1
|
Nency YM, Sekartini R, Wirahmadi A, Maria S, Farhanah N, Hapsari R, Mahati E, Maharani N, Mulyono M, Anantyo DT, Pramudo SG, Yeremia GF, Setyaningsih L, Puspita M, Sari RM. Safety and immunogenicity of SARS-CoV-2 protein subunit recombinant vaccine (Indovac®) in healthy populations aged 18 years and above in Indonesia: A phase I, observer-blind, randomized, controlled study. Hum Vaccin Immunother 2025; 21:2501467. [PMID: 40381203 PMCID: PMC12087486 DOI: 10.1080/21645515.2025.2501467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 04/14/2025] [Accepted: 04/30/2025] [Indexed: 05/20/2025] Open
Abstract
Indonesian vaccine producer (PT BIOFARMA), conducted a study to assess the safety and immunogenicity of a new COVID-19 vaccine candidate. This vaccine is based on a recombinant subunit protein platform, with the SARS-CoV-2 receptor-binding domain (RBD) as its target antigen. The study compared the candidate's safety and immunogenicity to the control group vaccine, the Sinovac vaccine, 28 days after administration. This was an observer-blinded and randomized Phase 1 trial which recruited 175 subjects. The subject received 0.5 ml of vaccine in two doses. The subjects were split into five treatment groups, consisting of different combinations of doses between RBD and CpG. The safety of this vaccine was evaluated within 7 days after the first dose and for 6 months after the second dose, while the immunogenicity was evaluated on days 14 and 28 after the second dose. The overall incidence of AEs was 54.86% from the beginning of the vaccination to 28 days after each injection. Most AEs were local pain and had no serious AEs. The study revealed a significant rise in the Geometric Mean Titer (GMT) of IgG antibodies in every group, indicating a strong immune response. The phase I data demonstrated that the new vaccine candidate showed promising results in safety and immunogenicity.
Collapse
MESH Headings
- Humans
- Indonesia
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/adverse effects
- COVID-19 Vaccines/administration & dosage
- Female
- Adult
- Male
- Middle Aged
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/adverse effects
- Vaccines, Synthetic/administration & dosage
- COVID-19/prevention & control
- COVID-19/immunology
- Antibodies, Viral/blood
- Immunogenicity, Vaccine
- Vaccines, Subunit/immunology
- Vaccines, Subunit/adverse effects
- Vaccines, Subunit/administration & dosage
- SARS-CoV-2/immunology
- Young Adult
- Single-Blind Method
- Adolescent
- Aged
- Spike Glycoprotein, Coronavirus/immunology
- Immunoglobulin G/blood
- Antibodies, Neutralizing/blood
Collapse
Affiliation(s)
- Yetty Movieta Nency
- Child Health Department, Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | - Rini Sekartini
- Department of Child Health, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Angga Wirahmadi
- Department of Child Health, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Suzy Maria
- Allergy and Clinical Immunology Division, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Nur Farhanah
- Department of Internal Medicine, Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | - Rebriarina Hapsari
- Department of Microbiology, Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | - Endang Mahati
- Department of Pharmacology and Therapy, Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | - Nani Maharani
- Department of Pharmacology and Therapy, Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | - Mulyono Mulyono
- Child Health Department, Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | - Dimas Tri Anantyo
- Child Health Department, Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | - Setyo Gundi Pramudo
- Department of Internal Medicine, Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | - Glenn Fernandez Yeremia
- COVID 19 Vaccine Bio Farma-Universitas Diponegoro (COBIU) Study, Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | | | - Mita Puspita
- Clinical Trial Division, PT Bio Farma, Bandung, Indonesia
| | | |
Collapse
|
2
|
El Gamal AA, Al-Massarani SM, Abdelmageed LWM, Basudan OA, Rehman MT, AlAjmi MF, Abdel-Kader MS, Parveen M, Hefnawy MM. New iridoids and phenolic glycosides from nuxia congesta R.Br. ex Fresen with potential inhibitors of SARS-CoV-2 main protease and spike RBD: an in silico approach. Nat Prod Res 2025:1-11. [PMID: 40411812 DOI: 10.1080/14786419.2025.2509883] [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: 10/27/2024] [Revised: 05/05/2025] [Accepted: 05/19/2025] [Indexed: 05/26/2025]
Abstract
The aerial parts of Nuxia congesta R.Br. ex Fresen were chromatographically purified, yielding five new compounds: two iridoids (1,2), one phenylpropanoid glycoside (3), and two rhamnosyl esters (4,5). The compounds were characterised via NMR and HRESIMS. The isolated compounds inhibitory action towards the COVID-19 major protease (Mpro) and receptor-binding domain (RBD) enzymes was evaluated. Comparative molecular docking and binding free energy analyses were conducted based on the binding affinities of the ligand binding domains of the therapeutic targets Mpro and RBD. The results indicated that all compounds effectively interacted with the binding sites of Mpro and RBD receptors, exhibiting a greater affinity for the Mpro receptor. The binding energy to Mpro ranged between -7.8 and -8.1 kcal/mol, however for RBD receptors it ranged from -7.0 to -7.3 kcal/mol. This research supports the efficacy of isolated compounds as antiviral medications, especially in the context of COVID-19.
Collapse
Affiliation(s)
- Ali A El Gamal
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Shaza M Al-Massarani
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Lena W M Abdelmageed
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Omer A Basudan
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Md Tabish Rehman
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed F AlAjmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Maged S Abdel-Kader
- Department of Pharmacognosy, College of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Mehtab Parveen
- Department of Chemistry, Faculty of Science, Aligarh Muslim University, Aligarh, India
| | - Mohamed M Hefnawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
3
|
Simovic A, Radomirovic M, Gligorijevic N, Milcic M, Bicanin M, Minic S, Stojanovic M, Stanic-Vucinic D, Cirkovic Velickovic T. Food-derived bioactive pigment phycocyanobilin binds to SARS-CoV-2 spike protein both covalently and noncovalently affecting its conformation and functionality. Arch Biochem Biophys 2025; 770:110475. [PMID: 40404003 DOI: 10.1016/j.abb.2025.110475] [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: 10/08/2024] [Revised: 05/16/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025]
Abstract
Phycocyanobilin (PCB), tetrapyrrole chromophore of Spirulina phycocyanin, is bilirubin analog and weak thiol-modifying agent. SARS-CoV-2 spike protein (SP) has bilirubin binding pocket, lacks free sulfhydryl, but it has two pairs of functionally important semi-stable disulfides reactive towards thiol-modifying agents. We investigated covalent and noncovalent binding of PCB to SP and its receptor-binding domain (RBD) and impact of covalent PCB conjugation to RBD on structure and binding to human angiotensin-converting enzyme 2 (ACE-2). PCB shows high-affinity for SP (Ka = 2.1 × 107 M-1), moderate-affinity for RBD (Ka = 8.4 × 104 M-1) and binds covalently to SP and RBD in reaction involving thiols. PCB binding alters RBD conformation. Molecular docking identified two binding sites of PCB to SP, bilirubin/biliverdin binding site and hydrophobic pocket of RBD in vicinity of Cys432, preferential target for covalent binding in in silico covalent docking of PCB to RBD. Redox proteomics mapped reactive Cys432, Cys391 and Cys525 in RBD. PCB-modified RBD exhibited reduced ability to bind to ACE-2. This is the first study demonstrating PCB reactivity towards semi-stable disulfides of proteins lacking free sulfhydryl groups. PCB may affect functionality and structure of SP and its RBD by noncovalent and covalent binding.
Collapse
Affiliation(s)
- Ana Simovic
- Centre of Excellence for Molecular Food Sciences and Department of Biochemistry, University of Belgrade, Faculty of Chemistry, 11000, Belgrade, Serbia
| | - Mirjana Radomirovic
- Centre of Excellence for Molecular Food Sciences and Department of Biochemistry, University of Belgrade, Faculty of Chemistry, 11000, Belgrade, Serbia
| | - Nikola Gligorijevic
- Center for Chemistry, University of Belgrade, Institute of Chemistry, Technology and Metallurgy, National Institute of the Republic of Serbia, 11000, Belgrade, Serbia
| | - Milos Milcic
- Centre of Excellence for Molecular Food Sciences and Department of Biochemistry, University of Belgrade, Faculty of Chemistry, 11000, Belgrade, Serbia
| | - Masa Bicanin
- Centre of Excellence for Molecular Food Sciences and Department of Biochemistry, University of Belgrade, Faculty of Chemistry, 11000, Belgrade, Serbia
| | - Simeon Minic
- Centre of Excellence for Molecular Food Sciences and Department of Biochemistry, University of Belgrade, Faculty of Chemistry, 11000, Belgrade, Serbia
| | - Marijana Stojanovic
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", National Institute of the Republic of Serbia, University of Belgrade, 11000, Belgrade, Serbia
| | - Dragana Stanic-Vucinic
- Centre of Excellence for Molecular Food Sciences and Department of Biochemistry, University of Belgrade, Faculty of Chemistry, 11000, Belgrade, Serbia
| | - Tanja Cirkovic Velickovic
- Centre of Excellence for Molecular Food Sciences and Department of Biochemistry, University of Belgrade, Faculty of Chemistry, 11000, Belgrade, Serbia; Serbian Academy of Sciences and Arts, 11000, Belgrade, Serbia.
| |
Collapse
|
4
|
Wijerathne SVT, Pandit R, Ezeuko CC, Matthews QL. Comparative Examination of Feline Coronavirus and Canine Coronavirus Effects on Extracellular Vesicles Acquired from A-72 Canine Fibrosarcoma Cell Line. Vet Sci 2025; 12:477. [PMID: 40431570 PMCID: PMC12115506 DOI: 10.3390/vetsci12050477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 05/02/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Introduction: Coronavirus (CoV) is an extremely contagious, enveloped positive-single-stranded RNA virus, which has become a global pandemic that causes several illnesses in humans and animals. Hence, it is necessary to investigate viral-induced reactions across diverse hosts. Herein, we propose utilizing naturally secreted extracellular vesicles (EVs), mainly focusing on exosomes to examine virus-host responses following CoV infection. Exosomes are small membrane-bound vesicles originating from the endosomal pathway, which play a pivotal role in intracellular communication and physiological and pathological processes. We suggested that CoV could impact EV formation, content, and diverse immune responses in vitro. Methods: In this study, we infected A-72, which is a canine fibroblast cell line, with a feline coronavirus (FCoV) and canine coronavirus (CCoV) independently in an exosome-free media at 0.001 multiplicity of infection (MOI), with incubation periods of 48 and 72 h. The cell viability was significantly downregulated with increased incubation time following FCoV and CCoV infection, which was identified by performing the 3-(4,5-dimethylthiazo-1-2yl)-2,5-diphenyltetrazolium bromide (MTT) assay. After the infection, EVs were isolated through ultracentrifugation, and the subsequent analysis involved quantifying and characterizing the purified EVs using various techniques. Results: NanoSight particle tracking analysis (NTA) verified that EV dimensions fell between 100 and 200 nm at both incubation periods. At both periods, total protein and RNA levels were significantly upregulated in A-72-derived EVs following FCoV and CCoV infections. However, total DNA levels were gradually upregulated with increased incubation time. Dot blot analysis indicated that the expression levels of ACE2, IL-1β, Flotillin-1, CD63, caspase-8, and Hsp90 were modified in A-72-derived EVs following both CoV infections. Conclusions: Our results indicated that FCoV and CCoV infections could modulate the EV production and content, which could play a role in the development of viral diseases. Investigating diverse animal CoV will provide in-depth insight into host exosome biology during CoV infection. Hence, our findings contribute to the comprehension and characterization of EVs in virus-host interactions during CoV infection.
Collapse
Affiliation(s)
- Sandani V. T. Wijerathne
- Microbiology Program, Alabama State University, Montgomery, AL 36104, USA; (S.V.T.W.); (R.P.); (C.C.E.)
| | - Rachana Pandit
- Microbiology Program, Alabama State University, Montgomery, AL 36104, USA; (S.V.T.W.); (R.P.); (C.C.E.)
| | - Chioma C. Ezeuko
- Microbiology Program, Alabama State University, Montgomery, AL 36104, USA; (S.V.T.W.); (R.P.); (C.C.E.)
| | - Qiana L. Matthews
- Microbiology Program, Alabama State University, Montgomery, AL 36104, USA; (S.V.T.W.); (R.P.); (C.C.E.)
