
|
Reddy KR, Kumari P, Vinay K, Singh J, Chauhan BS, Kumar P, Srikrishna S, Kumar D, Mishra M, Singaravel M, Prasad NK, Anuraag NS, Sahni C, Sharma S. Safety Study and Compositional Analysis of the Svarnvir-IV Tablet With Special Reference to Its Therapeutic Utility in SARS-CoV-2. Cureus 2024; 16:e75438. [PMID: 39791040 PMCID: PMC11713176 DOI: 10.7759/cureus.75438] [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] [Accepted: 12/10/2024] [Indexed: 01/12/2025] Open
Abstract
Aim Traditional Ayurvedic herbo-mineral medicines have proven their potential in managing COVID-19. Cell-based assays of the Svarnvir-IV tablet demonstrated the virucidal activity against SARS-CoV-2 and its therapeutic action, along with safety in cytotoxicity, has been proved. In the present study, in vivo, safety profile and compositional analysis of the Svarnvir-IV tablet were performed. Methods The safety and potency of the Svarnvir tablet were evaluated comprehensively through in vivo drug screening on Drosophila, along with elemental composition analysis of Svarnvir tablets using atomic absorption spectroscopy (AAS), inductively coupled plasma-mass spectroscopy (ICP-MS), X-ray diffraction (XRD), and scanning electron microscopy energy dispersive spectroscopy (SEM-EDS). Results The Svarnvir tablet was found safe in Drosophila and their larvae up to the dosage of 1 mg/ml. In comparison to the control, morphologically and physiologically healthy and active flies were observed without any change in circadian locomotor activity rhythms or activity patterns. In addition, the elemental composition of Svarnvir tablets was evaluated using AAS, ICP-MS, and SEM-EDS, and the microstructure was examined by means of XRD and SEM. Conclusions Overall, these findings will contribute to an accessible and safe therapeutic approach for traditional age-old Ayurvedic medication to combat SARS-CoV-2 variants.
Collapse
Affiliation(s)
- K Ramachandra Reddy
- Department of Rasa Shastra (Ayurvedic Pharmaceutics) Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, IND
| | - Priya Kumari
- Department of Rasa Shastra (Ayurvedic Pharmaceutics) Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, IND
| | - K Vinay
- College of Medicine, Bharti Vidyapeeth Deemed University (BVDU) Medical College, Sangli, IND
| | - Jyotsna Singh
- Department of Biochemistry, Faculty of Science, Banaras Hindu University, Varanasi, IND
| | - Brijesh S Chauhan
- Department of Zoology, Faculty of Science, Banaras Hindu University, Varanasi, IND
| | - Prabhat Kumar
- Department of Biochemistry, Faculty of Science, Banaras Hindu University, Varanasi, IND
| | - Saripella Srikrishna
- Department of Biochemistry, Faculty of Science, Banaras Hindu University, Varanasi, IND
| | - Deepak Kumar
- Department of Zoology, Faculty of Science, Banaras Hindu University, Varanasi, IND
| | - Madhumita Mishra
- Department of Zoology, Faculty of Science, Banaras Hindu University, Varanasi, IND
| | - Muniyandi Singaravel
- Department of Zoology, Faculty of Science, Banaras Hindu University, Varanasi, IND
| | - N K Prasad
- Department of Metallurgical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, IND
| | - N S Anuraag
- Department of Metallurgical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, IND
| | - Chetan Sahni
- Department of Anatomy, All India Institute of Medical Sciences, Gorakhpur, Gorakhpur, IND
| | - Sanchit Sharma
- School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, IND
- Department of Research and Development, Aimil Pharmaceuticals India Limited, New Delhi, IND
| |
Collapse
|
26
|
Li Y, Zhang X, Yi J, Chen Y, Liang J, Wang L, Ma J, Zhu R, Zhang X, Hu D, Jia Y, Yu X, Wang Y. Synergistic evolution: The dynamic adaptation of SARS-CoV-2 and human protective immunity in the real world. J Infect 2024; 89:106310. [PMID: 39393556 DOI: 10.1016/j.jinf.2024.106310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/13/2024]
Abstract
OBJECTIVES SARS-CoV-2 is continually evolving with new variants to evade protective immunity and cause new infections. This study aimed to assess infection-acquired immunity and hybrid immunity against re-infection or severe COVID-19. METHODS During 2020-2023, we collected 890 serum samples from individuals infected with SARS-CoV-2 variants including wild type, D614G, Alpha, Delta, BA.1, BA.2, BA.2.76, BA.5.2, BF.7, XBB, and EG.5. The levels of serum neutralizing antibodies (NAbs) against 18 diverse SARS-CoV-2 variants were determined using a bead-based high-throughput broad neutralizing-antibody assay. RESULTS In the initial wave of the COVID-19 pandemic, >75% of the patients demonstrated robust NAb responses against the ancestral SARS-CoV-2, during a period when vaccines were not yet available. After the emergence of the Omicron variant, the seroprevalence of anti-Omicron NAbs among the patients increased rapidly. By April 2023, when XBB variant was predominant, approximately 80% of the patients demonstrated >50% neutralization against the highly immune-evasive XBB lineages. Three serotypes of SARS-CoV-2, namely non-Omicron, Omicron, and XBB serotypes, were identified, with the strong likelihood of further changes occurring as the virus mutating. Generally, NAbs elicited by a previous serotype could not typically effectively protect against another serotype that emerges later in the evolutionary stages. CONCLUSION Our results firstly demonstrated the synergistic evolution between host immunity and SARS-CoV-2 variants in the real world, which would be helpful to develop future vaccines and public health strategies.
Collapse
Affiliation(s)
- Yunhui Li
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xiaohan Zhang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Jingkun Yi
- Department of Biomedical Informatics, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Yuan Chen
- Department of Clinical Laboratory, Peking University Ditan Teaching Hospital, Beijing 100015, China
| | - Jing Liang
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Li Wang
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Jiayue Ma
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Renlong Zhu
- Department of Clinical Laboratory, Peking University Ditan Teaching Hospital, Beijing 100015, China
| | - Xiaomei Zhang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Di Hu
- ProteomicsEra Medical Co., Ltd., Beijing 102206, China
| | - Yan Jia
- ProteomicsEra Medical Co., Ltd., Beijing 102206, China
| | - Xiaobo Yu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China.
| | - Yajie Wang
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China.
| |
Collapse
|
27
|
Al-Mankhee AA, Moatasim Y, El Taweel A, Gomaa M, Rabiee OA, Gado MM, Barakat AB, Ali MA, El-Shesheny R. Genomic Diversity and Evolution of Identified SARS-CoV-2 Variants in Iraq. Pathogens 2024; 13:1051. [PMID: 39770311 PMCID: PMC11728743 DOI: 10.3390/pathogens13121051] [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: 10/27/2024] [Revised: 11/21/2024] [Accepted: 11/27/2024] [Indexed: 01/16/2025] Open
Abstract
The COVID-19 pandemic caused by the SARS-CoV-2 virus continues to circulate worldwide, causing the deaths of millions of people. The continuous circulation of the virus, its genetic diversity, the emergence of new variants with increased transmissibility, and/or the capacity of the virus to escape from the immune system constitute a major public health concern. In our study, we aimed to characterize SARS-CoV-2 strains in Iraq from the first introduction until the end of 2023, and to identify their variants, lineages, clades, and mutation patterns. All published Iraqi full genome sequences (2020-2023) were obtained from Global Initiative on Sharing All Influenza Data (GISAID) and subjected to molecular characterization along with 19 samples of full genome sequences that were collected during the fifth and sixth waves of the SARS-CoV-2 pandemic in this study. Next-generation sequencing was performed using an Illumina MiSeq system, and phylogenetic analysis was performed for all the Iraqi sequences. Three established global platforms, GISAID, Nextstrain, and PANGO, were used for the classification of isolates into distinct clades, variants, and lineages. Six wave peaks of COVID-19 cases have been identified in Iraq, resulting in approximately 2,400,000 cumulative confirmed cases and more than 25,000 deaths. Our study revealed patterns of circulation and dominance of SARS-CoV-2 clades and their lineages in the pandemic waves in the country.
Collapse
Affiliation(s)
- Ahmed A. Al-Mankhee
- Al Hussein Teaching Hospital, Al Bat’haa 64011, Thi Qar, Iraq;
- Department of Microbiology, Faculty of Science, Ain Shams University, Cairo 11566, Egypt; (O.A.R.); (M.M.G.); (A.B.B.)
| | - Yassmin Moatasim
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt; (Y.M.); (A.E.T.); (M.G.)
| | - Ahmed El Taweel
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt; (Y.M.); (A.E.T.); (M.G.)
| | - Mokhtar Gomaa
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt; (Y.M.); (A.E.T.); (M.G.)
| | - Omar A. Rabiee
- Department of Microbiology, Faculty of Science, Ain Shams University, Cairo 11566, Egypt; (O.A.R.); (M.M.G.); (A.B.B.)
| | - Marwa M. Gado
- Department of Microbiology, Faculty of Science, Ain Shams University, Cairo 11566, Egypt; (O.A.R.); (M.M.G.); (A.B.B.)
| | - Ahmed B. Barakat
- Department of Microbiology, Faculty of Science, Ain Shams University, Cairo 11566, Egypt; (O.A.R.); (M.M.G.); (A.B.B.)
| | - Mohamed A. Ali
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt; (Y.M.); (A.E.T.); (M.G.)
| | - Rabeh El-Shesheny
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt; (Y.M.); (A.E.T.); (M.G.)
| |
Collapse
|
28
|
Lyu C, He Z, Hu X, Wang S, Qin M, Zhu L, Li Y, Yang F, Jiao Z, Zhang X, Lu G, Wang E, Hu Y, Zhai Y, Wang Y, Huang W, Wang D, Cui Y, Pang X, Liu X, Kamiya H, Ma G, Wei W. Lysosomal "TRAP": a neotype modality for clearance of viruses and variants. Nat Commun 2024; 15:10155. [PMID: 39578473 PMCID: PMC11584657 DOI: 10.1038/s41467-024-54505-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024] Open
Abstract
The binding of viruses to host-entry factor receptors is an essential step for viral infection. Many studies have shown that macrophages can internalize viruses and degrade them in lysosomes for clearance in vivo. Inspired by these natural behaviors and using SARS-CoV-2 as a testbed, we harvest lysosomes from activated macrophages and anchor the protein-receptor ACE2 as bait, thus constructing a lysosomal "TRAP" (lysoTRAP) that selectively captures, internalizes, and eventually degrades SARS-CoV-2. Through experiments with cells, female mice, female hamsters, and human lung organoids, we demonstrate that lysoTRAP effectively clears SARS-CoV-2. Importantly, unlike therapeutic agents targeting SARS-CoV-2 spike protein, lysoTRAP remains effective against nine pseudotyped variants and the authentic Omicron variant, demonstrating its resistance to SARS-CoV-2 mutations. In addition to the protein-receptor ACE2, we also extend lysoTRAP with the saccharide-receptor sialic acid and verify its excellent antiviral effect against H1N1, highlighting the flexibility of our "TRAP" platform in fighting against various viruses.