- Department of Biological Sciences, College of Science, Technology, Engineering, and Mathematics, Alabama State University, Montgomery, AL 36104, USA
| |
Collapse
|
5
|
Wang L, Tučs A, Ding S, Tsuda K, Sljoka A. HDXRank: A Deep Learning Framework for Ranking Protein Complex Predictions with Hydrogen-Deuterium Exchange Data. J Chem Theory Comput 2025. [PMID: 40367339 DOI: 10.1021/acs.jctc.5c00175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Accurate modeling of protein-protein complex structures is essential for understanding biological mechanisms. Hydrogen-deuterium exchange (HDX) experiments provide valuable insights into binding interfaces. Incorporating HDX data into protein complex modeling workflows offers a promising approach to improve prediction accuracy. Here, we developed HDXRank, a graph neural network (GNN)-based framework for candidate structure ranking utilizing alignment with HDX experimental data. Trained on a newly curated HDX data set, HDXRank captures nuanced local structural features critical for accurate HDX profile prediction. This versatile framework can be integrated with a variety of protein complex modeling tools, transforming the HDX profile alignment into a model quality metric. HDXRank demonstrates effectiveness at ranking models generated by rigid docking or AlphaFold, successfully prioritizing functionally relevant models and improving prediction quality across all tested protein targets. These findings underscore HDXRank's potential to become a pivotal tool for understanding molecular recognition in complex biological systems.
Collapse
Affiliation(s)
- Liyao Wang
- Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa 277-8561, Japan
- RIKEN Center for Advanced Intelligence Project, RIKEN, Tokyo 103-0027, Japan
| | - Andrejs Tučs
- Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa 277-8561, Japan
- RIKEN Center for Advanced Intelligence Project, RIKEN, Tokyo 103-0027, Japan
| | - Songting Ding
- Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa 277-8561, Japan
| | - Koji Tsuda
- Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa 277-8561, Japan
- RIKEN Center for Advanced Intelligence Project, RIKEN, Tokyo 103-0027, Japan
| | - Adnan Sljoka
- RIKEN Center for Advanced Intelligence Project, RIKEN, Tokyo 103-0027, Japan
| |
Collapse
|
6
|
Bermúdez-Puga S, Mendes B, Ramos-Galarza JP, Oliveira de Souza de Azevedo P, Converti A, Molinari F, Moore SJ, Almeida JR, Pinheiro de Souza Oliveira R. Revolutionizing agroindustry: Towards the industrial application of antimicrobial peptides against pathogens and pests. Biotechnol Adv 2025; 82:108605. [PMID: 40368115 DOI: 10.1016/j.biotechadv.2025.108605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 04/09/2025] [Accepted: 05/10/2025] [Indexed: 05/16/2025]
Abstract
Antibiotics are essential chemicals for medicine and agritech. However, all antibiotics are small molecules that pathogens evolve antimicrobial resistance (AMR). Alternatively, antimicrobial peptides (AMPs) offer potential to overcome or evade AMR. AMPs provide broad-spectrum activity, favourable biosafety profiles, and a rapid and efficient mechanism of action with low resistance incidence. These properties have driven innovative applications, positioning AMPs as promising contributors to advancements in various industrial sectors. This review evaluates the multifaceted nature of AMPs and their biotechnological applications in underexplored sectors. In the food industry, the application of AMPs helps to suppress the growth of microorganisms, thereby decreasing foodborne illnesses, minimizing food waste, and prolonging the shelf life of products. In animal husbandry and aquaculture, incorporating AMPs into the diet reduces the load of pathogenic microorganisms and enhances growth performance and survival rates. In agriculture, AMPs provide an alternative to decrease the use of chemical pesticides and antibiotics. We also review current methods for obtaining AMPs, including chemical synthesis, recombinant DNA technology, cell-free protein synthesis, and molecular farming, are also reviewed. Finally, we look to the peptide market to assess its status, progress, and transition from the discovery stage to benefits for society and high-quality products. Overall, our review exemplifies the other side of the coin of AMPs and how these molecules provide similar benefits to conventional antibiotics and pesticides in the agritech sector.
Collapse
Affiliation(s)
- Sebastián Bermúdez-Puga
- Microbial Biomolecules Laboratory, Faculty of Pharmaceutical Sciences, University of São Paulo, Rua do Lago 250, Cidade Universitária, São Paulo 05508-000, SP, Brazil
| | - Bruno Mendes
- School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6AH, UK
| | - Jean Pierre Ramos-Galarza
- Biomolecules Discovery Group, Universidad Regional Amazónica Ikiam, Km 7 Via Muyuna, Tena, Napo, Ecuador
| | - Pamela Oliveira de Souza de Azevedo
- Microbial Biomolecules Laboratory, Faculty of Pharmaceutical Sciences, University of São Paulo, Rua do Lago 250, Cidade Universitária, São Paulo 05508-000, SP, Brazil
| | - Attilio Converti
- Department of Civil, Chemical and Environmental Engineering, Pole of Chemical Engineering, University of Genoa, Via Opera Pia 15, 16145 Genoa, Italy
| | - Francesco Molinari
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Simon J Moore
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - José R Almeida
- Biomolecules Discovery Group, Universidad Regional Amazónica Ikiam, Km 7 Via Muyuna, Tena, Napo, Ecuador; School of Pharmacy, University of Reading, Reading RG6 6UB, UK
| | - Ricardo Pinheiro de Souza Oliveira
- Microbial Biomolecules Laboratory, Faculty of Pharmaceutical Sciences, University of São Paulo, Rua do Lago 250, Cidade Universitária, São Paulo 05508-000, SP, Brazil.
| |
Collapse
|
7
|
Costa SM, Simas MCDC, da Costa LJ, Silva R. Dynamics of SARS-CoV-2 Mutations in Wastewater Provide Insights into the Circulation of Virus Variants in the Population. Int J Mol Sci 2025; 26:4324. [PMID: 40362561 PMCID: PMC12072199 DOI: 10.3390/ijms26094324] [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: 03/14/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
SARS-CoV-2 high transmission and genomic mutations result in the emergence of new variants that impact COVID-19 vaccine efficacy and virus transmission by evading the host immune system. Wastewater-based epidemiology is an effective approach to monitor SARS-CoV-2 variants circulation in the population but is a challenge due to the presence of reaction inhibitors and the low concentrations of SARS-CoV-2 in this environment. Here, we aim to improve SARS-CoV-2 variant detection in wastewater by employing nested PCR followed by next-generation sequencing (NGS) of small amplicons of the S gene. Eight SARS-CoV-2 wastewater samples from Alegria Wastewater Treatment Plant, in Rio de Janeiro, Brazil, were collected monthly from February to September 2021. Samples were submitted to virus concentration, RNA extraction and nested PCR followed by NGS. The small amplicons were used to prepare libraries for sequencing without the need to perform any fragmentation step. We identified and calculated the frequencies of 29 mutations matching the Alpha, Beta, Gamma, Delta, Omicron, and P.2 variants. Omicron matching-mutations were detected before the lineage was classified as a variant of concern. SARS-CoV-2 wastewater sequences clustered with SARS-CoV-2 variants detected in clinical samples that circulated in 2021 in Rio de Janeiro. We show that sequencing of selected small amplicons of SARS-CoV-2 S gene allows the identification of SARS-CoV-2 variants matching mutations and their frequencies' calculation. This approach may be expanded using customizing primers for additional genomic regions, in order to differentiate current variants. Approaches that allow us to learn how variants emerge and how they relate to clinical outcomes are crucial for our understanding of the dynamics of virus variants circulation, providing valuable data for public health management.
Collapse
Affiliation(s)
- Sara Mesquita Costa
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (S.M.C.); (M.C.d.C.S.)
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
| | - Maria Clara da Costa Simas
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (S.M.C.); (M.C.d.C.S.)
| | - Luciana Jesus da Costa
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
| | - Rosane Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (S.M.C.); (M.C.d.C.S.)
| |
Collapse
|
8
|
Ali FH, Gentilcore G, Al-Jighefee HT, Taleb SA, Hssain AA, Qotba HA, Al Thani AA, Abu Raddad LJ, Nasrallah GK, Grivel JC, Yassine HM. Comprehensive analysis of human coronavirus antibody responses in ICU and non-ICU COVID-19 patients reveals IgG3 against SARS-CoV-2 spike protein as a key biomarker of disease severity. J Med Microbiol 2025; 74:002012. [PMID: 40359129 PMCID: PMC12075857 DOI: 10.1099/jmm.0.002012] [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: 07/08/2024] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction. Pre-existing immunity to human coronaviruses (HCoVs) may shape the immune response in COVID-19 patients. Increasing evidence suggests that immune cross-reactivity between SARS-CoV-2 and other coronaviruses may determine clinical prognosis.Hypothesis. SARS-CoV-2 disease severity is influenced by pre-existing immunity to HCoVs, with distinct antibody profiles and cross-reactivity patterns.Aim. To investigate the antibody response of ICU and non-ICU SARS-CoV-2 patients against different HCoV proteins and assess the potential impact of pre-existing immunity on SARS-CoV-2 disease outcomes.Methodology. This study used a comprehensive HCoVs antigen bead array to measure antibody response to pathogenic Middle East respiratory syndrome coronavirus (MERS-CoV), SARS-CoV, SARS-CoV-2 and the four seasonal HCoVs in 70 ICU and 63 non-ICU COVID-19 patients.Results. Our analysis demonstrates an overall higher antibody response in ICU than in non-ICU COVID-19 patients. Interestingly, the anti-S1 IgG and IgA were significantly higher among ICU than in non-ICU patients. Similarly, the anti-S1 IgG against NL63 showed a lower response among ICU compared to non-ICU. Cross-reactivity was evident between SARS-CoV-2 and SARS-CoV antibodies but not with MERS-CoV and seasonal HCoVs. The subclass analysis of antibodies recognizing SARS-CoV-2 revealed that anti-S1 IgG1, IgG3, IgA1 and IgA2 were significantly higher in ICU compared to non-ICU. The predominant IgA subtype among SARS-CoV-2 patients was IgA1. We applied machine learning algorithms to subclass serological responses to build classifiers that could distinguish between ICU patients and patients with milder COVID-19. Out of 90 variables used in two different types of models, the variable of highest influence in determining the ICU status was IgG3 against SARS-CoV-2 S, and the top 8 variables of influence included the presence of IgG3 against S-trimer as well as IgA against SARS-CoV-2 S.Conclusion. Understanding the complexities of humoral immunity in various patients is critical for early medical intervention, disease management, selective vaccination and passive immunotherapy.
Collapse
Affiliation(s)
- Fatma H. Ali
- Biomedical Research Center, QH Health, Qatar University, Doha, Qatar
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | | | | | - Sara Ahmad Taleb
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Ali Ait Hssain
- Medical Intensive Care Unit, Hamad Medical Corporation, Doha, Qatar
| | | | - Asmaa A. Al Thani
- Biomedical Research Center, QH Health, Qatar University, Doha, Qatar
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Laith J. Abu Raddad
- Department of Population Health Sciences, Infectious Disease Epidemiology Group, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Gheyath K. Nasrallah
- Biomedical Research Center, QH Health, Qatar University, Doha, Qatar
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | | | - Hadi M. Yassine
- Biomedical Research Center, QH Health, Qatar University, Doha, Qatar
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
9
|
Zhang E, Luo S, Xu X, Wang Q, Liu J, Gao P, Duan L. Molecular mechanistic exploration of conformational shifts induced by class IV anti-RBD antibody IY2A. Int J Biol Macromol 2025; 306:141417. [PMID: 39993688 DOI: 10.1016/j.ijbiomac.2025.141417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 02/26/2025]
Abstract
The SARS-CoV-2 virus mutates rapidly, reducing the effectiveness of antibodies. The novel Class IV antibody IY2A partially unfolds the receptor-binding domain (RBD), allowing tolerance of antigenic variations and effectively neutralizing Omicron variants. In this study, we used molecular dynamics simulations and alanine scanning combined with interaction entropy method to elucidate how IY2A maintains its binding affinity across Omicron variants. We compared IY2A with EY6A and evaluated how mutations affect IY2A inhibition. The findings revealed that the IY2A adopted a closer conformation when binding to Omicron variants than to the WT. Energy calculations indicate that van der Waals interactions primarily drive IY2A binding to the RBD. Following unfolding, IY2A interacts with the RBD via interatomic hydrogen bonds and hydrophobic contacts involving LEU368, PHE377, LYS378, and SER383. This study provides theoretical insights to guide the development of Class IV antibodies against emerging and future Omicron variants.