Collapse
Affiliation(s)
- Chengliang Lyu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, Tokyo, 184-8588, Japan
| | - Zhanlong He
- Institute of Medical Biology, Peking Union Medical College & Chinese Academy of Medical Sciences, Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Disease, Kunming, 650118, China
| | - Xiaoming Hu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shuang Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Meng Qin
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Li Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Yanyan Li
- Institute of Medical Biology, Peking Union Medical College & Chinese Academy of Medical Sciences, Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Disease, Kunming, 650118, China
| | - Fengmei Yang
- Institute of Medical Biology, Peking Union Medical College & Chinese Academy of Medical Sciences, Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Disease, Kunming, 650118, China
| | - Zhouguang Jiao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xiao Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guihong Lu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Erqiang Wang
- Sinovac Life Sciences Co., Ltd., Beijing, 100085, China
| | - Yaling Hu
- Sinovac Life Sciences Co., Ltd., Beijing, 100085, China
| | - Yu Zhai
- Sinovac Life Sciences Co., Ltd., Beijing, 100085, China
| | - Youchun Wang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC) and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, 102629, China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC) and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, 102629, China
| | - Dongshu Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Yimin Cui
- Department of Pharmacy, Peking University First Hospital, Beijing, 100034, China
- Institute of Clinical Pharmacology, Peking University, Beijing, 100191, China
| | - Xiaocong Pang
- Department of Pharmacy, Peking University First Hospital, Beijing, 100034, China
- Institute of Clinical Pharmacology, Peking University, Beijing, 100191, China
| | - Xiangzheng Liu
- Department of thoracic surgery, Peking University First Hospital, Beijing, 100034, China
| | - Hidehiro Kamiya
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, Tokyo, 184-8588, Japan
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China.
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China.
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China.
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China.
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
29
|
Barazorda-Ccahuana HL, Cárcamo Rodriguez EG, Centeno-Lopez A, Paco-Chipana M, Goyzueta-Mamani LD, Chavez-Fumagalli MA. Identification of compounds from natural Peruvian sources as potential inhibitors of SARS-CoV-2 Mpro mutations by virtual screening and computational simulations. F1000Res 2024; 13:246. [PMID: 39583212 PMCID: PMC11585855 DOI: 10.12688/f1000research.143633.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/19/2024] [Indexed: 11/26/2024] Open
Abstract
Background Although the COVID-19 pandemic has diminished in intensity, the virus continues to circulate globally. The SARS-CoV-2 main protease (Mpro) is a key enzyme in the life cycle of the virus, making it important for the development of treatments against future variants of the virus. In this work, Peruvian natural compounds were evaluated against different mutations of the SARS-CoV-2 Mpro. Methods In silico techniques such as virtual screening, all-atom molecular dynamics simulations, and energy estimation analysis were applied. Results Of the tested compounds by virtual screening, rutin was identified as the best binding agent against the different proposed Mpro mutations. In addition, computational simulations and energy estimation analysis demonstrated the high structural and energetic stability between the Mpro-rutin systems. Conclusions Overall, our study identified rutin as the most promising compound with a strong affinity for various Mpro mutations, potentially playing a key role in the development of new treatments for emerging viral variants.
Collapse
Affiliation(s)
- Haruna Luz Barazorda-Ccahuana
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Catolica de Santa Maria de Arequipa, Pedro Vilcapaza, Arequipa, 04000, Peru
| | - Eymi Gladys Cárcamo Rodriguez
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Catolica de Santa Maria de Arequipa, Pedro Vilcapaza, Arequipa, 04000, Peru
- Facultad de Ciencias Farmaceuticas, Bioquímicas y Biotecnológicas, Universidad Catolica de Santa Maria de Arequipa, Pedro Vilcapaza, Arequipa, 04000, Peru
| | - Angela Emperatriz Centeno-Lopez
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Catolica de Santa Maria de Arequipa, Pedro Vilcapaza, Arequipa, 04000, Peru
- Facultad de Ciencias Farmaceuticas, Bioquímicas y Biotecnológicas, Universidad Catolica de Santa Maria de Arequipa, Pedro Vilcapaza, Arequipa, 04000, Peru
| | - Margot Paco-Chipana
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Catolica de Santa Maria de Arequipa, Pedro Vilcapaza, Arequipa, 04000, Peru
| | - Luis Daniel Goyzueta-Mamani
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Catolica de Santa Maria de Arequipa, Pedro Vilcapaza, Arequipa, 04000, Peru
- Sustainable Innovative Biomaterials, Le Qara Research Center, Arequipa, Peru
| | - Miguel Angel Chavez-Fumagalli
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Catolica de Santa Maria de Arequipa, Pedro Vilcapaza, Arequipa, 04000, Peru
| |
Collapse
|
30
|
Akçeşme FB, Köprülü TK, Çam BE, İş Ş, Keskin BC, Akçeşme B, Baydili KN, Gezer B, Balkan J, Uçar B, Gürsoy O, Yıldız MT, Kurt H, Ünal N, Korkmaz C, Saral ÖB, Demirkol B, Çağ Y, Abakay H, Köse Ş, Türkez H, Çadırcı K, Altındiş M, Gülseren YD, Aslan N, Özel A, Karagöl MA, Mutluay N, Tekin Ş. Genomic Surveillance and Molecular Characterization of SARS-CoV-2 Variants During the Peak of the Pandemic in Türkiye. Biochem Genet 2024:10.1007/s10528-024-10962-8. [PMID: 39516327 DOI: 10.1007/s10528-024-10962-8] [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: 04/30/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
SARS-CoV-2 is a highly transmissible coronavirus and has caused a pandemic of acute respiratory disease. Genomic characterization of SARS-CoV-2 is important for monitoring and assessing its evolution. A total of 1.346 nasopharyngeal swab samples were collected but only 879 SARS-CoV-2 high-quality genomes were isolated, subjected to Next Generation Sequencing and analyzed both statistically and regarding mutations comprehensively. The distribution of clades and lineages in different cities of Türkiye and the association of SARS-CoV-2 variants with age groups and clinical characteristics of COVID-19 were also examined. Furthermore, the frequency of the clades and lineages was observed in 10 months. Finally, non-synonymous mutations not defined in specific SARS-CoV-2 variants (during that period) were identified by performing mutation analysis. B.1.1.7 (Alpha) and B.1.617.2 (Delta) SARS-CoV-2 variants which have also been identified in our study from March to December 2021. We observed a significant association of SARS-CoV-2 variants with age groups and cities. Also, E:T9I, S:A27S, S:A67V, S:D796Y, S:K417N, S:N440K, S:R158X, S:S477N (below 1%-frequency) were determined as specific mutations belonging and shared with the Omicron variant that appeared later. Our study has highlighted the importance of constant monitoring of the genetic diversity of SARS-CoV-2 to provide better prevention strategies and it contributes to the understanding of SARS-CoV-2 from the past to the present.
Collapse
Affiliation(s)
- Faruk Berat Akçeşme
- Division of Biostatistics and Medical Informatics, Department of Basic Medical Sciences, Hamidiye Faculty of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Tuğba Kul Köprülü
- Division of Medical Laboratory Techniques, Department of Medical Services and Techniques, Hamidiye Health Services Vocational School, University of Health Sciences, Istanbul, Turkey
- Experimental Medicine Application and Research Center, University of Health Sciences, Istanbul, Turkey
| | - Burçin Erkal Çam
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, Yıldız Technical University, Istanbul, Turkey
| | - Şeyma İş
- Division of Bioinformatics, Department of Molecular Biotechnology, Faculty of Science, Turkish-German University, Istanbul, Turkey
- Division of Medical Biology, Department of Basic Medical Sciences, Hamidiye Faculty of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Birsen Cevher Keskin
- Genome Research Center, Life Sciences, Marmara Research Center, TUBITAK, Kocaeli, Turkey
| | - Betül Akçeşme
- Division of Medical Biology, Department of Basic Medical Sciences, Hamidiye Faculty of Medicine, University of Health Sciences, Istanbul, Turkey.