Collapse
Affiliation(s)
- Enhao Zhang
- School of Physics and Electronics, Shandong Normal University, Jinan 250014, China
| | - Song Luo
- School of Physics and Electronics, Shandong Normal University, Jinan 250014, China
| | - Xiaole Xu
- School of Physics and Electronics, Shandong Normal University, Jinan 250014, China
| | - Qihang Wang
- School of Physics and Electronics, Shandong Normal University, Jinan 250014, China
| | - Jinxin Liu
- School of Physics and Electronics, Shandong Normal University, Jinan 250014, China
| | - Pengfei Gao
- School of Physics and Electronics, Shandong Normal University, Jinan 250014, China
| | - Lili Duan
- School of Physics and Electronics, Shandong Normal University, Jinan 250014, China.
| |
Collapse
|
10
|
Bamrung P, Toviwek B, Samsudin F, Chairatana P, Bond PJ, Pongprayoon P. The Binding of Brazilin from C. sappan to the Full-Length SARS-CoV-2 Spike Proteins. Int J Mol Sci 2025; 26:4100. [PMID: 40362339 PMCID: PMC12072004 DOI: 10.3390/ijms26094100] [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: 04/01/2025] [Revised: 04/15/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
The emergence of coronavirus disease (COVID-19) caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has become a global issue since 2019. The prominent characteristic of SARS-CoV-2 is the presence of the spike (S) protein protruding from the virus particle envelope. The S protein is a major drug and vaccine target because it initiates the key step in infection. Medicinal herbs are a potential treatment option to enhance immunity to fight viral infections. Caesalpinia sappan L. has been reported to display promising anti-viral activities. Specifically, brazilin (BRA), a major bioactive compound in C. sappan, was reported to play a role in inhibiting viral infection. Thus, the ability of BRA as a COVID-19 treatment was tested. The S protein was used as the BRA target of this work. Understanding the binding mechanism of BRA to the S protein is crucial for future utilisation of C. sappan as a COVID-19 treatment or other coronavirus-caused pandemics. Here, we performed molecular docking of BRA onto the S protein receptor binding domain (RBD) and multimerisation (MM) pockets. Molecular dynamics (MD) simulations were conducted to study the stability of binding to glycosylated and non-glycosylated S protein constructs. BRA can bind to the Receptor-binding motif (RBM) on an RBD surface stably; however, it is too large to fit into the MM pocket, resulting in dissociation. Nonetheless, BRA is bound by residues near the S1/S2 interface. We found that glycosylation has no effect on BRA binding, as the proposed binding site is far from any glycans. Our results thus indicate that C. sappan may act as a promising preventive and therapeutic alternative for COVID-19 treatment.
Collapse
Affiliation(s)
- Phonphiphat Bamrung
- Department of Chemistry, Faculty of Science, Kasetsart University, Chatuchak, Bangkok 10900, Thailand; (P.B.); (B.T.)
| | - Borvornwat Toviwek
- Department of Chemistry, Faculty of Science, Kasetsart University, Chatuchak, Bangkok 10900, Thailand; (P.B.); (B.T.)
| | - Firdaus Samsudin
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore;
| | - Phoom Chairatana
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand;
| | - Peter John Bond
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore;
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Prapasiri Pongprayoon
- Department of Chemistry, Faculty of Science, Kasetsart University, Chatuchak, Bangkok 10900, Thailand; (P.B.); (B.T.)
- Center for Advanced Studies in Nanotechnology for Chemical, Food and Agricultural Industries, KU Institute for Advanced Studies, Kasetsart University, Bangkok 10900, Thailand
| |
Collapse
|
11
|
Robert Kolar M, Mitra D, Kobzarenko V. Efficient discovery of frequently co-occurring mutations in a sequence database with matrix factorization. PLoS Comput Biol 2025; 21:e1012391. [PMID: 40273414 DOI: 10.1371/journal.pcbi.1012391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
We have developed a robust method for efficiently tracking multiple co-occurring mutations in a sequence database. Evolution often hinges on the interaction of several mutations to produce significant phenotypic changes that lead to the proliferation of a variant. However, identifying numerous simultaneous mutations across a vast database of sequences poses a significant computational challenge. Our approach leverages a matrix factorization technique to automatically and efficiently pinpoint subsets of positions where co-mutations occur, appearing in a substantial number of sequences within the database. We validated our method using SARS-CoV-2 receptor-binding domains, comprising approximately seven hundred thousand sequences of the Spike protein, demonstrating superior performance compared to a reasonably exhaustive brute-force method. Furthermore, we explore the biological significance of the identified co-mutational positions (CMPs) and their potential impact on the virus's evolution and functionality, identifying key mutations in Delta and Omicron variants. This analysis underscores the significant role of identified CMPs in understanding the evolutionary trajectory. By tracking the "birth" and "death" of CMPs, we can elucidate the persistence and impact of specific groups of mutations across different viral strains, providing valuable insights into the virus' adaptability and thus, possibly aiding vaccine design strategies.
Collapse
Affiliation(s)
- Michael Robert Kolar
- BiC Lab, Department of Electrical Engineering and Computer Science, Florida Institute of Technology, Melbourne, Florida, United States of America
| | - Debasis Mitra
- BiC Lab, Department of Electrical Engineering and Computer Science, Florida Institute of Technology, Melbourne, Florida, United States of America
| | - Valerie Kobzarenko
- BiC Lab, Department of Electrical Engineering and Computer Science, Florida Institute of Technology, Melbourne, Florida, United States of America
| |
Collapse
|
12
|
Pan Y, Yan J, Zhang Y, Lin J, Liang Z, Sun J. Centrifugation-Based Purification Protocol Optimization Enhances Structural Preservation of Nucleopolyhedrovirus Budded Virion Envelopes. INSECTS 2025; 16:424. [PMID: 40332984 PMCID: PMC12027964 DOI: 10.3390/insects16040424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/04/2025] [Accepted: 04/13/2025] [Indexed: 05/08/2025]
Abstract
The structural integrity of viral envelopes is a critical determinant of infectivity for enveloped viruses, directly influencing vector stability, functional accuracy of surface-displayed epitopes, and preservation of native conformational states required for membrane protein studies. However, conventional purification methods often disrupt envelope integrity and cause envelope proteins to lose their activity. Here, we systematically compared discontinuous, continuous, and optimized continuous sucrose density gradient centrifugation protocols for purifying Autographa californica multiple nucleopolyhedrovirus (AcMNPV). Through cryo-EM, we demonstrated that our optimized continuous sucrose gradient protocol significantly increased the proportion of AcMNPV budded virions with intact envelopes from 36% to 81%, while preserving the metastable prefusion conformation of the fusion protein GP64. This advancement should prove useful for structural studies of viral envelope proteins and may enhance applications in gene therapy and vaccine development utilizing enveloped viruses.
Collapse
Affiliation(s)
- Yong Pan
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding & Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (Y.P.); (J.Y.); (Y.Z.); (J.L.); (Z.L.)
| | - Jiming Yan
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding & Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (Y.P.); (J.Y.); (Y.Z.); (J.L.); (Z.L.)
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yinong Zhang
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding & Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (Y.P.); (J.Y.); (Y.Z.); (J.L.); (Z.L.)
| | - Jiasheng Lin
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding & Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (Y.P.); (J.Y.); (Y.Z.); (J.L.); (Z.L.)
| | - Zhiquan Liang
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding & Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (Y.P.); (J.Y.); (Y.Z.); (J.L.); (Z.L.)
| | - Jingchen Sun
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding & Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (Y.P.); (J.Y.); (Y.Z.); (J.L.); (Z.L.)
| |
Collapse
|
13
|
Pedrini M, Pozzi L, Sacchi F, Citarella A, Fasano V, Seneci P, Pieraccini S, Ruberto L, Peña HP, Garzino-Demo A, Vitiello A, Sernicola L, Borsetti A, Calistri A, Parolin C, Passarella D. Design, synthesis and in vitro validation of bivalent binders of SARS-CoV-2 spike protein: Obeticholic, betulinic and glycyrrhetinic acids as building blocks. Bioorg Med Chem 2025; 121:118124. [PMID: 39999646 DOI: 10.1016/j.bmc.2025.118124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/12/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025]
Abstract
SARS-CoV-2 is the virus responsible for the COVID-19 pandemic, which caused over 6.7 million deaths worldwide. The Spike protein plays a crucial role in the infection process, mediating the binding of the virus to its cellular receptor, angiotensin-converting enzyme 2 (ACE2), and its subsequent entry into target cells. Previous studies identified, through virtual screening, several natural products capable of binding to two distinct pockets of the Spike protein: triterpenoids binding to pocket 1 and bile acid derivatives binding to pocket 5. Building on these findings, our study advances the field by developing bivalent compounds 1-4 that through a spacer combine a triterpenoid (betulinic acid or glycyrrhetinic acid) with a semisynthetic bile acid derivative (obeticholic acid). These bivalent compounds are designed to simultaneously bind both pockets of the Spike protein, offering significant advantages over single molecules or the combination of the two natural products. In vitro cell assays using pseudotyped recombinant lentiviral particles with selected SARS-CoV-2 Spike proteins demonstrated that 1 and 2 exhibit enhanced activity in reducing viral entry into target cells compared to individual natural products, thus highlighting their potential as superior antiviral agents with reduced side effects.
Collapse
Affiliation(s)
- Martina Pedrini
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Luca Pozzi
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Francesca Sacchi
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Andrea Citarella
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy.
| | - Valerio Fasano
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Pierfausto Seneci
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Stefano Pieraccini
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Lorenzo Ruberto
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Helena Perez Peña
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Alfredo Garzino-Demo
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121 Padova, Italy; Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, United States; Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Adriana Vitiello
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121 Padova, Italy
| | - Leonardo Sernicola
- National HIV/AIDS Research Center (CNAIDS), Istituto Superiore di Sanità, 00162 Roma, Italy
| | - Alessandra Borsetti
- National HIV/AIDS Research Center (CNAIDS), Istituto Superiore di Sanità, 00162 Roma, Italy
| | - Arianna Calistri
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121 Padova, Italy
| | - Cristina Parolin
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121 Padova, Italy
| | - Daniele Passarella
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy.
| |
Collapse
|
14
|
Jana ID, Kanjo K, Roy S, Bhasin M, Bhattacharya S, Banerjee I, Jana S, Chatterjee A, Chakrabarti AK, Chakraborty S, Mukherjee B, Varadarajan R, Mondal A. Early 2022 breakthrough infection sera from India target the conserved cryptic class 5 epitope to counteract immune escape by SARS-CoV-2 variants. J Virol 2025; 99:e0005125. [PMID: 40135898 PMCID: PMC11998512 DOI: 10.1128/jvi.00051-25] [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: 01/11/2025] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
During the coronavirus disease 2019 (COVID-19) pandemic, the vast majority of epitope mapping studies have focused on sera from mRNA-vaccinated populations from high-income countries. In contrast, here, we report an analysis of 164 serum samples isolated from patients with breakthrough infection in India during early 2022 who received two doses of the ChAdOx viral vector vaccine. Sera were screened for neutralization breadth against wild-type (WT), Kappa, Delta, and Omicron BA.1 viruses. Three sera with the highest neutralization breadth and potency were selected for epitope mapping, using charged scanning mutagenesis coupled with yeast surface display and next-generation sequencing. The mapped sera primarily targeted the recently identified class 5 cryptic epitope and, to a lesser extent, the class 1 and class 4 epitopes. The class 5 epitope is completely conserved across all severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants and for most sarbecoviruses. Based on these observations, an additional 26 sera were characterized, and all showed a broad neutralizing activity, including against XBB.1.5. This is in contrast with the results obtained with the sera from individuals receiving multiple doses of original and updated mRNA vaccines, where impaired neutralization of XBB and later variants of concern (VOCs) were observed. Our study demonstrates that two doses of the ChAdOx vaccine in a highly exposed population were sufficient to drive substantial neutralization breadth against emerging and upcoming variants of concern. These data highlight the important role of hybrid immunity in conferring broad protection and inform future vaccine strategies to protect against rapidly mutating viruses. IMPORTANCE Worldwide implementation of coronavirus disease 2019 (COVID-19) vaccines and the parallel emergence of newer severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants have shaped the humoral immune response in a population-specific manner. While characterizing this immune response is important for monitoring disease progression at the population level, it is also imperative for developing effective countermeasures in the form of novel vaccines and therapeutics. India has implemented the world's second largest COVID-19 vaccination drive and encountered a large number of post-vaccination "breakthrough" infections. From a cohort of patients with breakthrough infection, we identified individuals whose sera showed broadly neutralizing immunity against different SARS-CoV-2 variants. Interestingly, these sera primarily target a novel cryptic epitope, which was not identified in previous population-level studies conducted in Western countries. This rare cryptic epitope remains conserved across all SARS-CoV-2 variants, including recently emerged ones and for the SARS-like coronaviruses that may cause future outbreaks, thus representing a potential target for future vaccines.
Collapse
Affiliation(s)
- Indrani Das Jana
- Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Kawkab Kanjo
- Molecular Biophysics Unit (MBU), Indian Institute of Science, Bengaluru, India
| | - Subhanita Roy
- Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Munmun Bhasin
- Molecular Biophysics Unit (MBU), Indian Institute of Science, Bengaluru, India
| | - Shatarupa Bhattacharya
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Indranath Banerjee
- B.C. Roy Technology Hospital, Indian Institute of Technology Kharagpur, Kharagpur, India
| | | | | | - Alok Kumar Chakrabarti
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Suman Chakraborty
- Department of Mechanical Engineering, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Budhaditya Mukherjee
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | | | - Arindam Mondal
- Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
15
|
Jerome JR, Wilson KL, Fialho J, Goodchild G, Prakash MD, McLeod C, Richmond PC, Apostolopoulos V, Flanagan KL, Plebanski M. Optimisation of the cultured ELISpot/Fluorospot technique for the selective investigation of SARS-CoV-2 reactive central memory T cells. Front Immunol 2025; 16:1547220. [PMID: 40303392 PMCID: PMC12037488 DOI: 10.3389/fimmu.2025.1547220] [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: 12/17/2024] [Accepted: 03/17/2025] [Indexed: 05/02/2025] Open
Abstract
Introduction This study presents an optimised cultured ELISpot protocol for detecting central memory T-cell interferon gamma (IFNγ) responses against SARS-CoV-2 peptides following an initial priming with either peptides, or whole spike protein. Methods Key variations optimised include the culture length, timing of exogenous survival signals (IL-2), and endpoint analysis modality and cell density to enhance assay sensitivity without compromising specificity for central memory T-cell IFNγ recall responses to cognate antigen. Results We noted a culture duration of 10 days, combined with a delayed IL-2 administration on day 5 to enhance assay sensitivity while maintaining response specificity towards cognate antigen when compared with shorter culture periods or earlier exogenous survival signal provision. With regards to lower-frequency T-cell interactions, as we observed with our donor SARS-CoV-2 epitope responses, our findings suggest Fluorospot to be preferable to the chromogenic ELISpot modality, and an immediate cell washing after culture collection to better facilitate cognate antigen responses. Fluorospot enabled a higher cell density while minimising the generation of visual artefacts, meanwhile immediate cell washing was critical for improving endpoint assay sensitivity. CCR7+ cell depletion was used to demonstrate our optimised protocol to selectively demonstrate central memory T-cell responses. Lastly, we provide evidence for the capacity of our assay to delineate individual responding peptides following peptide pool priming, and to explore cross-reactivity between viral variant peptides. Conclusion This work advances the methodology for investigating T-cell immunity, particularly in the context of SARS-CoV-2, and emphasises the balance between enhancing specific cognate central memory responses while limiting non-specific activation.
Collapse
Affiliation(s)
- Jack R. Jerome
- School of Health and Biomedical Science, Royal Melbourne Institute of Technology University, Melbourne, VIC, Australia
- Accelerator for Translational Research in Clinical Trials (ATRACT) Centre, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University, Melbourne, VIC, Australia
| | - Kirsty L. Wilson
- School of Health and Biomedical Science, Royal Melbourne Institute of Technology University, Melbourne, VIC, Australia
- Accelerator for Translational Research in Clinical Trials (ATRACT) Centre, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University, Melbourne, VIC, Australia
| | - Joshuah Fialho
- School of Health and Biomedical Science, Royal Melbourne Institute of Technology University, Melbourne, VIC, Australia
- Accelerator for Translational Research in Clinical Trials (ATRACT) Centre, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University, Melbourne, VIC, Australia
| | - Georgia Goodchild
- School of Health and Biomedical Science, Royal Melbourne Institute of Technology University, Melbourne, VIC, Australia
- Accelerator for Translational Research in Clinical Trials (ATRACT) Centre, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University, Melbourne, VIC, Australia
| | - Monica D. Prakash
- School of Health and Biomedical Science, Royal Melbourne Institute of Technology University, Melbourne, VIC, Australia
- Accelerator for Translational Research in Clinical Trials (ATRACT) Centre, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University, Melbourne, VIC, Australia
| | - Charlie McLeod
- Wesfarmers Centre of Vaccines and Infectious Diseases, Kids Research Institute of Australia, Perth, WA, Australia
- Centre for Child Health Research, University of Western Australia, Perth, WA, Australia
- Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Infectious Diseases Department, Perth Children’s Hospital, Perth, WA, Australia
| | - Peter C. Richmond
- Wesfarmers Centre of Vaccines and Infectious Diseases, Kids Research Institute of Australia, Perth, WA, Australia
- Infectious Diseases Department, Perth Children’s Hospital, Perth, WA, Australia
- Division of Paediatrics, University of Western Australia School of Medicine, Perth, WA, Australia
- Department of Immunology, Perth Children’s Hospital, Perth, WA, Australia
| | - Vasso Apostolopoulos
- School of Health and Biomedical Science, Royal Melbourne Institute of Technology University, Melbourne, VIC, Australia
| | - Katie L. Flanagan
- School of Health and Biomedical Science, Royal Melbourne Institute of Technology University, Melbourne, VIC, Australia
- Tasmanian Vaccine Trial Centre, Clifford Craig Foundation, Launceston General Hospital, Launceston, TAS, Australia
- School of Health Sciences and School of Medicine, University of Tasmania, Launceston, TAS, Australia
| | - Magdalena Plebanski
- School of Health and Biomedical Science, Royal Melbourne Institute of Technology University, Melbourne, VIC, Australia
- Accelerator for Translational Research in Clinical Trials (ATRACT) Centre, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University, Melbourne, VIC, Australia
| |
Collapse
|
16
|
Liu Y, Li C, Wu Z, Zhao Y, Yin T, Liu X, Hui J, Wang Q, Pan Y, Shan Y, Qu X. Self-assembled epitope-based nanoparticles targeting the SARS-CoV-2 spike protein enhanced the immune response and induced potential broad neutralizing activity. Front Cell Infect Microbiol 2025; 15:1560330. [PMID: 40270771 PMCID: PMC12014594 DOI: 10.3389/fcimb.2025.1560330] [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: 01/14/2025] [Accepted: 03/20/2025] [Indexed: 04/25/2025] Open
Abstract
Introduction The ongoing COVID-19 has caused a global pandemic, resulting in millions of infections and deaths. While current vaccines target the SARS-CoV-2 spike (S) protein, its high mutation rate significantly compromises vaccine efficacy. We aimed to evaluate the potential of epitope-based nanoparticles (NPs) to induce broad cross-protection and durable immune responses against SARS-CoV-2. Methods Four conserved epitopes derived from the receptor-binding domain (RBD) and S2 subunit of the spike protein were integrated into Helicobacter pylori ferritin to create epitope-based NPs named S18-F, RBM-F, UH-F, and HR2-F. The immunogenicity of the epitope-based NPs was evaluated through animal experiments to measure epitope-specific antibody titers and assess neutralizing activity against SARS-CoV-2 pseudovirus. To characterize cellular immune responses, splenic lymphocyte proliferation following epitope stimulation was measured, and cytokine secretion profiles including IFN-γ, IL-2, IL-4, and IL-10 were analyzed to determine Th1/Th2 immune polarization. Antibody-dependent cellular cytotoxicity (ADCC) assays were performed to evaluate NP-enhanced recognition and elimination of infected target cells. Results These NPs induced high titers of epitope-specific antibodies lasting three months post-immunization. Sera from the RBM-F, UH-F, and HR2-F groups exhibited neutralizing activity against the SARS-CoV-2 pseudovirus WH-1 in vitro. Splenic lymphocytes from the S18-F, RBM-F, and UH-F groups showed significantly increased proliferation. Lymphocytes from the RBM-F group demonstrated increased secretion of IFN-γ, IL-2, IL-4, and IL-10 cytokines, indicating a balanced Th1 and Th2 immune response. Immune sera from the S18-F and mixed-immunized groups exhibited antibody-dependent cellular cytotoxicity. Discussion The results indicate that these NPs induce robust humoral and cellular immune responses, potentially offering a promising strategy for effective vaccine development against SARS-CoV-2.
Collapse
Affiliation(s)
- Yue Liu
- Department of Echocardiography, The First Hospital of Jilin University, Changchun, China
- Institute of Virology and AIDS Research, The First Hospital of Jilin University, Changchun, China
| | - Chenxi Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Zirui Wu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Yu Zhao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Tieyan Yin
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Xiaopan Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Jiaru Hui
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Qingyu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Yi Pan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Yaming Shan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Xinglong Qu
- Department of Respiratory, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
17
|
Ilyas S, Manan A, Lee D. Deep Learning-Based Comparative Prediction and Functional Analysis of Intrinsically Disordered Regions in SARS-CoV-2. Int J Mol Sci 2025; 26:3411. [PMID: 40244295 PMCID: PMC11989790 DOI: 10.3390/ijms26073411] [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: 02/10/2025] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
This study explores the role of intrinsically disordered regions (IDRs) in the SARS-CoV-2 proteome and their potential as targets for small-molecule drug discovery. Experimentally validated intrinsic disordered regions from the literature were utilized to assess the prediction of intrinsic disorder across a selection of SARS-CoV-2 proteins. The disorder propensities of proteins using four deep learning-based disorder prediction models: ADOPT, PONDR®VLXT, PONDR®VSL2, and flDPnn, were analyzed. ADOPT, VSL2, and VLXT identified a flexible linker (129-147), while VSL2 and VLXT predicted disorder in the Cu(II) binding region (163-167) of NSP1. ADOPT did not predict disordered regions in NSP11; however, VSL2 and VLXT identified disorder in the experimentally validated regions. The IDR in ORF3a is crucial for protein localization and immune modulation, affecting inflammatory pathways. VSL2 predicted significant disorder in the N-terminal domain (18-23), which aligns with experimental data (1-41), overlapping with the TRAF-binding motif, while ADOPT indicated high disorder in the C-terminal domain (255-275), consistent with VSL2 and flDPnn. All tools identified disorder in the N-terminal (1-68), central linker (181-248), and C-terminal (370-419) regions of the nucleocapsid (N) protein, suggesting flexibility and accuracy. The S2 subunit of the spike protein displayed more predicted disorder than the S1 subunit across ADOPT, VSL2, and flDPnn. These IDRs are essential for viral functions, like protein localization, immune modulation, receptor binding, and membrane fusion. This study highlights the importance of IDR in modulating key inflammatory pathways, suggesting that they could serve as promising targets for small-molecule drug development to combat COVID-19.