| | - Kürşad Nuri Baydili
- Department of Biostatistics and Medical Informatics, Hamidiye Faculty of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Bahar Gezer
- Experimental Medicine Application and Research Center, University of Health Sciences, Istanbul, Turkey
- Department of Molecular Medicine, Hamidiye Faculty of Health Sciences, University of Health Sciences, Istanbul, Turkey
| | - Jülide Balkan
- Experimental Medicine Application and Research Center, University of Health Sciences, Istanbul, Turkey
- Department of Molecular Medicine, Hamidiye Faculty of Health Sciences, University of Health Sciences, Istanbul, Turkey
| | - Bihter Uçar
- Genome Research Center, Life Sciences, Marmara Research Center, TUBITAK, Kocaeli, Turkey
- Department of Biology, Faculty of Science, Marmara University, Istanbul, Turkey
| | - Osman Gürsoy
- Department of Computer Sciences and Engineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Mehmet Taha Yıldız
- Hamidiye Institute of Science, Molecular Medicine, Hamidiye Health Services Vocational School, University of Health Sciences, Istanbul, Turkey
| | - Halil Kurt
- Department of Medical Biology, Hamidiye International Faculty of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Nevzat Ünal
- Department of Microbiology, Adana City Training and Research Hospital, Adana, Turkey
| | - Celalettin Korkmaz
- Division of Thoracic Diseases, Department of Internal Medicine, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Özlem Bayraktar Saral
- Clinic for Infectious Diseases and Clinical Microbiology, Trabzon Kanuni Training and Research Hospital, Trabzon, Turkey
| | - Barış Demirkol
- Department of Chest Diseases, Basaksehir Cam and Sakura City Hospital, University of Health Sciences, Istanbul, Turkey
| | - Yasemin Çağ
- Division of Infectious Diseases and Clinical Microbiology, Department of Internal Medicine, Faculty of Medicine, İstanbul Medeniyet University, Istanbul, Turkey
| | - Hilal Abakay
- Clinic for Infectious Diseases, İzmir Tepecik Training and Research Hospital, İzmir, Turkey
| | - Şükran Köse
- Division of Infectious Diseases, Department of Internal Medicine, Dokuz Eylül Univesity, İzmir, Turkey
| | - Hasan Türkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Kenan Çadırcı
- Department of Internal Medicine, Erzurum Regional Education and Research Hospital, Erzurum, Turkey
| | - Mustafa Altındiş
- Division of Medical Microbiology, Department of Basic Medical Sciences, Faculty of Medicine, Sakarya University, Sakarya, Turkey
| | | | - Nuray Aslan
- Emergency Service, Sakarya University Training and Research Hospital, Sakarya, Turkey
| | - Abdulkadir Özel
- Experimental Medicine Application and Research Center, University of Health Sciences, Istanbul, Turkey
- Dr. Orhan Öcalgiray Molecular Biology-Biotechnology and Genetics Research Center (ITU-MOBGAM), Faculty of Science and Letters, İstanbul Technical University, Istanbul, Turkey
| | - Muhammet Atıf Karagöl
- Department of Chest Diseases, Basaksehir Cam and Sakura City Hospital, University of Health Sciences, Istanbul, Turkey
| | - Neslihan Mutluay
- Department of Medical Microbiology, Adana City Training and Research Hospital, Adana, Turkey
- Division of Medical Microbiology, Department of Basic Medical Sciences, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - Şaban Tekin
- Division of Medical Biology, Department of Basic Medical Sciences, Hamidiye Faculty of Medicine, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
31
|
Erkihun M, Ayele B, Asmare Z, Endalamaw K. Current Updates on Variants of SARS-CoV- 2: Systematic Review. Health Sci Rep 2024; 7:e70166. [PMID: 39502131 PMCID: PMC11534727 DOI: 10.1002/hsr2.70166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 06/11/2024] [Accepted: 07/29/2024] [Indexed: 11/08/2024] Open
Abstract
Background Coronavirus disease 2019 is caused by the severe acute respiratory syndrome coronavirus 2, which has become a pandemic. Severe acute respiratory syndrome coronavirus 2 is an enveloped, unsegmented, positive-sense, single-stranded RNA virus that belongs to the family Coronaviridae. Aim The objective of this review is to conduct a qualitative analysis of the current updates on epidemiology, evolution, and vaccine variants for SARS-CoV-2. Method The search strategy was done from the database based on the PRISMA criteria for qualitative analysis of this review. Literature on variants of severe acute respiratory syndrome coronavirus 2, published in English in the last 5 years (2019-2023), were included. From 179 a total of 105 articles were reviewed, searched, and retrieved from the electronic databases PubMed. The search was done using keywords like COVID-19, SARS-CoV-2, variants, mutations, and vaccines, and articles were managed using EndNote X8 software. The scope of view for this review was the course of the pandemic by emerging variants and how man is struggling to overcome this sudden pandemic through vaccines. The narrative skeleton was constructed based on the article's scope of view. Result From the parent severe acute respiratory syndrome coronavirus 2, many variants emerged during the course of this pandemic. They are mainly categorized into two variants: variants of interest and variants of concern based on the impact on public health. The World Health Organization leveled five variants: Alpha (strain B.1.1.7), Beta (strain B.1.351), Gamma (strain P.1), Delta (strain B.1.617.2), and Omicron (B.1.1.529). Conclusions It is crucial to stay informed about the latest developments in the understanding of SARS-CoV-2 variants, as new variants can emerge and impact the course of the pandemic. Health authorities and researchers continuously have to monitor and study these variants to assess their characteristics, transmissibility, severity, and the effectiveness of vaccines against them. One has to always refer to the latest information from reputable health journals or organizations for the most up-to-date and accurate details on COVID-19 variants.
Collapse
Affiliation(s)
- Mulat Erkihun
- Department of Medical Laboratory Sciences, School of Health Sciences, College of Medicine and Health SciencesDebre Tabor UniversityDebre TaborEthiopia
| | - Bayu Ayele
- Laboratory Service UnitFelege Hiwot Comprehensive Specialized HospitalBahir DarEthiopia
| | - Zelalem Asmare
- Department of Medical Laboratory Sciences, College of Health SciencesWoldia UniversityWoldiaEthiopia
| | - Kirubel Endalamaw
- Department of Diagnostic Laboratory at Shegaw Motta General HospitalMotta TownEthiopia
| |
Collapse
|
32
|
Chen P, Bergman P, Blennow O, Hansson L, Mielke S, Nowak P, Gao Y, Söderdahl G, Österborg A, Smith CIE, Vesterbacka J, Wullimann D, Cuapio A, Akber M, Bogdanovic G, Muschiol S, Åberg M, Loré K, Chen MS, Ljungman P, Buggert M, Aleman S, Ljunggren HG. Real-world assessment of immunogenicity in immunocompromised individuals following SARS-CoV-2 mRNA vaccination: a two-year follow-up of the prospective clinical trial COVAXID. EBioMedicine 2024; 109:105385. [PMID: 39395230 DOI: 10.1016/j.ebiom.2024.105385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND Immunocompromised patients with primary and secondary immunodeficiencies have shown impaired responses to SARS-CoV-2 mRNA vaccines, necessitating recommendations for additional booster doses. However, longitudinal data reflecting the real-world impact of such recommendations remains limited. METHODS This study represents a two-year follow-up of the COVAXID clinical trial, where 364 of the original 539 subjects consented to participate. 355 individuals provided blood samples for evaluation of binding antibody (Ab) titers and pseudo-neutralisation capacity against both the ancestral SARS-CoV-2 strain and prevalent Omicron variants. T cell responses were assessed in a subset of these individuals. A multivariate analysis determined the correlation between Ab responses and the number of vaccine doses received, documented infection events, immunoglobulin replacement therapy (IGRT), and specific immunosuppressive drugs. The original COVAXID clinical trial was registered in EudraCT (2021-000175-37) and clinicaltrials.gov (NCT04780659). FINDINGS Several of the patient groups that responded poorly to the initial primary vaccine schedule and early booster doses presented with stronger immunogenicity-related responses including binding Ab titres and pseudo-neutralisation at the 18- and 24-month sampling time point. Responses correlated positively with the number of vaccine doses and infection. The vaccine response was blunted by an immunosuppressive state due to the underlying specific disease and/or to specific immunosuppressive treatment. INTERPRETATION The study results highlight the importance of continuous SARS-CoV-2 vaccine booster doses in building up and sustaining Ab responses in specific immunocompromised patient populations. FUNDING The present studies were supported by the European Research Council, Karolinska Institutet, Knut and Alice Wallenberg Foundation, Nordstjernan AB, Region Stockholm, and the Swedish Research Council.
Collapse
Affiliation(s)
- Puran Chen
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Peter Bergman
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Laboratory Medicine, Clinical Immunology, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Ola Blennow
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Transplantation, Karolinska University Hospital, Stockholm, Sweden; Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Lotta Hansson
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Stephan Mielke
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Karolinska Comprehensive Cancer Center, Stockholm, Sweden; Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Piotr Nowak
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - Yu Gao
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gunnar Söderdahl
- Department of Transplantation, Karolinska University Hospital, Stockholm, Sweden; Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Anders Österborg
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - C I Edvard Smith
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jan Vesterbacka
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - David Wullimann
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Angelica Cuapio
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mira Akber
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gordana Bogdanovic
- Dept of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden; Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sandra Muschiol
- Dept of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden; Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Mikael Åberg
- Department of Medical Sciences, Clinical Chemistry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Karin Loré
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Margaret Sällberg Chen
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Per Ljungman
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Karolinska Comprehensive Cancer Center, Stockholm, Sweden; Department of Medicine Huddinge, Division of Hematology, Karolinska Institutet, Stockholm, Sweden
| | - Marcus Buggert
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Soo Aleman
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden.
| | - Hans-Gustaf Ljunggren
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
33
|
Wee J, Chen J, Wei GW. Preventing future zoonosis: SARS-CoV-2 mutations enhance human-animal cross-transmission. Comput Biol Med 2024; 182:109101. [PMID: 39243518 PMCID: PMC11560627 DOI: 10.1016/j.compbiomed.2024.109101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/13/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024]
Abstract
The COVID-19 pandemic has driven substantial evolution of the SARS-CoV-2 virus, yielding subvariants that exhibit enhanced infectiousness in humans. However, this adaptive advantage may not universally extend to zoonotic transmission. In this work, we hypothesize that viral adaptations favoring animal hosts do not necessarily correlate with increased human infectivity. In addition, we consider the potential for gain-of-function mutations that could facilitate the virus's rapid evolution in humans following adaptation in animal hosts. Specifically, we identify the SARS-CoV-2 receptor-binding domain (RBD) mutations that enhance human-animal cross-transmission. To this end, we construct a multitask deep learning model, MT-TopLap trained on multiple deep mutational scanning datasets, to accurately predict the binding free energy changes upon mutation for the RBD to ACE2 of various species, including humans, cats, bats, deer, and hamsters. By analyzing these changes, we identified key RBD mutations such as Q498H in SARS-CoV-2 and R493K in the BA.2 variant that are likely to increase the potential for human-animal cross-transmission.
Collapse
Affiliation(s)
- JunJie Wee
- Department of Mathematics, Michigan State University, East Lansing, MI 48824, USA
| | - Jiahui Chen
- Department of Mathematical Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| | - Guo-Wei Wei
- Department of Mathematics, Michigan State University, East Lansing, MI 48824, USA; Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA; Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
34
|
Yang YF, Lin YJ, You SH, Lu TH, Chen CY, Wang WM, Ling MP, Chen SC, Liao CM. A Regional-Scale Assessment-Based SARS-CoV-2 Variants Control Modeling with Implications for Infection Risk Characterization. Infect Drug Resist 2024; 17:4791-4805. [PMID: 39498414 PMCID: PMC11533883 DOI: 10.2147/idr.s480086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/25/2024] [Indexed: 11/07/2024] Open
Abstract
Background The emergence and progression of highly divergent SARS-CoV-2 variants have posed increased risks to global public health, triggering the significant impacts on countermeasures since 2020. However, in addition to vaccination, the effectiveness of non-pharmaceutical interventions, such as social distancing, masking, or hand washing, on different variants of concern (VOC) remains largely unknown. Objective This study provides a mechanistic approach by implementing a control measure model and a risk assessment framework to quantify the impacts of control measure combinations on the transmissions of five VOC (Alpha, Beta, Delta, Gamma, and Omicron), along with a different perspective of risk assessment application. Materials and Methods We applied uncontrollable ratios as an indicator by adopting basic reproduction number (R 0) data collected from a regional-scale survey. A risk assessment strategy was established by constructing VOC-specific dose-response profiles to implicate practical uses in risk characterization when exposure data are available. Results We found that social distancing alone was ineffective without vaccination in almost all countries and VOC when the median R 0 was greater than two. Our results indicated that Omicron could not be contained, even when all control measure combinations were applied, due to its low threshold of infectivity (~3×10-4 plague-forming unit (PFU) mL-1). Conclusion To facilitate better decision-making in future interventions, we provide a comprehensive evaluation of how combined control measures impact on different countries and various VOC. Our findings indicate the potential application of threshold estimates of infectivity in the context of risk communication and policymaking for controlling future emerging SARS-CoV-2 variant infections.
Collapse
Affiliation(s)
- Ying-Fei Yang
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei, 10617, Taiwan
| | - Yi-Jun Lin
- Institute of Food Safety and Health Risk Assessment, National Yang Ming Chiao Tung University, Taipei, 11230, Taiwan
| | - Shu-Han You
- Institute of Food Safety and Risk Management, National Taiwan Ocean University, Keelung City, 20224, Taiwan
| | - Tien-Hsuan Lu
- Department of Science Education and Application, National Taichung University of Education, Taichung, 403514, Taiwan
| | - Chi-Yun Chen
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL, 32610, USA
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, 32608, USA
| | - Wei-Min Wang
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei, 10617, Taiwan
| | - Min-Pei Ling
- Department of Food Science, National Taiwan Ocean University, Keelung City, 20224, Taiwan
| | - Szu-Chieh Chen
- Department of Public Health, Chung Shan Medical University, Taichung, 40201, Taiwan
- Department of Family and Community Medicine, Chung Shan Medical University Hospital, Taichung, 40201, Taiwan
| | - Chung-Min Liao
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei, 10617, Taiwan
| |
Collapse
|
35
|
Yoo JW, Kim WY, Chung CR, Cho YJ, Lee J, Jegal Y, Kim J, Joh JS, Park TY, Baek AR, Park JH, Chae G, Hwang JH, Song JW. Early pulmonary fibrosis-like changes between delta and pre-delta periods in patients with severe COVID-19 pneumonia on mechanical ventilation. Sci Rep 2024; 14:26101. [PMID: 39478105 PMCID: PMC11525473 DOI: 10.1038/s41598-024-77405-7] [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: 02/13/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
It remains unclear whether pulmonary fibrosis-like changes differ in patients with different SARS-CoV-2 variants. This study aimed to compare pulmonary fibrotic changes between two SARS-CoV-2 variant periods (delta vs. pre-delta) in critically ill patients with SARS-CoV-2 pneumonia. Clinical data and chest CT images of patients with SARS-CoV-2 pneumonia receiving mechanical ventilation were collected from 10 hospitals in South Korea over two periods: delta (July-December, 2021; n = 64) and pre-delta (February, 2020-June, 2021; n = 120). Fibrotic changes on chest CT were evaluated through visual assessment. Of 184 patients, the mean age was 64.6 years, and 60.5% were ale. Fibrosis-like changes on chest CT (median 51 days from enrollment to follow up CT scan, interquartile range 27-76 days) were identified in 75.3%. Delta group showed more fibrosis-like changes (≥ 2) (69.8% vs. 43.1%, P = 0.001) and more frequent reticulation and architectural distortion+/-parenchymal band than pre-delta group. Even after propensity score matching with clinical variables, delta group had more severe (≥ 2) fibrosis-like changes (71.4% vs. 38.8%, P = 0.001), and more frequent reticulation and architectural distortion+/-parenchymal band than pre-delta group. Our data suggest that critically ill patients with SARS-CoV-2 in delta period had more severe pulmonary fibrosis-like changes than those in pre-delta period.
Collapse
Affiliation(s)
- Jung-Wan Yoo
- Department of Internal Medicine, Gyeongsang National University Hospital, Jinju, Republic of Korea
| | - Won-Young Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chung Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Chi Ryang Chung
- Department of Critical Care Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Young-Jae Cho
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jinwoo Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yangjin Jegal
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Junghyun Kim
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Hallym University Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong, Republic of Korea
| | - Joon-Sung Joh
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, National Medical Center, Seoul, Republic of Korea
| | - Tae Yun Park
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul Metropolitan Government, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Ae-Rin Baek
- Division of Allergy and Pulmonology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Joo Hun Park
- Department of Pulmonary and Critical Care Medicine, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Ganghee Chae
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Jung Hwa Hwang
- Department of Radiology, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Jin Woo Song
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu 05505, Seoul, Republic of Korea.
| |
Collapse
|
36
|
Chen N, Decker KE, Schulz SR, Kempf A, Nehlmeier I, Moldenhauer AS, Dopfer-Jablonka A, Behrens GMN, Stankov MV, Manthey L, Jäck HM, Hoffmann M, Pöhlmann S, Arora P. Comparative Analysis of Host Cell Entry Efficiency and Neutralization Sensitivity of Emerging SARS-CoV-2 Lineages KP.2, KP.2.3, KP.3, and LB.1. Vaccines (Basel) 2024; 12:1236. [PMID: 39591139 PMCID: PMC11598761 DOI: 10.3390/vaccines12111236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/28/2024] Open
Abstract
New SARS-CoV-2 lineages continue to evolve and may exhibit new characteristics regarding host cell entry efficiency and potential for antibody evasion. Here, employing pseudotyped particles, we compared the host cell entry efficiency, ACE2 receptor usage, and sensitivity to antibody-mediated neutralization of four emerging SARS-CoV-2 lineages, KP.2, KP.2.3, KP.3, and LB.1. The XBB.1.5 and JN.1 lineages served as controls. Our findings reveal that KP.2, KP.2.3, KP.3, and LB.1 lineages enter host cells efficiently and in an ACE2-dependent manner, and that KP.3 is more adept at entering Calu-3 lung cells than JN.1. However, the variants differed in their capacity to employ ACE2 orthologues from animal species for entry, suggesting differences in ACE2 interactions. Moreover, we demonstrate that only two out of seven therapeutic monoclonal antibody (mAbs) in preclinical development retain robust neutralizing activity against the emerging JN.1 sublineages tested, while three mAbs displayed strongly reduced neutralizing activity and two mAbs lacked neutralizing activity against any of the lineages tested. Furthermore, our results show that KP.2, KP.2.3, KP.3, and LB.1 lineages evade neutralization by antibodies induced by infection or vaccination with greater efficiency than JN.1, particularly in individuals without hybrid immunity. This study indicates that KP.2, KP.2.3, KP.3, and LB.1 differ in ACE2 interactions and the efficiency of lung cell entry and suggest that evasion of neutralizing antibodies drove the emergence of these variants.
Collapse
Affiliation(s)
- Nianzhen Chen
- Infection Biology Unit, German Primate Center—Leibniz Institute for Primate Research, 37077 Göttingen, Germany; (N.C.); (K.E.D.); (A.K.); (I.N.); (A.-S.M.); (M.H.)
- Faculty of Biology and Psychology, Georg-August-University Göttingen, 37073 Göttingen, Germany
| | - Katharina Emma Decker
- Infection Biology Unit, German Primate Center—Leibniz Institute for Primate Research, 37077 Göttingen, Germany; (N.C.); (K.E.D.); (A.K.); (I.N.); (A.-S.M.); (M.H.)
- Faculty of Biology and Psychology, Georg-August-University Göttingen, 37073 Göttingen, Germany
| | - Sebastian R. Schulz
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander University of Erlangen-Nürnberg, 91054 Erlangen, Germany; (S.R.S.); (H.-M.J.)
| | - Amy Kempf
- Infection Biology Unit, German Primate Center—Leibniz Institute for Primate Research, 37077 Göttingen, Germany; (N.C.); (K.E.D.); (A.K.); (I.N.); (A.-S.M.); (M.H.)
- Faculty of Biology and Psychology, Georg-August-University Göttingen, 37073 Göttingen, Germany
| | - Inga Nehlmeier
- Infection Biology Unit, German Primate Center—Leibniz Institute for Primate Research, 37077 Göttingen, Germany; (N.C.); (K.E.D.); (A.K.); (I.N.); (A.-S.M.); (M.H.)
| | - Anna-Sophie Moldenhauer
- Infection Biology Unit, German Primate Center—Leibniz Institute for Primate Research, 37077 Göttingen, Germany; (N.C.); (K.E.D.); (A.K.); (I.N.); (A.-S.M.); (M.H.)
| | - Alexandra Dopfer-Jablonka
- Department of Rheumatology and Immunology, Hannover Medical School, 30625 Hannover, Germany; (A.D.-J.); (G.M.N.B.); (M.V.S.); (L.M.)
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 30625 Hannover, Germany
| | - Georg M. N. Behrens
- Department of Rheumatology and Immunology, Hannover Medical School, 30625 Hannover, Germany; (A.D.-J.); (G.M.N.B.); (M.V.S.); (L.M.)
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 30625 Hannover, Germany
- Center for Individualized Infection Medicine (CiiM), 30625 Hannover, Germany
| | - Metodi V. Stankov
- Department of Rheumatology and Immunology, Hannover Medical School, 30625 Hannover, Germany; (A.D.-J.); (G.M.N.B.); (M.V.S.); (L.M.)
| | - Luis Manthey
- Department of Rheumatology and Immunology, Hannover Medical School, 30625 Hannover, Germany; (A.D.-J.); (G.M.N.B.); (M.V.S.); (L.M.)
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander University of Erlangen-Nürnberg, 91054 Erlangen, Germany; (S.R.S.); (H.-M.J.)
| | - Markus Hoffmann
- Infection Biology Unit, German Primate Center—Leibniz Institute for Primate Research, 37077 Göttingen, Germany; (N.C.); (K.E.D.); (A.K.); (I.N.); (A.-S.M.); (M.H.)
- Faculty of Biology and Psychology, Georg-August-University Göttingen, 37073 Göttingen, Germany
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center—Leibniz Institute for Primate Research, 37077 Göttingen, Germany; (N.C.); (K.E.D.); (A.K.); (I.N.); (A.-S.M.); (M.H.)
- Faculty of Biology and Psychology, Georg-August-University Göttingen, 37073 Göttingen, Germany
| | - Prerna Arora
- Infection Biology Unit, German Primate Center—Leibniz Institute for Primate Research, 37077 Göttingen, Germany; (N.C.); (K.E.D.); (A.K.); (I.N.); (A.-S.M.); (M.H.)
- Faculty of Biology and Psychology, Georg-August-University Göttingen, 37073 Göttingen, Germany
| |
Collapse
|
37
|
Sharma S, Roy D, Cherian S. In-silico evaluation of the T-cell based immune response against SARS-CoV-2 omicron variants. Sci Rep 2024; 14:25413. [PMID: 39455652 PMCID: PMC11511884 DOI: 10.1038/s41598-024-75658-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
During of COVID-19 pandemic, Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has continuously evolved, resulting in the emergence of several new variants of concerns (VOCs) with numerous mutations. These VOCs dominate in various regions due to increased transmissibility and antibody evasion, potentially reducing vaccine effectiveness. Nonetheless, it remains uncertain whether the recent SARS-CoV-2 VOCs have the ability to circumvent the T cell immunity elicited by either COVID-19 vaccination or natural infection. To address this, we conducted in-silico analysis to examine the impact of VOC-specific mutations at the epitope level and T cell cross-reactivity with the ancestral SARS-CoV-2. According to the in-silico investigation, T cell responses triggered by immunization or prior infections still recognize the variants in spite of mutations. These variants are expected to either maintain their dominant epitope HLA patterns or bind with new HLAs, unlike the epitopes of the ancestral strain. Our findings indicate that a significant proportion of immuno-dominant CD8 + and CD4 + epitopes are conserved across all the variants, implying that existing vaccines might maintain efficacy against new variations. However, further in-vitro and in-vivo studies are needed to validate the in-silico results and fully elucidate immune sensitivities to VOCs.