Collapse
Affiliation(s)
- Sidra Ilyas
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea
| | - Abdul Manan
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea;
| | - Donghun Lee
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea
| |
Collapse
|
18
|
Fu Z, Lin S, Chen H, Guo H, Li J, Chen Y, Lu Y, Liu J, Huang W, Pang Y. Generating Self-Adjuvated Nanofiber Vaccines by Coating Bacterial Flagella with Antigens. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2415887. [PMID: 39981905 DOI: 10.1002/adma.202415887] [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: 10/17/2024] [Revised: 01/28/2025] [Indexed: 02/22/2025]
Abstract
Bacteria-based vaccines have received increasing attention given the ability to induce strong systemic immune responses. However, the application of bacteria as therapeutic agents inevitably suffers from infection-associated side effects due to the living characteristics. Here, the use of bacteria-derived flagella is described to construct self-adjuvated nanofiber vaccines. With the help of charge-reversal mediated by decoration with cationic polymers, the flagella can be coated with negatively charged antigens through electrostatic interaction. By virtue of the large aspect ratio, the resulting nanofiber vaccines show prolonged retention at the injection site and increased uptake by dendritic cells and macrophages. Thanks to the innate immunogenicity, self-adjuvated flagella robustly promote dendritic cell maturation and macrophage polarization, resulting in the elicitation of antigen-specific T-cell and B-cell immune responses. In ovalbumin-overexpressing melanoma-bearing mice, immunization with ovalbumin-carried vaccines not only exhibits a favorable tolerance, but also displays superior inhibition efficacies on tumor growth and metastasis separately under the therapeutic and prophylactic settings. The flexibility of this approach is further demonstrated for vaccine fabrication by coating with the SARS-CoV-2 Spike protein S1 subunit. Bacterial flagella-based self-adjuvated nanofiber platform proposes a versatile strategy to develop various vaccines for disease prevention and treatment.
Collapse
Affiliation(s)
- Zhenzhen Fu
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Sisi Lin
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Huan Chen
- Shanghai Frontiers Science Center of Drug Target ldentification and Delivery,School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Haiyan Guo
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Juanjuan Li
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yanmei Chen
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yue Lu
- Shanghai Frontiers Science Center of Drug Target ldentification and Delivery,School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Wei Huang
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Xiangfu Laboratory, Jiaxing, Zhejiang, 314102, China
| | - Yan Pang
- Shanghai Frontiers Science Center of Drug Target ldentification and Delivery,School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
19
|
Çelik G, Karaoğlu ŞA, Suyabatmaz Ş, Bozdeveci A, Yılmaz GT, Yaylı N, Akpınar R, Çiçek AÇ. Synthesis, biological evaluation and molecular docking studies of flavonol-3-O-β-D-glycoside as a potential inhibitor of SARS-CoV-2 main protease (3CLpro) in drug development for COVID-19. Int J Biol Macromol 2025; 298:139621. [PMID: 39818399 DOI: 10.1016/j.ijbiomac.2025.139621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 01/18/2025]
Abstract
The COVID-19 pandemic began in March 2020 and has affected many countries and infected over a million people. It has had a serious impact on people's physical and mental health, daily life and the global economy. Today, many drugs show limited efficacy in the treatment of COVID-19 and studies to develop effective drugs continue. Here, we aim to the synthesise and characterise of the flavonol-3-O-glycoside derivatives, the following and evaluated molecular docking studies with antimicrobial activity, inhibition of SARS-CoV-2 main protease enzyme (3CLpro) and nuclease activity. Molecular docking simulations of the synthesized flavonol-3-O-glycoside derivatives, especially compounds 5a, 5d, 5h, 5i and 5m, showed a stronger interaction with SARS-CoV-2 3CLpro in the active site. Two compounds from the target compounds, 5h and 5m, were found to be specifically effective against M. smegmatis and yeasts. In particular, compounds 5a, 5d, 5h, 5i and 5m, which exhibited high activity against the SARS-CoV-2 main protease enzyme, were found to be effective at low concentrations. We determined the IC50 values for the compounds that showed an inhibitory effect as well as their nuclease activities, which further emphasising the potential of our results. Among these, compound 5d showed a significant competitive inhibitor of 3CLpro. Furthermore, nuclease activity studies identified compound 5d as the most potent. The above results suggest that the flavonol-3-O-glycoside derivatives could be promising new antiviral agents for the development of 3CLpro inhibitors to combat COVID-19.
Collapse
Affiliation(s)
- Gonca Çelik
- Department of Chemistry, Faculty of Science, Karadeniz Technical University, Trabzon 61080, Türkiye.
| | - Şengül Alpay Karaoğlu
- Department of Biology, Faculty of Science, Recep Tayyip Erdoğan University, Rize 53100, Türkiye
| | - Şeyma Suyabatmaz
- Department of Biology, Faculty of Science, Recep Tayyip Erdoğan University, Rize 53100, Türkiye
| | - Arif Bozdeveci
- Department of Biology, Faculty of Science, Recep Tayyip Erdoğan University, Rize 53100, Türkiye
| | - Gizem Tatar Yılmaz
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Karadeniz Technical University, Trabzon 61080, Türkiye; Karadeniz Technical University, Institute of Health Sciences, Department of Bioinformatics, 61080 Trabzon, Türkiye; Yılmaz Bilişim R&D Consulting Software Engineering and Services Trade Limited Company, 61081 Trabzon, Türkiye
| | - Nurettin Yaylı
- Department of Pharmacognosy, Faculty of Pharmacy, Karadeniz Technical University, Trabzon 61080, Türkiye
| | - Rahşan Akpınar
- Laboratory of Bee Diseases, Samsun Veterinary Control Institute, Samsun 55200, Türkiye
| | - Ayşegül Çopur Çiçek
- Department of Basic Medical Sciences, Faculty of Medicine, Istanbul Medipol University, Istanbul 34815, Türkiye
| |
Collapse
|
20
|
Sahin F, Atasoy BT, Yalcin S, Bitirim VC. Membrane-targeted immunogenic compositions using exosome mimetic approach for vaccine development against SARS-CoV-2 and other pathogens. Sci Rep 2025; 15:10899. [PMID: 40157987 PMCID: PMC11954949 DOI: 10.1038/s41598-025-95503-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025] Open
Abstract
The COVID-19 pandemic has underscored the urgent need for a vaccine strategy that is safe, effective, rapid, cost-efficient, and scalable for large-scale deployment during widespread infectious outbreaks. Here, we present a new vaccination strategy that meets these critical requirements. The SARS-CoV-2 S protein consists of the S1 and S2 subunits. The S2 subunit acts as the viral cell membrane fusion protein, and mutations in its C-terminal region facilitate the transport of the entire S protein to the cell membrane. When we expressed the SARS-CoV-2 S protein with a deletion of 21 amino acids from its C-terminal region in various cell types, we observed a dense presence of the protein in the cell membrane, as determined by IHC, dot blot, and ELISA. In the cell membrane-SARS-CoV-2 S protein complex, the cell membrane functions as an exosome mimic, carrying protein antigens (S protein) in their most natural form, as no further protocols are used to attach antigens to the membrane. We demonstrate that using the membrane-S protein component as a vaccine yields a more robust and protective immune response, with enhanced safety compared to mRNA-based or inactivated virus-based vaccines against SARS-CoV-2. Additionally, we show that fusing the transmembrane domain of the Vesicular Stomatitis Virus (VSV) G protein with the SARS-CoV-2 S1 protein effectively transports the S1 protein to the cell membrane, similar to SARS-CoV-2 S Δ21. We propose that designing the S2 subunit of the SARS-CoV-2 S protein, or its analogues such as the VSV-G protein, as carriers for fusing bacterial, viral, or tumor proteins with antigenic properties-and transporting them to the cell membrane-could result in a comprehensive vaccination protocol applicable to all bacteria, viruses, and even tumors.
Collapse
Affiliation(s)
- Fikret Sahin
- Department of Medical Microbiology, University of Ankara, Ankara, 06100, Turkey.
- Faculty of Medicine, Department of Medical Microbiology, Ankara University, Morphology Building, Sihhiye, Ankara, 06100, Turkey.
| | - Buse Turegun Atasoy
- Department of Medical Microbiology, University of Ankara, Ankara, 06100, Turkey
| | - Suleyman Yalcin
- Microbiology Reference Laboratory, General Directorate of Public Health, Ankara, 06800, Turkey
| | | |
Collapse
|
21
|
Sánchez A, García-Pardo G, Martí A, Gómez-Bertomeu F, Chafino S, Massanella M, Flores-Piñas M, Cedó L, Vidal F, Peraire J, Rull A. Omics for searching plasma biomarkers associated with unfavorable COVID-19 progression in hypertensive patients. Sci Rep 2025; 15:10343. [PMID: 40133696 PMCID: PMC11937446 DOI: 10.1038/s41598-025-94725-4] [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: 09/25/2024] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
Hypertension is one of the most common risk factors for COVID-19 clinical progression. The identification of plasma biomarkers for anticipating worse clinical outcomes and to better understand the shared mechanisms between hypertension and COVID-19 are needed. A hypothesis-generating study was designed to compare plasma proteomics and metabolomics between 22 hypertensives (HT) and 41 non-hypertensives (nHT) patients with the most unfavorable COVID-19 progression. A total of 43 molecules were significantly differed between HT (n = 22) and nHT (n = 41). Random Forest (RF) analysis identified myo-inositol, gelsolin and phosphatidylcholine (PC) 32:1 as the top molecules for distinguishing between HT and nHT. Plasma myo-inositol and gelsolin were higher (P = 0.03 and P = 0.02, respectively) and plasma PC 32:1 was lower (P = 0.03) in HT compared to nHT. Biological processes like stress response and blood coagulation, along with KEGG pathways including ascorbate and aldarate metabolism (P = 0.021) and linoleic acid metabolism (P = 0.028), were altered in hypertensive patients with the most unfavorable COVID-19 progression. There is a clear link between hypertension and severe COVID-19. Key biological pathways to consider for improving the prognosis and quality of life of hypertensive patients who become infected with SARS-CoV-2 include oxidative stress, ascorbate and aldarate metabolism, lipid metabolism, immune system and inflammation.
Collapse
Affiliation(s)
- Alba Sánchez
- Infection and Immunity (INIM), Institut Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Graciano García-Pardo
- Infection and Immunity (INIM), Institut Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Anna Martí
- Infection and Immunity (INIM), Institut Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
| | - Frederic Gómez-Bertomeu
- Infection and Immunity (INIM), Institut Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Silvia Chafino
- Infection and Immunity (INIM), Institut Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Massanella
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- IrsiCaixa, Hospital Universitari Germans Trias i Pujol, 08916, Badalona, Spain
| | - Marina Flores-Piñas
- Infection and Immunity (INIM), Institut Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
| | - Lídia Cedó
- Grup de Recerca en Diabetis i Malalties Metabòliques Associades (DIAMET), Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari Joan XXIII, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Francesc Vidal
- Infection and Immunity (INIM), Institut Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Joaquim Peraire
- Infection and Immunity (INIM), Institut Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.
- Universitat Rovira i Virgili (URV), Tarragona, Spain.
| | - Anna Rull
- Infection and Immunity (INIM), Institut Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.
- Universitat Rovira i Virgili (URV), Tarragona, Spain.
| |
Collapse
|
22
|
Fang JL, Shrestha L, Beland FA. Flow cytometric analysis of the SARS coronavirus 2 antibodies in human plasma. Sci Rep 2025; 15:10300. [PMID: 40133428 PMCID: PMC11937374 DOI: 10.1038/s41598-025-92389-8] [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: 10/09/2024] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
COVID-19 is an infectious disease caused by the severe acute respiratory syndrome coronavirus (SARS-CoV-2). Anti-SARS-CoV-2 antibodies can provide information on patient immunity, identify asymptomatic patients, and track the spread of COVID-19. Efforts have been made to develop methods to detect anti-SARS-CoV-2 antibodies in humans. Here, we describe a flow cytometric assay for the simultaneous detection of anti-SARS-CoV-2 IgG and IgM in human plasma. To assess the antibody response against the different SARS-CoV-2 structural proteins, five viral recombinant proteins, including spike protein subunit 1 (S1), N-terminal domain of S1 (S1A), spike receptor-binding domain (RBD), spike protein subunit 2 (S2), and nucleocapsid protein (N), were generated. A comparison of the antibody profiles detected by the assay with plasma from 100 healthy blood donors collected prior to the COVID-19 pandemic and plasma from 100 virologically confirmed COVID-19 patients demonstrated a clear discrimination between the two groups. Among the COVID-19 patients, the antibody responses for the viral proteins, as determined by their prevalence, were anti-RBD IgG = anti-N IgG > anti-S1 IgG > anti-S1A IgG > anti-S2 IgG, and anti-RBD IgM > anti-S1 IgM > anti-N IgM > anti-S2 IgM. The prevalence of anti-SARS-CoV-2 IgG and IgM was not associated with sex, age, race, days after the onset of symptoms, or severity of illness, except for a higher prevalence of anti-S2 IgG being observed in men than in women. The levels of anti-RBD IgG were higher in patients 65 years and older and in patients who had severe symptoms. Similarly, patients who had severe symptoms exhibited higher levels of anti-S1 and anti-S1A IgG than patients who had mild or moderate symptoms. The levels of anti-RBD IgM tended to be higher in men but did not differ among age, race, days after the onset of symptoms, or severity of illness. Our study indicates that the flow cytometric assay, especially using RBD as target antigen, can be used to detect simultaneously anti-SARS-CoV-2 IgG and IgM antibodies in human plasma.