Collapse
Affiliation(s)
- Shivangi Sharma
- Bioinformatics and Data Management Group, ICMR-National Institute of Virology, Pune, Maharashtra, 411001, India
| | - Diya Roy
- Bioinformatics and Data Management Group, ICMR-National Institute of Virology, Pune, Maharashtra, 411001, India
| | - Sarah Cherian
- Bioinformatics and Data Management Group, ICMR-National Institute of Virology, Pune, Maharashtra, 411001, India.
| |
Collapse
|
38
|
de Los Rios Kobara I, Jayewickreme R, Lee MJ, Wilk AJ, Blomkalns AL, Nadeau KC, Yang S, Rogers AJ, Blish CA. Interferon-mediated NK cell activation is associated with limited neutralization breadth during SARS-CoV-2 infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619639. [PMID: 39484382 PMCID: PMC11527016 DOI: 10.1101/2024.10.22.619639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Best known for their ability to kill infected or malignant cells, natural killer (NK) cells are also underappreciated regulators of the antibody response to viral infection. In mice, NK cells can kill T follicular helper (Tfh) cells, decreasing somatic hypermutation and vaccine responses. Although human NK cell activation correlates with poor vaccine response, the mechanisms of human NK cell regulation of adaptive immunity are poorly understood. We found that in human ancestral SARS-CoV-2 infection, broad neutralizers, who were capable of neutralizing Alpha, Beta, and Delta, had fewer NK cells that expressed inhibitory and immaturity markers whereas NK cells from narrow neutralizers were highly activated and expressed interferon-stimulated genes (ISGs). ISG-mediated activation in NK cells from healthy donors increased cytotoxicity and functional responses to induced Tfh-like cells. This work reveals that NK cell activation and dysregulated inflammation may play a role in poor antibody response to SARS-CoV-2 and opens exciting avenues for designing improved vaccines and adjuvants to target emerging pathogens.
Collapse
|
39
|
Paul D, Verma J, Kumar S, Talukdar D, Jana P, Narendrakumar L, Kumar R, Tanwar S, Gosain M, Porey Karmakar S, Pareek M, Mani S, Chaudhuri S, Kshetrapal P, Wadhwa N, Bhatnagar S, Garg PK, Das B. A rapid point-of-care population-scale dipstick assay to identify and differentiate SARS-CoV-2 variants in COVID-19-positive patients. Front Microbiol 2024; 15:1459644. [PMID: 39498137 PMCID: PMC11532176 DOI: 10.3389/fmicb.2024.1459644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 10/09/2024] [Indexed: 11/07/2024] Open
Abstract
Delta and Omicron variants of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) are remarkably contagious, and have been recognized as variants of concern (VOC). The acquisition of spontaneous substitutions or insertion-deletion mutations (indels) in the spike protein-encoding gene substantially increases the binding affinity of the receptor binding domain (RBD)-hACE2 complex and upsurges the transmission of both variants. In this study, we analyzed thousands of genome sequences from 30 distinct SARS-CoV-2 variants, focusing on the unique nucleic acid signatures in the spike gene specific to the Delta and Omicron variants. Using these variant-specific sequences, we synthesized a range of oligonucleotides and optimized a multiplex PCR (mPCR) assay capable of accurately identifying and differentiating between the Delta and Omicron variants. Building on this mPCR assay, we developed a dipstick format by incorporating a tag linker sequence at the 5' end of the forward primer and adding biotin to the 3' end of the oligonucleotides, enhancing the assay's usability and accessibility. Streptavidin-coated latex beads and the dipstick imprinted with a probe for the tag linker sequence in the test strips were used for the detection assay. Our dipstick-based assay, developed as a rapid point-of-care test for identifying and differentiating SARS-CoV-2 variants has the potential to be used in low-resource settings and scaled up to the population level.
Collapse
Affiliation(s)
- Deepjyoti Paul
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Jyoti Verma
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Shakti Kumar
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Daizee Talukdar
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Pradipta Jana
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Lekshmi Narendrakumar
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Roshan Kumar
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Subhash Tanwar
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Mudita Gosain
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| | - Sonali Porey Karmakar
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Madhu Pareek
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Shailendra Mani
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Susmita Chaudhuri
- Multidisciplinary Clinical and Translational Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Pallavi Kshetrapal
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| | - Nitya Wadhwa
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| | - Shinjini Bhatnagar
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| | - Pramod Kumar Garg
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, India
| | - Bhabatosh Das
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
40
|
Kawser Z, Rahman S, Westeel E, Habib MT, Hossain M, Bulbul MRH, Mukta SA, Islam MZ, Hossain MZ, Afrad MH, Khan MH, Shirin T, Ahmed MS, Berland JL, Komurian-Pradel F, Qadri F. SARS-CoV-2 variant survey: Comparison of RT-PCR screening with TGS and variant distribution across two divisions of Bangladesh. PLoS One 2024; 19:e0311993. [PMID: 39418288 PMCID: PMC11486398 DOI: 10.1371/journal.pone.0311993] [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: 04/29/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND The widespread increase in multiple variants of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) since 2020 is causing significant health concerns worldwide. While whole-genome sequencing (WGS) has played a leading role in surveillance programs, many local laboratories lack the expertise and resources. Thus, we aimed to investigate the circulating SARS-CoV-2 variants and evaluate the performance of multiplexed real-time reverse transcription-PCR (RT-PCR) for screening and monitoring the emergence of new SARS-CoV-2 variants in Bangladesh. METHODS A total of 600 confirmed SARS-CoV-2-positive cases were enrolled either prospectively or retrospectively from two divisions of Bangladesh. The samples were screened by variant RT-PCR targeting five mutations of the spike gene (N501Y, P681R, L452R, E484K, E484Q). A subsample of the study population was also selected for third-generation sequencing (TGS) and the results were compared to the variant RT-PCR screening. An in-depth comparison was made between the two methods in terms of congruence and cost-benefit. RESULT Seven variants were detected among samples, with similar distributions of the variants across both divisions. Variant RT-PCR for the targeted mutations lead to a 98.5% call rate; only nine samples failed to be determined. No association was found regarding the demographic features, clinical criteria, or routine RT-PCR Ct values across the variants. The clade diversity of the sequenced subpopulation (n = 99) exhibited similar distributions across the two study sites and other epidemiologic variables. Variant RT-PCR successfully distinguished variants of concern (VOCs) and variants of interest (VOIs); however, 8% discrepancy was observed for the closest lineages. Moreover, the variant RT-PCR represented an ideal balance of cost, time, and accuracy that outweigh their limitations. CONCLUSION Based on the strong agreement of variant RT-PCR with TGS, such rapid, easily accessible approaches of rapid strain typing are essential in the context of pandemic responses to guide both treatment decisions and public health measures.
Collapse
Affiliation(s)
- Zannat Kawser
- Institute for Developing Science and Health Initiatives, Dhaka, Bangladesh
| | - Saikt Rahman
- Institute for Developing Science and Health Initiatives, Dhaka, Bangladesh
| | - Emilie Westeel
- Fondation Mérieux, Direction Médicale et Scientifique, Lyon, France
| | | | - Mohabbat Hossain
- Institute for Developing Science and Health Initiatives, Dhaka, Bangladesh
- Department of Genetic Engineering and Biotechnology, University of Chittagong, Chittagong, Bangladesh
| | - Md. Rakibul Hassan Bulbul
- Institute for Developing Science and Health Initiatives, Dhaka, Bangladesh
- Bangladesh Institute of Tropical and Infectious Disease (BITID), Fouzderhat, Chittagong, Bangladesh
| | | | - Md. Zahirul Islam
- Institute for Developing Science and Health Initiatives, Dhaka, Bangladesh
- Bangladesh Institute of Tropical and Infectious Disease (BITID), Fouzderhat, Chittagong, Bangladesh
| | - Md. Zakir Hossain
- Bangladesh Institute of Tropical and Infectious Disease (BITID), Fouzderhat, Chittagong, Bangladesh
| | - Mokibul Hassan Afrad
- International Centre for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | | | - Tahmina Shirin
- Institute of Epidemiology, Disease Control, and Research, Dhaka, Bangladesh
| | - Md. Shakeel Ahmed
- Bangladesh Institute of Tropical and Infectious Disease (BITID), Fouzderhat, Chittagong, Bangladesh
| | - Jean-Luc Berland
- Fondation Mérieux, Direction Médicale et Scientifique, Lyon, France
| | | | - Firdausi Qadri
- Institute for Developing Science and Health Initiatives, Dhaka, Bangladesh
- International Centre for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| |
Collapse
|
41
|
Cong B, Dong X, Yang Z, Yu P, Chai Y, Liu J, Zhang M, Zang Y, Kang J, Feng Y, Liu Y, Feng W, Wang D, Deng W, Li F, Song Z, Wang Z, Chen X, Qin H, Yu Q, Li Z, Liu S, Xu X, Zhong N, Ren X, Qin C, Liu L, Wang J, Cao X. Single-cell spatiotemporal analysis of the lungs reveals Slamf9 + macrophages involved in viral clearance and inflammation resolution. Cell Discov 2024; 10:104. [PMID: 39414783 PMCID: PMC11484945 DOI: 10.1038/s41421-024-00734-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/08/2024] [Indexed: 10/18/2024] Open
Abstract
How the lung achieves immune homeostasis after a pulmonary infection is not fully understood. Here, we analyzed the spatiotemporal changes in the lungs over a 2-week natural recovery from severe pneumonia in a Syrian hamster model of SARS-CoV-2 infection. We find that SARS-CoV-2 infects multiple cell types and causes massive cell death at the early stage, including alveolar macrophages. We identify a group of monocyte-derived Slamf9+ macrophages, which are induced after SARS-CoV-2 infection and resistant to impairment caused by SARS-CoV-2. Slamf9+ macrophages contain SARS-CoV-2, recruit and interact with Isg12+Cst7+ neutrophils to clear the viruses. After viral clearance, Slamf9+ macrophages differentiate into Trem2+ and Fbp1+ macrophages, contributing to inflammation resolution at the late stage, and finally replenish alveolar macrophages. These findings are validated in a SARS-CoV-2-infected hACE2 mouse model and confirmed with publicly available human autopsy single-cell RNA-seq data, demonstrating the potential role of Slamf9+ macrophages and their coordination with neutrophils in post-injury tissue repair and inflammation resolution.
Collapse
Affiliation(s)
- Boyi Cong
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuan Dong
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Zongheng Yang
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Pin Yu
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Yangyang Chai
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiaqi Liu
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Meihan Zhang
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China
| | | | | | - Yu Feng
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Yi Liu
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | | | - Dehe Wang
- Changping Laboratory, Beijing, China
| | - Wei Deng
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Fengdi Li
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhiqi Song
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Ziqiao Wang
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaosu Chen
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China
| | - Hua Qin
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China
| | - Qinyi Yu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhiqing Li
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Navy Medical University, Shanghai, China
- Guangzhou Laboratory, Guangzhou, Guangdong, China
| | - Shuxun Liu
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Navy Medical University, Shanghai, China
- Guangzhou Laboratory, Guangzhou, Guangdong, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | | | | | - Chuan Qin
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China.
| | - Longqi Liu
- BGI-Shenzhen, Shenzhen, Guangdong, China.
| | - Jian Wang
- BGI-Shenzhen, Shenzhen, Guangdong, China.
| | - Xuetao Cao
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China.
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
42
|
Cong B, Dong X, Yang Z, Yu P, Chai Y, Liu J, Zhang M, Zang Y, Kang J, Feng Y, Liu Y, Feng W, Wang D, Deng W, Li F, Song Z, Wang Z, Chen X, Qin H, Yu Q, Li Z, Liu S, Xu X, Zhong N, Ren X, Qin C, Liu L, Wang J, Cao X. Single-cell spatiotemporal analysis reveals alveolar dendritic cell-T cell immunity hubs defending against pulmonary infection. Cell Discov 2024; 10:103. [PMID: 39414763 PMCID: PMC11484931 DOI: 10.1038/s41421-024-00733-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/08/2024] [Indexed: 10/18/2024] Open
Abstract
How immune cells are spatiotemporally coordinated in the lung to effectively monitor, respond to, and resolve infection and inflammation in primed form needs to be fully illustrated. Here we apply immunocartography, a high-resolution technique that integrates spatial and single-cell RNA sequencing (scRNA-seq) through deconvolution and co-localization analyses, to the SARS-CoV-2-infected Syrian hamster model. We generate a comprehensive transcriptome map of the whole process of pulmonary infection from physiological condition, infection initiation, severe pneumonia to natural recovery at organ scale and single-cell resolution, with 142,965 cells and 45 lung lobes from 25 hamsters at 5 time points. Integrative analysis identifies that alveolar dendritic cell-T cell immunity hubs, where Ccr7+Ido1+ dendritic cells, Cd160+Cd8+ T cells, and Tnfrsf4+Cd4+ T cells physiologically co-localize, rapidly expand during SARS-CoV-2 infection, eliminate SARS-CoV-2 with the aid of Slamf9+ macrophages, and then restore to physiological levels after viral clearance. We verify the presence of these cell subpopulations in the immunity hubs in normal and SARS-CoV-2-infected hACE2 mouse models, as well as in publicly available human scRNA-seq datasets, demonstrating the potential broad relevance of our findings in lung immunity.
Collapse
Affiliation(s)
- Boyi Cong
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuan Dong
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Zongheng Yang
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Pin Yu
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Yangyang Chai
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiaqi Liu
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Meihan Zhang
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China
| | | | | | - Yu Feng
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Yi Liu
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | | | - Dehe Wang
- Changping Laboratory, Beijing, China
| | - Wei Deng
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Fengdi Li
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhiqi Song
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Ziqiao Wang
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaosu Chen
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China
| | - Hua Qin
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China
| | - Qinyi Yu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhiqing Li
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Navy Medical University, Shanghai, China
- Guangzhou Laboratory, Guangzhou, Guangdong, China
| | - Shuxun Liu
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Navy Medical University, Shanghai, China
- Guangzhou Laboratory, Guangzhou, Guangdong, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | | | | | - Chuan Qin
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China.
| | - Longqi Liu
- BGI-Shenzhen, Shenzhen, Guangdong, China.
| | - Jian Wang
- BGI-Shenzhen, Shenzhen, Guangdong, China.
| | - Xuetao Cao
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China.
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
43
|
Li PH, Xu H, Xie CY, Ji ZL, Deng YY, Li X, Fang M. The short-term and long-term prognosis of discharged COVID-19 patients in Guangdong during the first wave of pandemic. Sci Rep 2024; 14:23994. [PMID: 39402204 PMCID: PMC11473718 DOI: 10.1038/s41598-024-76013-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
COVID-19 survivors concerning about the rehabilitation and sustained sequelae of Coronavirus Disease 2019 (COVID-19) infection. We aimed to investigate the sequelae of patients' psychological and physical condition and its related factors in the early and late stages. This longitudinal study tracked 281 COVID-19 patients discharged from hospitals in Guangdong, China, for one year. Assessments occurred at 2,4,12,24 and 48 weeks post-discharge. We define 2 weeks, 4 weeks, and 12 weeks as early stage, and 24 weeks and 48 weeks as late stage. Psychological health was measured using the Patient Health Questionnaire-9 (PHQ-9), Generalized Anxiety Disorder-7 (GAD-7), Posttraumatic Stress Disorder Checklist for DSM-5 (PCL-5), and Pittsburgh Sleep Quality Index (PSQI) scales. Physical health was assessed through laboratory tests, chest computed tomography (CT) scans, and pulmonary function tests. Data were analyzed using multivariate regression models to evaluate the influence of demographic and clinical variables on health outcomes. COVID-19 survivors exhibited psychological and physical sequelae in both the early and late stages. Compared to the early stage, the proportions of patients with depression (early stage 14.6%, late stage 4.6%), anxiety (early stage 8.9%, late stage 5.3%), PTSD(early stage 3.6%, late stage 0.7%), abnormal liver function (early stage 24.6%, late stage 11.0%), abnormal cardiac function (early stage 10.0%, late stage 7.8%), abnormal renal function (early stage 20.6%, late stage 11.0%) and abnormal pulmonary function (early stage 40.9%, late stage 13.5%) were significantly reduced in the late stage. Factors such as gender, age, severity of COVID-19, hospitalization duration, and various comorbidities were significantly associated with these sequelae. We noticed that psychological and physical sequelae occurred to COVID-19 survivors in short and long stages, and these would gradually decrease as time went on. Male gender, age > 50 years old, severe clinical condition, longer hospitalization time and comorbidity history were related factors that significantly affected the rehabilitation of COVID-19 patients.
Collapse
Grants
- No. 2020B1111330006 The Health Commission of Guangdong Province, the Department of Science and Technology of Guangdong Province, and the Special Project on Emergency Response to Control of Novel Coronavirus Infection of Guangdong Province
- No. 2020B1111330006 The Health Commission of Guangdong Province, the Department of Science and Technology of Guangdong Province, and the Special Project on Emergency Response to Control of Novel Coronavirus Infection of Guangdong Province
- No. 2020B1111330006 The Health Commission of Guangdong Province, the Department of Science and Technology of Guangdong Province, and the Special Project on Emergency Response to Control of Novel Coronavirus Infection of Guangdong Province
- No. 2020B1111330006 The Health Commission of Guangdong Province, the Department of Science and Technology of Guangdong Province, and the Special Project on Emergency Response to Control of Novel Coronavirus Infection of Guangdong Province
- No. 2020B1111330006 The Health Commission of Guangdong Province, the Department of Science and Technology of Guangdong Province, and the Special Project on Emergency Response to Control of Novel Coronavirus Infection of Guangdong Province
- No. 2020B1111330006 The Health Commission of Guangdong Province, the Department of Science and Technology of Guangdong Province, and the Special Project on Emergency Response to Control of Novel Coronavirus Infection of Guangdong Province
- No. 2020B1111330006 The Health Commission of Guangdong Province, the Department of Science and Technology of Guangdong Province, and the Special Project on Emergency Response to Control of Novel Coronavirus Infection of Guangdong Province
Collapse
Affiliation(s)
- Pei-Hong Li
- Graduate College, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Hui Xu
- Department of Emergency, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China
| | - Cheng-Yuan Xie
- Department of Critical Care Medicine, Sun Yat-Sen University 2nd Affiliated Hospital, Guangzhou, 510000, Guangdong, China
| | - Zhong-Liang Ji
- Department of Emergency, Shenzhen University General Hospital, Shenzhen, 518055, Guangdong, China
| | - Yi-Yu Deng
- Department of Critical Care Medicine, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Xin Li
- Department of Emergency, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China
| | - Ming Fang
- Department of Critical Care Medicine, Xiaolan Affiliated Hospital, Southern Medical University, Zhongshan, 528415, Guangdong, China.
- , 65 Ju Cheng Avenue Middle Section, 528415, Xiaolan Town, Zhongshan, P.R. China.
| |
Collapse
|
44
|
Uyar Y, Mart Kömürcü SZ, Artik Y, Cesur NP, Tanrıverdi A, Şanlı K. The evaluation of SARS-CoV-2 mutations at the early stage of the pandemic in Istanbul population. Ann Clin Microbiol Antimicrob 2024; 23:93. [PMID: 39390548 PMCID: PMC11468081 DOI: 10.1186/s12941-024-00750-y] [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: 07/29/2024] [Accepted: 09/21/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Determination of SARS-CoV-2 variant is significant to prevent the spreads of COVID-19 disease. METHODS We aimed to evaluate the variants of SARS-CoV-2 rate in positive patients in Kanuni Sultan Suleyman Training and Research Hospital (KSS-TRH), Istanbul, Türkiye between 1st January and 30th November 2021 by using RT-PCR method. RESULTS Herein, 825,169 patients were evaluated (male:58.53% and female:41.47%) whether COVID-19 positive or not [( +):21.3% and (-):78.7%] and 175,367 patient was described as positive (53.2%-female and 46.8%-male) by RT-PCR. COVID-19 positive rate is observed highest in the 6-15- and 66-75-year age range. The frequencies were obtained as SARS-CoV-2 positive (without mutation of B.1.1.7 [B.1.1.7 (U.K), E484K, L452R, B.1.351 (S. Africa/Brazil) spike mutations] as 66.1% (n: 115,899), B.1.1.7 Variant as 23.2% (n:40,686), Delta mutation (L452R) variant as 9.8% (n:17,182), B.1.351 variant as 0.8% (n:1370) and E484K as 0.1% (n: 230). In April 2021, general SARS-CoV-2 and B.1.1.7 variant were dominantly observed. Up to July 2021, B.1.617.2 (Delta variant/ Indian variant) and E484K has been not observed. B.1.351 variant of SARS-CoV-2 has been started in February 2021 at the rarest ratio and March 2021 is the top point. September 2021 is the pick point of E484K. African/Brazil variant of SARS-CoV-2 has been started in February 2021 at the rarest ratio and March 2021 is the top point. September 2021 is the pick point of E484K. When the gender type is compared within the variants, women were found to be more prevalent in all varieties. CONCLUSIONS The meaning of these mutations is very important to understand the transmission capacity of the COVID-19 disease, pandemic episode, and diagnosis of the virus with mutation types. Understanding the variant type is important for monitoring herd immunity and the spread of the disease.