Collapse
Affiliation(s)
- Jia-Long Fang
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA.
| | - Leeza Shrestha
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Frederick A Beland
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| |
Collapse
|
23
|
Pujadas V, Chin C, Sankpal NV, Buhrmaster J, Arjuna A, Walia R, Smith MA, Eickelberg O, Bremner RM, Mohanakumar T, Sureshbabu A. Alveolar epithelial type 2 cell specific loss of IGFBP2 activates inflammation in COVID-19. Respir Res 2025; 26:111. [PMID: 40121473 PMCID: PMC11929192 DOI: 10.1186/s12931-025-03187-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 03/10/2025] [Indexed: 03/25/2025] Open
Abstract
The coronavirus disease 2019 (COVID-19) global pandemic is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, our understanding of SARS-CoV-2-induced inflammation in alveolar epithelial cells remains very limited. The contributions of intracellular insulin-like growth factor binding protein-2 (IGFBP2) to SARS-CoV-2 pathogenesis are also unclear. In this study, we have uncovered a critical role for IGFBP2, specifically in alveolar epithelial type 2 cells (AEC2), in the immunopathogenesis of COVID-19. Using bulk RNA sequencing, we show that IGFBP2 mRNA expression is significantly downregulated in primary AEC2 cells isolated from fibrotic lung regions from patients with COVID-19-acute respiratory distress syndrome (ARDS) compared to those with idiopathic pulmonary fibrosis (IPF) alone or IPF with a history of COVID-19. Using multicolor immunohistochemistry, we demonstrated that IGFBP2 and its selective ligands IGF1 and IGF2 were significantly reduced in AEC2 cells from patients with COVID-ARDS, IPF alone, or IPF with COVID history than in those from age-matched donor controls. Further, we demonstrated that lentiviral expression of Igfbp2 significantly reduced mRNA expression of proinflammatory cytokines-Tnf-α, Il1β, Il6, Stat3, Stat6 and chemokine receptors-Ccr2 and Ccr5-in mouse lung epithelial cells challenged with SARS-CoV-2 spike protein injury (S2; 500 ng/mL). Finally, we demonstrated higher levels of cytokines-TNF-α; IL-6 and chemokine receptor-CCR5 in AEC2 cells from COVID-ARDS patients compared to the IPF alone and the IPF with COVID history patients. Altogether, these data suggest that anti-inflammatory properties of IGFBP2 in AEC2 cells and its localized delivery may serve as potential therapeutic strategy for patients with COVID-19.
Collapse
Affiliation(s)
- Valentina Pujadas
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Chiahsuan Chin
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Narendra V Sankpal
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - James Buhrmaster
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Ashwini Arjuna
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Rajat Walia
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Michael A Smith
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Oliver Eickelberg
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Ross M Bremner
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Thalachallour Mohanakumar
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Angara Sureshbabu
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA.
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA.
| |
Collapse
|
24
|
Luo S, Yin L, Liu X, Wang X. Advances in Virus Biorecognition and Detection Techniques for the Surveillance and Prevention of Infectious Diseases. BIOSENSORS 2025; 15:198. [PMID: 40136995 PMCID: PMC11940537 DOI: 10.3390/bios15030198] [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] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Abstract
Viral infectious diseases pose a serious threat to global public health due to their high transmissibility, rapid mutation rates, and limited treatment options. Recent outbreaks of diseases such as plague, monkeypox, avian influenza, and coronavirus disease 2019 (COVID-19) have underscored the urgent need for efficient diagnostic and surveillance technologies. Focusing on viral infectious diseases that seriously threaten human health, this review summarizes and analyzes detection techniques from the perspective of combining viral surveillance and prevention advice, and discusses applications in improving diagnostic sensitivity and specificity. One of the major innovations of this review is the systematic integration of advanced biorecognition and detection technologies, such as bionanosensors, rapid detection test strips, and microfluidic platforms, along with the exploration of artificial intelligence in virus detection. These technologies address the limitations of traditional methods and enable the real-time monitoring and early warning of viral outbreaks. By analyzing the application of these technologies in the detection of pathogens, new insights are provided for the development of next-generation diagnostic tools to address emerging and re-emerging viral threats. In addition, we analyze the current progress of developed vaccines, combining virus surveillance with vaccine research to provide new ideas for future viral disease prevention and control and vaccine development, and call for global attention and the development of new disease prevention and detection technologies.
Collapse
Affiliation(s)
- Shuwen Luo
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China;
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China;
| | - Lihong Yin
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China;
| | - Xiaohui Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China;
| | - Xuemei Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China;
| |
Collapse
|
25
|
Hailu G, Legesse M, Mulu A, Medhin G, Tsegaye MM, Alemayehu DH, Ayele A, Gebreegziabxier A, Tayachew A, Aguine A, Dejene H, Tessema SK, Onywera H, Stanislas AE, Abate E, Marcello A, Bitew M. SARS-CoV-2 Genetic Variants Identified in Selected Regions of Ethiopia Through Whole Genome Sequencing: Insights from the Fifth Wave of COVID-19. Genes (Basel) 2025; 16:351. [PMID: 40149502 PMCID: PMC11942139 DOI: 10.3390/genes16030351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 02/19/2025] [Accepted: 02/23/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND The COVID-19 pandemic highlighted SARS-CoV-2 variants with increased transmissibility and immune evasion. In Ethiopia, where cases surged, the understanding of the virus's dynamics was limited. This study analyzed SARS-CoV-2 variants during the fifth wave, crucial for guiding vaccines, therapeutics, diagnostics, and understanding disease severity. METHOD From June to August 2022, 150 SARS-CoV-2-positive samples were randomly selected from the Ethiopian Public Health Institute repository. Sixty-three high-quality genome sequences were analyzed. RESULTS Of the 63 sequences, 70% were from males and 30% from females, with a median age of 34. Omicron dominated (97%, 61/63), primarily clade 22A (64%, 40/63), followed by 22B (18%, 11/63) and 21K (14%, 9/63). Delta accounted for 3.2% (2/63). Omicron was identified in all (25) vaccinated study participants. Ethiopian sequences showed limited evolutionary divergence and lower genetic diversity compared to global sequences. CONCLUSION Omicron was the predominant variant during Ethiopia's fifth wave, indicating recent community transmission. Despite minor genetic diversity differences, ongoing surveillance remains critical for tracking variants and informing public health interventions.
Collapse
Affiliation(s)
- Getnet Hailu
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa P.O. Box 1176, Ethiopia; (G.H.); (M.L.); (G.M.); (A.T.); (E.A.)
- Ethiopian Public Health Institute, Addis Ababa P.O. Box 1242, Ethiopia; (A.G.); (A.A.)
| | - Mengistu Legesse
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa P.O. Box 1176, Ethiopia; (G.H.); (M.L.); (G.M.); (A.T.); (E.A.)
| | - Andargachew Mulu
- Armaur Hansson Research Institute, Addis Ababa P.O. Box 1005, Ethiopia; (A.M.); (M.M.T.); (D.H.A.); (A.A.)
| | - Girmay Medhin
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa P.O. Box 1176, Ethiopia; (G.H.); (M.L.); (G.M.); (A.T.); (E.A.)
| | - Mesfin Mengesha Tsegaye
- Armaur Hansson Research Institute, Addis Ababa P.O. Box 1005, Ethiopia; (A.M.); (M.M.T.); (D.H.A.); (A.A.)
| | - Dawit Hailu Alemayehu
- Armaur Hansson Research Institute, Addis Ababa P.O. Box 1005, Ethiopia; (A.M.); (M.M.T.); (D.H.A.); (A.A.)
| | - Abaysew Ayele
- Armaur Hansson Research Institute, Addis Ababa P.O. Box 1005, Ethiopia; (A.M.); (M.M.T.); (D.H.A.); (A.A.)
| | | | - Adamu Tayachew
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa P.O. Box 1176, Ethiopia; (G.H.); (M.L.); (G.M.); (A.T.); (E.A.)
- Ethiopian Public Health Institute, Addis Ababa P.O. Box 1242, Ethiopia; (A.G.); (A.A.)
| | - Adimkewu Aguine
- Ethiopian Public Health Institute, Addis Ababa P.O. Box 1242, Ethiopia; (A.G.); (A.A.)
| | - Haileyesus Dejene
- College of Veterinary and Animal Science, University of Gondar, Gondar P.O. Box 196, Ethiopia;
| | - Sofonias K. Tessema
- Africa Centres for Disease Control and Prevention (Africa CDC), Addis Ababa P.O. Box 3243, Ethiopia; (S.K.T.); (H.O.)
| | - Harris Onywera
- Africa Centres for Disease Control and Prevention (Africa CDC), Addis Ababa P.O. Box 3243, Ethiopia; (S.K.T.); (H.O.)
| | | | - Ebba Abate
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa P.O. Box 1176, Ethiopia; (G.H.); (M.L.); (G.M.); (A.T.); (E.A.)
| | - Alessandro Marcello
- Laboratory of Molecular Virology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano 99, 34149 Trieste, Italy;
| | - Molalegne Bitew
- Bio and Emerging Technology Institute, Addis Ababa P.O. Box 5954, Ethiopia
| |
Collapse
|
26
|
Chakraborty D, Singh R, Rajmani RS, Kumar S, Ringe RP, Varadarajan R. Stabilizing Prefusion SARS-CoV-2 Spike by Destabilizing the Postfusion Conformation. Vaccines (Basel) 2025; 13:315. [PMID: 40266205 PMCID: PMC11946859 DOI: 10.3390/vaccines13030315] [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: 12/31/2024] [Revised: 03/01/2025] [Accepted: 03/11/2025] [Indexed: 04/24/2025] Open
Abstract
Background/Objectives: As with many viral fusion proteins, the native conformation of SARS-CoV-2 Spike is metastable. Most COVID-19 vaccines utilize a stabilized Spike (Spike-2P) containing two proline substitutions, and subsequently, a further stabilized variant with four additional proline substitutions, Spike-6P, has been developed. In an alternative approach, we introduced two aspartic acid residues (2D) in the HR1 region of Spike at positions that are exposed and buried in the pre- and postfusion states, respectively, to destabilize the postfusion conformation. Methods: The recombinant protein constructs were expressed in a mammalian cell culture and characterized for their yield and antigenicity, and the formulations were then used to immunize hamsters. After two immunizations, the hamsters were challenged with live B.1.351 SARS-CoV-2 virus for an evaluation of the protective efficacy. Results: The introduction of the two aspartic acid mutations resulted in an approximately six-fold increase in expression, comparable to that in Spike-2P. When the 2D mutations were combined with the above four proline mutations (Spike-4P-2D), this led to a further three- to four-fold enhancement of protein expression, similar to that seen in Spike-6P. When formulated with the oil-in-water emulsion adjuvant Sepivac SWE, the 2P, 2D, 6P, and 4P-2D Spike variants all protected female hamsters against heterologous challenge with the B.1.351 SARS-CoV-2 virus and elicited high titers of neutralizing antibodies. Conclusions: We suggest that destabilization of the postfusion conformation through the introduction of charged amino acids at sites that are exposed in the pre- and buried in the postfusion conformation offers a general strategy to enhance the yield and stability of the native, prefusion conformation of viral surface proteins.