Collapse
Affiliation(s)
- Yavuz Uyar
- Cerrahpaşa Faculty of Medicine, Department of Medical Microbiology, Istanbul University-Cerrahpaşa, 34147, Istanbul, Türkiye.
| | - Selen Zeliha Mart Kömürcü
- Republic of Türkiye, Istanbul Provincial Directorate of Health, Ministry of Health, University of Health Science, Kanuni Sultan Suleyman Training and Research Hospital, Kücükcekmece, 34303, Istanbul, Türkiye
| | - Yakup Artik
- Health Institutes of Türkiye (TUSEB), COVID-19 Diagnostic Center, Istanbul Provincial Directorate of Health, Republic of Türkiye Ministry of Health, University of Health Science, Kanuni Sultan Suleyman Training and Research Hospital, Kücükcekmece, 34303, Istanbul, Türkiye
| | - Nevra Pelin Cesur
- Health Institutes of Türkiye (TUSEB), COVID-19 Diagnostic Center, Istanbul Provincial Directorate of Health, Republic of Türkiye Ministry of Health, University of Health Science, Kanuni Sultan Suleyman Training and Research Hospital, Kücükcekmece, 34303, Istanbul, Türkiye
| | - Arzu Tanrıverdi
- Republic of Türkiye, Istanbul Provincial Directorate of Health, Ministry of Health, University of Health Science, Kanuni Sultan Suleyman Training and Research Hospital, Kücükcekmece, 34303, Istanbul, Türkiye
| | - Kamuran Şanlı
- Republic of Türkiye, Istanbul Provincial Directorate of Health, Ministry of Health, University of Health Science, Başakşehir Çam and Sakura City Hospital, Başakşehir, 34480, Istanbul, Türkiye
| |
Collapse
|
45
|
Djorwé S, Malki A, Nzoyikorera N, Nyandwi J, Zebsoubo SP, Bellamine K, Bousfiha A. Genetic diversity and genomic epidemiology of SARS-CoV-2 during the first 3 years of the pandemic in Morocco: comprehensive sequence analysis, including the unique lineage B.1.528 in Morocco. Access Microbiol 2024; 6:000853.v4. [PMID: 39376591 PMCID: PMC11457919 DOI: 10.1099/acmi.0.000853.v4] [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: 05/22/2024] [Accepted: 09/16/2024] [Indexed: 10/09/2024] Open
Abstract
During the 3 years following the emergence of the COVID-19 pandemic, the African continent, like other regions of the world, was substantially impacted by COVID-19. In Morocco, the COVID-19 pandemic has been marked by the emergence and spread of several SARS-CoV-2 variants, leading to a substantial increase in the incidence of infections and deaths. Nevertheless, the comprehensive understanding of the genetic diversity, evolution, and epidemiology of several viral lineages remained limited in Morocco. This study sought to deepen the understanding of the genomic epidemiology of SARS-CoV-2 through a retrospective analysis. The main objective of this study was to analyse the genetic diversity of SARS-CoV-2 and identify distinct lineages, as well as assess their evolution during the pandemic in Morocco, using genomic epidemiology approaches. Furthermore, several key mutations in the functional proteins across different viral lineages were highlighted along with an analysis of the genetic relationships amongst these strains to better understand their evolutionary pathways. A total of 2274 genomic sequences of SARS-CoV-2 isolated in Morocco during the period of 2020 to 2023, were extracted from the GISAID EpiCoV database and subjected to analysis. Lineages and clades were classified according to the nomenclature of GISAID, Nextstrain, and Pangolin. The study was conducted and reported in accordance with STROBE (Strengthening the Reporting of Observational Studies in Epidemiology) guidelines. An exhaustive analysis of 2274 genomic sequences led to the identification of 157 PANGO lineages, including notable lineages such as B.1, B.1.1, B.1.528, and B.1.177, as well as variants such as B.1.1.7, B.1.621, B.1.525, B.1.351, B.1.617.1, B.1.617.2, and its notable sublineages AY.33, AY.72, AY.112, AY.121 that evolved over time before being supplanted by Omicron in December 2021. Among the 2274 sequences analysed, Omicron and its subvariants had a prevalence of 59.5%. The most predominant clades were 21K, 21L, and 22B, which are respectively related phylogenetically to BA.1, BA.2, and BA.5. In June 2022, Morocco rapidly observed a recrudescence of cases of infection, with the emergence and concurrent coexistence of subvariants from clade 22B such as BA.5.2.20, BA.5, BA.5.1, BA.5.2.1, and BF.5, supplanting the subvariants BA.1 (clade display 21K) and BA.2 (clade display 21L), which became marginal. However, XBB (clade 22F) and its progeny such XBB.1.5(23A), XBB.1.16(23B), CH.1.1(23C), XBB.1.9(23D), XBB.2.3(23E), EG.5.1(23F), and XBB.1.5.70(23G) have evolved sporadically. Furthermore, several notable mutations, such as H69del/V70del, G142D, K417N, T478K, E484K, E484A, L452R, F486P, N501Y, Q613H, D614G, and P681H/R, have been identified. Some of these SARS-CoV-2 mutations are known to be involved in increasing transmissibility, virulence, and antibody escape. This study has identified several distinct lineages and mutations involved in the genetic diversity of Moroccan isolates, as well as the analysis of their evolutionary trends. These findings provide a robust basis for better understanding the distinct mutations and their roles in the variation of transmissibility, pathogenicity, and antigenicity (immune evasion/reinfection). Furthermore, the noteworthy number of distinct lineages identified in Morocco highlights the importance of maintaining continuous surveillance of COVID-19. Moreover, expanding vaccination coverage would also help protect patients against more severe clinical disease.
Collapse
Affiliation(s)
- Soulandi Djorwé
- Laboratory of Physiopathology and Molecular Genetics, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca (Morocco), Avenue Cdt Driss El Harti, PB 7955 Sidi Othman, Casablanca, Morocco
- Bourgogne Laboratory of Medical and Scientific Analysis, 136, residence belhcen, Bd Bourgogne, Casablanca, Morocco
| | - Abderrahim Malki
- Laboratory of Physiopathology and Molecular Genetics, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca (Morocco), Avenue Cdt Driss El Harti, PB 7955 Sidi Othman, Casablanca, Morocco
| | - Néhémie Nzoyikorera
- National Reference Laboratory, National Institute of Public Health, Bujumbura, Burundi
- Higher Institute of Biosciences and Biotechnology, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
- Laboratory of Microbial Biotechnology and Infectiology Research, Mohammed VI Center for Research & Innovation, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Joseph Nyandwi
- Département de Médecine, Faculté de Médecine, Université du Burundi, Bujumbura, Burundi
- Ministère de la Santé Publique et de la Lutte contre le Sida, Institut National de Santé Publique de Bujumbura, Bujumbura, Burundi
| | - Samuel Privat Zebsoubo
- School of Advanced Studies in Biotechnology and Private Health (EHEB), 183 Bd de la Resistance, Casablanca 20250, Morocco
| | - Kawthar Bellamine
- Bourgogne Laboratory of Medical and Scientific Analysis, 136, residence belhcen, Bd Bourgogne, Casablanca, Morocco
| | - Amale Bousfiha
- Laboratory of Physiopathology and Molecular Genetics, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca (Morocco), Avenue Cdt Driss El Harti, PB 7955 Sidi Othman, Casablanca, Morocco
| |
Collapse
|
46
|
Rosales R, McGovern BL, Rodriguez ML, Leiva-Rebollo R, Diaz-Tapia R, Benjamin J, Rai DK, Cardin RD, Anderson AS, Sordillo EM, van Bakel H, Simon V, García-Sastre A, White KM. Nirmatrelvir and molnupiravir maintain potent in vitro and in vivo antiviral activity against circulating SARS-CoV-2 omicron subvariants. Antiviral Res 2024; 230:105970. [PMID: 39067667 DOI: 10.1016/j.antiviral.2024.105970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 06/27/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Variants of SARS-CoV-2 pose significant challenges in public health due to their increased transmissibility and ability to evade natural immunity, vaccine protection, and monoclonal antibody therapeutics. The emergence of the highly transmissible Omicron variant and subsequent subvariants, characterized by an extensive array of over 32 mutations within the spike protein, intensifies concerns regarding vaccine evasion. In response, multiple antiviral therapeutics have received FDA emergency use approval, targeting the SARS-CoV-2 RNA-dependent RNA polymerase (RdRp) and main protease (Mpro) regions, known to have relatively fewer mutations across novel variants. In this study, we evaluated the efficacy of nirmatrelvir (PF-07321332) and other clinically significant SARS-CoV-2 antivirals against a diverse panel of SARS-CoV-2 variants, encompassing the newly identified Omicron subvariants XBB1.5 and JN.1, using live-virus antiviral assays. Our findings demonstrate that while the last Omicron subvariants exhibited heightened pathogenicity in our animal model, nirmatrelvir and other clinically relevant antivirals consistently maintained their efficacy against all tested variants, including the XBB1.5 subvariant.
Collapse
Affiliation(s)
- Romel Rosales
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Briana L McGovern
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - M Luis Rodriguez
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rocio Leiva-Rebollo
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Randy Diaz-Tapia
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jared Benjamin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Devendra K Rai
- Worldwide Research and Development, Pfizer Inc., Pearl River, NY, 10965, USA
| | - Rhonda D Cardin
- Worldwide Research and Development, Pfizer Inc., Pearl River, NY, 10965, USA
| | | | - Emilia Mia Sordillo
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Harm van Bakel
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Artificial Intelligence And Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Viviana Simon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kris M White
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
47
|
Shahriyari N, Omidvar S, Mohsenzadeh‐Ledari F, Azizi A, Gholinia H. The Impact of Mindfulness-Based Counseling on the Mental Health of Women With a History of COVID-19 During Pregnancy: A Quasi-Experimental Study. Brain Behav 2024; 14:e70062. [PMID: 39350632 PMCID: PMC11442985 DOI: 10.1002/brb3.70062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 08/23/2024] [Accepted: 09/02/2024] [Indexed: 10/04/2024] Open
Abstract
INTRODUCTION With the spread of COVID-19, certain population groups, including pregnant women, were more susceptible than others. This disease can lead to postpartum complications, including mental disorders, in mothers. Few studies have investigated the impact of mindfulness-based interventions on mental health, and the most effective counseling approach to promote mental health has not been identified. OBJECTIVE This study aimed to determine the impact of online mindfulness-based counseling on improving mental health among women with a history of COVID-19 during pregnancy in Iran. METHODS The present study was a quasi-experimental design conducted on 100 women with a history of coronavirus infection during pregnancy referred to the Mother's Clinic of Yahya Nejad and Ayatollah Rouhani Educational-Treatment Hospital, affiliated with Babol University of Medical Sciences, Iran, via convenience sampling. The women were randomly assigned to the intervention (mindfulness-based counseling) and control groups. The intervention group received eight 45-min weekly mindfulness-based counseling sessions over 8 weeks. Data were collected via a demographic information questionnaire and the Goldberg General Health Questionnaire before and after the intervention, which were completed by both groups. Independent t-tests and analysis of covariances (ANCOVAs) were used to compare the outcomes of the two groups. RESULTS After controlling for confounding variables, the mean mental health scores before and after counseling were 29.42 ± 4.49 and 19.80 ± 3.88, respectively, in the intervention group and 26.26 ± 2.29 and 25.92 ± 2.15, respectively, in the control group. The mean mental health score in the intervention group was significantly lower than that in the control group (F = 266.7, p < 0.001). The mean scores for somatic symptoms (F = 89.30, p < 0.001), depression symptoms (F = 142.71, p < 0.001), anxiety and insomnia symptoms (F = 120.56, p < 0.001), and social dysfunction scores (F = 127.77, p < 0.001) were significantly different between the two groups after counseling. CONCLUSION The findings indicated that online mindfulness-based counseling positively affects mental health and its domains during the postpartum period. However, further randomized clinical trials are needed before a definitive conclusion can be drawn. TRIAL REGISTRATION We were not allowed to register according to the law of our country.
Collapse
Affiliation(s)
- Najmeh Shahriyari
- Student Research CommitteeBabol University of Medical SciencesBabolIran
| | - Shabnam Omidvar
- Social Determinants of Health Research Center, Health Research InstituteBabol University of Medical SciencesBabolIran
| | - Farideh Mohsenzadeh‐Ledari
- Social Determinants of Health Research Center, Health Research InstituteBabol University of Medical SciencesBabolIran
| | - Alireza Azizi
- Social Determinants of Health Research Center, Health Research InstituteBabol University of Medical SciencesBabolIran
| | | |
Collapse
|
48
|
Yamasaki M, Saso W, Yamamoto T, Sato M, Takagi H, Hasegawa T, Kozakura Y, Yokoi H, Ohashi H, Tsuchimoto K, Hashimoto R, Fukushi S, Uda A, Muramatsu M, Takayama K, Maeda K, Takahashi Y, Nagase T, Watashi K. Anti-SARS-CoV-2 gapmer antisense oligonucleotides targeting the main protease region of viral RNA. Antiviral Res 2024; 230:105992. [PMID: 39181215 DOI: 10.1016/j.antiviral.2024.105992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/28/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Given the worldwide risk for the outbreak of emerging/re-emerging respiratory viruses, establishment of new antiviral strategies is greatly demanded. In this study, we present a scheme to identify gapmer antisense oligonucleotides (ASOs) targeting severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA that efficiently inhibit viral replication. We synthesized approximately 300 gapmer ASOs designed to target various SARS-CoV-2 RNA regions and evaluated their activity in cell-based assays. Through a multistep screening in cell culture systems, we identified that ASO#41, targeting the coding region for viral main protease, reduced SARS-CoV-2 RNA levels in infected cells and inhibited virus-induced cytopathic effects. Antiviral effect of ASO#41 was also observed in iPS cell-derived human lung organoids. ASO#41 depleted intracellular viral RNAs during genome replication in an endogenous RNaseH-dependent manner. ASO#41 showed a wide range of antiviral activity against SARS-CoV-2 variants of concern including Alpha, Delta, and Omicron. Intranasal administration to mice exhibited intracellular accumulation of ASO#41 in the lung and significantly reduced the viral infectious titer, with milder body weight loss due to SARS-CoV-2 infection. Further chemical modification with phosphoryl guanidine-containing backbone linkages provided an elevation of anti-SARS-CoV-2 activity, with 23.4 nM of 50% antiviral inhibitory concentration, one of the strongest anti-SARS-CoV-2 ASOs reported so far. Our study presents an approach to identify active ASOs against SARS-CoV-2, which is potentially useful for establishing an antiviral strategy by targeting genome RNA of respiratory viruses.
Collapse
Affiliation(s)
- Masako Yamasaki
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan; Department of Virology II, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan; Department of Applied Biological Sciences, Tokyo University of Science, Noda, 278-8510, Japan
| | - Wakana Saso
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Takuya Yamamoto
- Infectious Diseases Unit, Tokushima Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Tokushima, 771-0192, Japan
| | - Masayoshi Sato
- Infectious Diseases Unit, Tokushima Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Tokushima, 771-0192, Japan
| | - Hiroko Takagi
- Infectious Diseases Unit, Tokushima Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Tokushima, 771-0192, Japan
| | - Tetsuya Hasegawa
- Department of Medicinal Chemistry, Tokushima Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Tokushima, 771-0192, Japan
| | - Yuji Kozakura
- Department of Drug Discovery Strategy, Office of Bioinformatics, Otsuka Pharmaceutical Co., Ltd., Tokushima, 771-0192, Japan
| | - Hiroyuki Yokoi
- Department of Drug Metabolism and Pharmacokinetics, Preclinical Research, Tokushima Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Tokushima, 771-0192, Japan
| | - Hirofumi Ohashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Kana Tsuchimoto
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Rina Hashimoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Shuetsu Fukushi
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Akihiko Uda
- Department of Veterinary Science, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Masamichi Muramatsu
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan; Foundation for Biomedical Research and Innovation at Kobe, Institute of Biomedical Research and Innovation, Kobe, 650-0047, Japan
| | - Kazuo Takayama
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Ken Maeda
- Department of Veterinary Science, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Tsuyoshi Nagase
- Department of Medicinal Chemistry, Tokushima Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Tokushima, 771-0192, Japan
| | - Koichi Watashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan; Department of Virology II, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan; Department of Applied Biological Sciences, Tokyo University of Science, Noda, 278-8510, Japan; MIRAI, JST, Tokyo, 102-0076, Japan.
| |
Collapse
|
49
|
Moraes Dos Santos L, Gutembergue de Mendonça J, Jerônimo Gomes Lobo Y, Henrique Franca de Lima L, Bruno Rocha G, C de Melo-Minardi R. Deep learning for discriminating non-trivial conformational changes in molecular dynamics simulations of SARS-CoV-2 spike-ACE2. Sci Rep 2024; 14:22639. [PMID: 39349594 PMCID: PMC11443059 DOI: 10.1038/s41598-024-72842-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 09/11/2024] [Indexed: 10/04/2024] Open
Abstract
Molecular dynamics (MD) simulations produce a substantial volume of high-dimensional data, and traditional methods for analyzing these data pose significant computational demands. Advances in MD simulation analysis combined with deep learning-based approaches have led to the understanding of specific structural changes observed in MD trajectories, including those induced by mutations. In this study, we model the trajectories resulting from MD simulations of the SARS-CoV-2 spike protein-ACE2, specifically the receptor-binding domain (RBD), as interresidue distance maps, and use deep convolutional neural networks to predict the functional impact of point mutations, related to the virus's infectivity and immunogenicity. Our model was successful in predicting mutant types that increase the affinity of the S protein for human receptors and reduce its immunogenicity, both based on MD trajectories (precision = 0.718; recall = 0.800; [Formula: see text] = 0.757; MCC = 0.488; AUC = 0.800) and their centroids. In an additional analysis, we also obtained a strong positive Pearson's correlation coefficient equal to 0.776, indicating a significant relationship between the average sigmoid probability for the MD trajectories and binding free energy (BFE) changes. Furthermore, we obtained a coefficient of determination of 0.602. Our 2D-RMSD analysis also corroborated predictions for more infectious and immune-evading mutants and revealed fluctuating regions within the receptor-binding motif (RBM), especially in the [Formula: see text] loop. This region presented a significant standard deviation for mutations that enable SARS-CoV-2 to evade the immune response, with RMSD values of 5Å in the simulation. This methodology offers an efficient alternative to identify potential strains of SARS-CoV-2, which may be potentially linked to more infectious and immune-evading mutations. Using clustering and deep learning techniques, our approach leverages information from the ensemble of MD trajectories to recognize a broad spectrum of multiple conformational patterns characteristic of mutant types. This represents a strategic advantage in identifying emerging variants, bypassing the need for long MD simulations. Furthermore, the present work tends to contribute substantially to the field of computational biology and virology, particularly to accelerate the design and optimization of new therapeutic agents and vaccines, offering a proactive stance against the constantly evolving threat of COVID-19 and potential future pandemics.
Collapse
Affiliation(s)
- Lucas Moraes Dos Santos
- Department of Computer Science, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| | | | - Yan Jerônimo Gomes Lobo
- Department of Exact and Biological Sciences, Federal University of São João Del Rei, São João del Rei, Minas Gerais, Brazil
| | | | - Gerd Bruno Rocha
- Department of Chemistry, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | - Raquel C de Melo-Minardi
- Department of Computer Science, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
50
|
Liu Q, Chen Y, Qi H. Advances in Genotyping Detection of Fragmented Nucleic Acids. BIOSENSORS 2024; 14:465. [PMID: 39451678 PMCID: PMC11506436 DOI: 10.3390/bios14100465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024]
Abstract
Single nucleotide variant (SNV) detection is pivotal in various fields, including disease diagnosis, viral screening, genetically modified organism (GMO) identification, and genotyping. However, detecting SNVs presents significant challenges due to the fragmentation of nucleic acids caused by cellular apoptosis, molecular shearing, and physical degradation processes such as heating. Fragmented nucleic acids often exhibit variable lengths and inconsistent breakpoints, complicating the accurate detection of SNVs. This article delves into the underlying causes of nucleic acid fragmentation and synthesizes the strengths and limitations of next-generation sequencing technology, high-resolution melting curves, molecular probes, and CRISPR-based approaches for SNV detection in fragmented nucleic acids. By providing a detailed comparative analysis, it seeks to offer valuable insights for researchers working to overcome the challenges of SNV detection in fragmented samples, ultimately advancing the accurate and efficient detection of single nucleotide variants across diverse applications.
Collapse
Affiliation(s)
- Qian Liu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; (Q.L.); (Y.C.)
- Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Yun Chen
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; (Q.L.); (Y.C.)
- Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Hao Qi
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; (Q.L.); (Y.C.)
- Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| |
Collapse
|