Collapse
Affiliation(s)
- Debajyoti Chakraborty
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560012, India; (D.C.); (R.S.R.)
| | - Randhir Singh
- Mynvax Private Limited, 3rd Floor, Brigade MLR Centre, No.50, Vani Vilas Road, Basavanagudi, Bengaluru 560004, India;
| | - Raju S. Rajmani
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560012, India; (D.C.); (R.S.R.)
| | - Sahil Kumar
- Virology Unit, Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR), Chandigarh 160036, India; (S.K.); (R.P.R.)
| | - Rajesh P. Ringe
- Virology Unit, Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR), Chandigarh 160036, India; (S.K.); (R.P.R.)
| | - Raghavan Varadarajan
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560012, India; (D.C.); (R.S.R.)
| |
Collapse
|
27
|
Mani N, Suresh R, Chakraborty S. Cleaved vs. Uncleaved: How Furin Cleavage Reshapes the Conformational Landscape of SARS-CoV-2 Spike. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642945. [PMID: 40161653 PMCID: PMC11952566 DOI: 10.1101/2025.03.12.642945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The SARS-CoV-2 Spike protein is the primary target for vaccine design, with immunogens typically engineered to enhance stability by introducing proline mutations (2P) and mutating or deleting the Furin Cleavage Site (FCS). While these modifications improve structural integrity, studies suggest that furin cleavage can play a functional role in Spike protein dynamics, potentially enhancing ACE2 receptor binding. However, the impact of this cleavage on the unbound form of the Spike protein remains unclear. In this study, we use extensive all-atom molecular dynamics (MD) simulations to compare the structural and dynamic properties of cleaved and uncleaved Spike proteins in their pre-fusion, unbound state. Our results show that Furin cleavage significantly alters allosteric communication within the protein, increasing correlated motions between the Receptor Binding Domain (RBD) and N-terminal Domain (NTD), which may facilitate receptor engagement. Principal Component Analysis (PCA) reveals that the cleaved and uncleaved Spike proteins sample distinct conformational landscapes, with the cleaved form displaying enhanced flexibility and a broader range of RBD tilt angles. Additionally, Furin cleavage primes the S2 subunit by expanding the central helix, potentially influencing the transition to the post-fusion state. Glycan clustering patterns further suggest an adaptive structural response to cleavage, particularly in the NTD and RBD regions. These findings highlight the potential functional consequences of FCS deletion in immunogen design and underscore the importance of considering the native cleavage state in vaccine and therapeutic development.
Collapse
Affiliation(s)
- Natesan Mani
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115; Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115
| | - Raghavendran Suresh
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115; Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115
| | - Srirupa Chakraborty
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115; Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115
| |
Collapse
|
28
|
Koylass N, Sachithanandham J, Osei-Owusu J, Chen KH, Cheng HY, Pekosz A, Qiu Z. The proton-activated chloride channel inhibits SARS-CoV-2 spike protein-mediated viral entry through the endosomal pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642872. [PMID: 40161729 PMCID: PMC11952384 DOI: 10.1101/2025.03.12.642872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
SARS-CoV-2 binds to its obligatory receptor, angiotensin-converting enzyme 2 (ACE2) and capitalizes on decreasing endosomal acidity and cathepsin-mediated spike protein cleavage to enter cells. Endosomal acidification is driven by V-ATPase which pumps protons (H + ) into the lumen. The driving force for H + is maintained by the import of chloride (Cl - ) which is mediated by intracellular CLC transporters. We have recently identified the Proton-Activated Chloride (PAC) channel as a negative regulator of endosomal acidification. PAC responds to low pH and releases Cl - from the lumen to prevent endosomal hyperacidification. However, its role in SARS-CoV-2 viral entry remains unexplored. Here, we show that overexpressing the PAC channel in ACE2 expressing HEK 293T cells markedly inhibited the SARS-CoV-2 spike-mediated viral entry. Several lines of evidence suggest that this effect was due to the suppression of the endosomal entry pathway. First, the abilities of PAC to regulate endosomal acidification and inhibit pseudoviral entry were both dependent on its endosomal localization and channel activity. Second, the inhibitory effect on viral entry was similar to the suppression mediated by E64-d, a cathepsin inhibitor, while no major additive effect for both treatments was observed. Third, this inhibition was also attenuated in cells expressing TMPRSS2, which provides the alternative entry pathway through cell surface. Importantly, PAC overexpression also inhibited the number and size of plaques formed by two live SARS-CoV-2 isolates (B.1 and Omicron XBB.1.16) in Vero E6 cells. Altogether, our data indicates that PAC plays a vital role in inhibiting SARS-CoV-2 viral entry and identifies this endosomal channel as a potential novel target against the infection of SARS-CoV-2 and other viruses, which rely on the endosomal pathway.
Collapse
|
29
|
Park YJ, Jankowski W, Hurst NC, Fry JW, Schwabe NF, Tan LCC, Sauna ZE. Functional Activity and Binding Specificity of Small Ankyrin Repeat Proteins Called Ankyrons Against SARS-CoV-2 Variants. AAPS J 2025; 27:58. [PMID: 40069439 DOI: 10.1208/s12248-025-01043-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/15/2025] [Indexed: 03/25/2025] Open
Abstract
Effective management of COVID-19 requires clinical tools to treat the disease in addition to preventive vaccines. Several recombinant mAbs and their cocktails have been developed to treat COVID-19 but these have limitations. Here, we evaluate small ankyrin repeat proteins called Ankyrons that were generated to bind with high affinity to the SARS-CoV-2 virus. Ankyrons are ankyrin repeat proteins comprised of repetitions a structural module. Each module consists of a β-turn followed by two antiparallel α-helices. The Ankyrons™ are directly selected in vitro from a highly diverse library of around a trillion clones in ribosome display and like antibodies can bind with high affinity to almost any target. We assessed Ankyrons that were generated against the wild-type SARS-CoV-2 and the Delta (B.1.617.2) and Omicron (BA.1) variants in a binding assay. We determined that all Ankyrons were specific in that they did not bind to MERS. While all Ankyrons bound with high affinity to the variant they were generated against, some also showed cross-reactivity to all three SARS-CoV-2 variants. Binding assays are useful for screening analytes but do not provide information about clinical effectiveness. Therefore, we used a pseudovirus-based neutralization assay to show that five of the Ankyrons evaluated neutralized all three strains of SARS-CoV-2. We have provided a workflow for the evaluation of novel Ankyrons against a viral target. This suggests that Ankyrons could be useful for rapidly developing new research tools for studying other emerging infectious diseases rapidly with the optional further potential for developing Ankyrons into diagnostic and even therapeutic applications.
Collapse
Affiliation(s)
- Yun-Jong Park
- Hemostasis Branch 1, Division of Hemostasis, Office of Plasma Protein Therapeutics, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration (FDA), 10903 New Hampshire Ave, Silver Spring, Maryland, 20993, USA
| | - Wojciech Jankowski
- Hemostasis Branch 1, Division of Hemostasis, Office of Plasma Protein Therapeutics, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration (FDA), 10903 New Hampshire Ave, Silver Spring, Maryland, 20993, USA
| | - Nicholas C Hurst
- ProImmune Limited, Magdalen Centre, Oxford Science Park, Oxford, OX4 4GA, UK
| | - Jeremy W Fry
- ProImmune Limited, Magdalen Centre, Oxford Science Park, Oxford, OX4 4GA, UK
| | - Nikolai F Schwabe
- ProImmune Limited, Magdalen Centre, Oxford Science Park, Oxford, OX4 4GA, UK
| | - Linda C C Tan
- ProImmune Limited, Magdalen Centre, Oxford Science Park, Oxford, OX4 4GA, UK
| | - Zuben E Sauna
- Hemostasis Branch 1, Division of Hemostasis, Office of Plasma Protein Therapeutics, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration (FDA), 10903 New Hampshire Ave, Silver Spring, Maryland, 20993, USA.
| |
Collapse
|
30
|
Blizard GS, Dwivedi G, Pan YG, Hou C, Etersque JM, Said H, Chevrier A, Lavertu M, Ni H, Davis B, Tam Y, Cao Q, Mach RH, Weissman D, Alameh MG, Sellmyer MA. Monitoring mRNA vaccine antigen expression in vivo using PET/CT. Nat Commun 2025; 16:2234. [PMID: 40044669 PMCID: PMC11882883 DOI: 10.1038/s41467-025-57446-w] [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: 06/11/2024] [Accepted: 02/20/2025] [Indexed: 03/09/2025] Open
Abstract
Noninvasive visualization of the distribution and persistence of mRNA vaccine antigen expression in mammalian systems has implications for the development and evaluation of future mRNA vaccines. Here, we genetically fuse E. coli dihydrofolate reductase (eDHFR) to the delta furin diproline modified SARS-CoV-2 spike glycoprotein (S2P∆f) mRNA vaccine and image its expression in female mice and male non-human primates using [18F]fluoropropyl-trimethoprim ([18F]FP-TMP). Whole body positron emission tomography (PET) imaging revealed transient expression of the vaccine antigen in the injection site and draining lymph nodes (dLNs). Fusion of eDHFR did not impact S2P immunogenicity and no humoral or cellular immune response was detected against eDHFR in either species. In this work, we show that eDHFR can be used as an mRNA-encoded PET reporter gene to monitor the spatiotemporal dynamics of mRNA vaccine antigen expression in vivo. This technique could be applied in clinical translation of future mRNA vaccines or therapeutics.
Collapse
Affiliation(s)
- Gabrielle S Blizard
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Garima Dwivedi
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yi-Gen Pan
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Catherine Hou
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jean M Etersque
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hooda Said
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Anik Chevrier
- Chemical Engineering Department, Polytechnique Montreal, Montreal, QC, Canada
| | - Marc Lavertu
- Chemical Engineering Department, Polytechnique Montreal, Montreal, QC, Canada
| | - Houping Ni
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin Davis
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ying Tam
- Acuitas Therapeutics, Vancouver, BC, Canada
| | - Quy Cao
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert H Mach
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Drew Weissman
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamad-Gabriel Alameh
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Mark A Sellmyer
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
31
|
Gramm AJ, Braet SM, Srinivasu BY, Venkatakrishnan V, Bass EJ, Kearns FL, Calvó-Tusell C, Amaro RE, Stahelin RV, Wales TE, Anand GS. SARS CoV-2 spike adopts distinct conformational ensembles in situ. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.04.641425. [PMID: 40093071 PMCID: PMC11908172 DOI: 10.1101/2025.03.04.641425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Engineered recombinant Spike (S) has been invaluable for determining S structure and dynamics and is the basis for the design of most prevalent vaccines. While these vaccines have been highly efficacious for short-term protection from infection, protection waned with the emergence of variants (alpha through omicron). Here we report differences in conformational dynamics between native, membrane-embedded full-length S and recombinant S. Our virus-like particle (VLP) model mimics the native SARS CoV-2 virion by displaying S assembled with auxiliary E, M, and N proteins in a native membrane environment that captures the entirety of quaternary interactions mediated by S. Display of S on VLP obviates the requirement for stabilizing modifications that have been engineered into recombinant S for enhanced expression and solubility. Amide hydrogen/deuterium exchange mass spectrometry (HDXMS) reveals altered interprotomer contacts in VLP S trimers attributable to the presence of auxiliary proteins, membrane anchoring, and lack of engineered modifications. Our results reveal decreased dynamics in the S2 subunit and at sites spanning interprotomer contacts in VLP S with minimal differences in the N-terminal domain (NTD) and receptor binding domain (RBD). This carries implications for display of epitopes beyond NTD and RBD. In summary, despite affording efficient structural characterization, recombinant S distorts the intrinsic conformational ensemble of native S displayed on the virus surface.
Collapse
|
32
|
Devi S, Yadav N, Yadav R. Nanotechnology-Based Modern Biosensors for the Detection of SARS-CoV-2 Virus. Indian J Microbiol 2025; 65:177-188. [PMID: 40371028 PMCID: PMC12069202 DOI: 10.1007/s12088-024-01404-5] [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: 04/08/2024] [Accepted: 09/28/2024] [Indexed: 05/16/2025] Open
Abstract
The emergence of the COVID-19 pandemic has pointed out the urgent need for rapid and accurate diagnostic tools to detect the SARS-CoV-2 virus. Nanotechnology-based biosensors have emerged as a promising solution due to their high sensitivity, specificity, and speed in detecting biological molecules. This article focuses on the advancements in using nanotechnology for the development of modern biosensors tailored for the detection of the SARS-CoV-2 virus. Various nanomaterials, such as quantum dots, metallic nanoparticles, and nanowires, have been harnessed to enhance the performance of biosensors, offering improved detection limits and specificity. Besides this, innovative detection platforms, such as field-effect transistors, surface plasmon resonance, and electrochemical sensors, have revolutionized the landscape of SARS-CoV-2 diagnostics. These nanotechnology-based biosensors demonstrate the potential for point-of-care testing, enabling rapid and on-site detection with minimal sample preparation. The scalability, cost-effectiveness, and portability of these biosensors make them suitable for mass screening efforts in various healthcare settings, including hospitals, clinics, and community centers. The development of reliable biosensors for SARS-CoV-2 detection aligns with global efforts to curb the spread of the virus through early identification and containment strategies.
Collapse
Affiliation(s)
- Sandhya Devi
- Department of Biotechnology formerly known as Department of Bio and Nano Technology, Guru Jambheshwar University of Science and Technology, Hisar, Haryana 125001 India
| | - Neha Yadav
- Department of Biotechnology formerly known as Department of Bio and Nano Technology, Guru Jambheshwar University of Science and Technology, Hisar, Haryana 125001 India
- Central Instrumentation Laboratory, Central University of Punjab, Bathinda, Punjab 151401 India
| | - Rakesh Yadav
- Department of Biotechnology formerly known as Department of Bio and Nano Technology, Guru Jambheshwar University of Science and Technology, Hisar, Haryana 125001 India
| |
Collapse
|
33
|
Shimizu T, Abe S, Kawaguchi Y, Takata H, Ando H, Ishida T. A spleen-targeted vaccine for SARS-CoV-2 - Inducting neutralizing antibodies in mice. J Pharm Sci 2025; 114:1615-1624. [PMID: 39892872 DOI: 10.1016/j.xphs.2025.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 01/25/2025] [Accepted: 01/25/2025] [Indexed: 02/04/2025]
Abstract
The development of vaccines against infectious diseases is of the utmost importance to prevent global pandemics such as COVID-19. The application of antigens and adjuvants to efficiently target antigen presenting cells (APCs) is paramount for the development of efficient vaccines. In our previous study, we showed that splenic marginal zone-B (MZ-B) cells are promising APCs in addition to dendritic cells (DCs). In this study we achieved the targeted delivery of sufficient antigen to MZ-B cells by utilizing an intravenous (IV) immunization system we originally developed. This system involves the sequential injection of empty PEGylated liposomes (PEG-Lip) and antigen-containing PEG-Lip within a prescribed interval. Herein, we describe the application of this IV immunization system as a COVID-19 vaccine to induce specific antibodies against SARS-CoV-2. To establish efficacy, SARS-CoV-2 spike proteins were used as an antigen, and α-galactosylceramide (GC) was used as an adjuvant in this study. Three days after priming with empty PEG-Lip, we injected PEG-Lip containing spike protein and α-GC. Our IV immunization system successfully induced higher levels of anti-spike antibodies when spike protein derived from HEK-293, but not E. coli., was injected into mice. The levels were less produced using conventional immunization via subcutaneous (s.c.) injections of complete Freund's adjuvant without priming. Interestingly, a lower dose (0.2 µg) of spike protein antigen encapsulated into PEG-Lip induced a higher level of anti-spike antibodies than that produced using a significantly higher dose (5 µg). The induced anti-spike antibodies inhibited the interaction between the receptor binding domain of the spike protein and the angiotensin-converting enzyme 2. This indicates that the induced antibodies tend to neutralize SARS-CoV-2. Collectively, the specific delivery of spike proteins to spleen, probably MZ-B cells, via nano-carriers could be a promising approach for the development of global pandemic vaccines that require only minimum dosages of antigen.
Collapse
Affiliation(s)
- Taro Shimizu
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima, 770-8505, Japan.
| | - Shunji Abe
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima, 770-8505, Japan
| | - Yoshino Kawaguchi
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima, 770-8505, Japan
| | - Haruka Takata
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima, 770-8505, Japan; Innovative Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University, 770-8505, Tokushima, Japan
| | - Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima, 770-8505, Japan; Innovative Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University, 770-8505, Tokushima, Japan
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima, 770-8505, Japan; Innovative Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University, 770-8505, Tokushima, Japan.
| |
Collapse
|
34
|
Berkell M, Górska A, Smet M, Bachelet D, Gentilotti E, Guedes M, Franco-Yusti AM, Mazzaferri F, Forero EL, Matheeussen V, Visseaux B, Palacios-Baena ZR, Caroccia N, Florence AM, Charpentier C, van Leer C, Giannella M, Friedrich AW, Rodríguez-Baño J, Ghosn J, Kumar-Singh S, Laouénan C, Tacconelli E, Malhotra-Kumar S. Quasi-species prevalence and clinical impact of evolving SARS-CoV-2 lineages in European COVID-19 cohorts, January 2020 to February 2022. Euro Surveill 2025; 30:2400038. [PMID: 40084424 PMCID: PMC11912139 DOI: 10.2807/1560-7917.es.2025.30.10.2400038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 09/05/2024] [Indexed: 03/16/2025] Open
Abstract
BackgroundEvolution of SARS-CoV-2 is continuous.AimBetween 01/2020 and 02/2022, we studied SARS-CoV-2 variant epidemiology, evolution and association with COVID-19 severity.MethodsIn nasopharyngeal swabs of COVID-19 patients (n = 1,762) from France, Italy, Spain, and the Netherlands, SARS-CoV-2 was investigated by reverse transcription-quantitative PCR and whole-genome sequencing, and the virus variant/lineage (NextStrain/Pangolin) was determined. Patients' demographic and clinical details were recorded. Associations between mild/moderate or severe COVID-19 and SARS-CoV-2 variants and patient characteristics were assessed by logistic regression. Rates and genomic locations of mutations, as well as quasi-species distribution (≥ 2 heterogeneous positions, ≥ 50× coverage) were estimated based on 1,332 high-quality sequences.ResultsOverall, 11 SARS-CoV-2 clades infected 1,762 study patients of median age 59 years (interquartile range (IQR): 45-73), with 52.5% (n = 925) being male. In total, 101 non-synonymous substitutions/insertions correlated with disease prognosis (severe, n = 27; mild-to-moderate, n = 74). Several hotspots (mutation rates ≥ 85%) occurred in Alpha, Delta, and Omicron variants of concern (VOCs) but none in pre-Alpha strains. Four hotspots were retained across all study variants, including spike:D614G. Average number of mutations per open-reading-frame (ORF) increased in the spike gene (average < 5 per genome in January 2020 to > 15 in 2022), but remained stable in ORF1ab, membrane, and nucleocapsid genes. Quasi-species were most prevalent in 20A/EU2 (48.9%), 20E/EU1 (48.6%), 20A (38.8%), and 21K/Omicron (36.1%) infections. Immunocompromised status and age (≥ 60 years), while associated with severe COVID-19 or death irrespective of variant (odds ratio (OR): 1.60-2.25; p ≤ 0.014), did not affect quasi-species' prevalence (p > 0.05).ConclusionSpecific mutations correlate with COVID-19 severity. Quasi-species potentially shaping VOCs' emergence are relevant to consider.
Collapse
Affiliation(s)
- Matilda Berkell
- Laboratory of Medical Microbiology, Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
- Shared first author
- Molecular Pathology Group, Cell Biology & Histology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Anna Górska
- Shared first author
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Mathias Smet
- Laboratory of Medical Microbiology, Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
- Shared first author
- Molecular Pathology Group, Cell Biology & Histology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Delphine Bachelet
- Shared first author
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, IAME, Paris, France
- AP-HP Nord, Hôpital Bichat, Department of Epidemiology Biostatistics and Clinical Research, Paris, France
| | - Elisa Gentilotti
- Shared second author
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Mariana Guedes
- Shared second author
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Anna Maria Franco-Yusti
- Shared second author
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, IAME, Paris, France
- Service de Virologie, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | - Fulvia Mazzaferri
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Erley Lizarazo Forero
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Groningen, The Netherlands
| | - Veerle Matheeussen
- Laboratory of Medical Microbiology, Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Benoit Visseaux
- Service de Virologie, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | - Zaira R Palacios-Baena
- Infectious Diseases and Microbiology Unit, University Hospital Virgen Macarena, Department of Medicine, University of Seville, Biomedicine Institute of Seville/CSIC, Seville, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Natascia Caroccia
- Infectious Diseases Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Aline-Marie Florence
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, IAME, Paris, France
- AP-HP Nord, Hôpital Bichat, Department of Epidemiology Biostatistics and Clinical Research, Paris, France
| | - Charlotte Charpentier
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, IAME, Paris, France
- Service de Virologie, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | - Coretta van Leer
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Groningen, The Netherlands
| | - Maddalena Giannella
- Infectious Diseases Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Alex W Friedrich
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Groningen, The Netherlands
| | - Jesús Rodríguez-Baño
- Infectious Diseases and Microbiology Unit, University Hospital Virgen Macarena, Department of Medicine, University of Seville, Biomedicine Institute of Seville/CSIC, Seville, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Jade Ghosn
- AP-HP Nord, Hôpital Bichat, Department of Infectious and Tropical Diseases, Paris, France
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, IAME, Paris, France
| | - Samir Kumar-Singh
- Laboratory of Medical Microbiology, Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
- Molecular Pathology Group, Cell Biology & Histology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Cedric Laouénan
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, IAME, Paris, France
- AP-HP Nord, Hôpital Bichat, Department of Epidemiology Biostatistics and Clinical Research, Paris, France
| | - Evelina Tacconelli
- Shared senior author
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Surbhi Malhotra-Kumar
- Laboratory of Medical Microbiology, Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
- Shared senior author
| |
Collapse
|
35
|
Augustus AR, Radhakrishnan Y, Bhaskar JP, Ramamurthi S, Shunmugiah KP. Tannic acid modulates SARS-CoV-2 pathogenesis by curbing key host receptors and oxidative stress. Toxicol In Vitro 2025; 103:105971. [PMID: 39551113 DOI: 10.1016/j.tiv.2024.105971] [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: 01/04/2024] [Revised: 10/23/2024] [Accepted: 11/12/2024] [Indexed: 11/19/2024]
Abstract
The novel coronavirus SARS-CoV-2, which wrecked havoc around the world in the recent years through COVID-19, gains entry into the host cell through various receptors. Development of therapies targeting host-pathogen interaction will be a key to curb the infection as it potentially suppresses viral attachment and entry into the host. Boundless bioactives abundant in natural resources are the important source of new as well as safer alternatives. Tannic acid, a polyphenolic compound found abundantly in various plant sources, has gained much attention owing to its multifaceted pharmacological properties. This research paper presents a comprehensive investigation on antioxidant, anti-inflammatory and anti-viral abilities of tannic acid, substantiated through a triad of methodologies: in silico, in vitro and in vivo approaches. In vitro experiments, confirmed the antioxidant and anti-inflammatory efficacy as well as the host receptor modulating potential of tannic acid. In silico docking analyses elucidated the molecular interactions between tannic acid and key host receptors involved in inflammation and viral pathogenesis. Furthermore, the in vivo studies involving Danio rerio provided a holistic understanding of the systemic impact of tannic acid, including its antioxidant effects by mitigating the oxidative stress.
Collapse
Affiliation(s)
- Akshaya Rani Augustus
- Department of Biotechnology, Alagappa University, Karaikudi 630 003, Tamil Nadu, India
| | | | | | | | | |
Collapse
|
36
|
Cheng RL, Lim JPL, Fortuna MA, Reyes DV, Hans EADR, Nellas RB. Exploring the effects of N234 and N343 linked glycans to SARS CoV 2 spike protein pocket accessibility using Gaussian accelerated molecular dynamics simulations. Sci Rep 2025; 15:7052. [PMID: 40016249 PMCID: PMC11868416 DOI: 10.1038/s41598-025-85153-5] [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: 04/05/2024] [Accepted: 01/01/2025] [Indexed: 03/01/2025] Open
Abstract
The N234 and N343-linked glycans of the SARS-CoV 2 spike protein are known to stabilize the up-conformation of its receptor-binding domains (RBDs), enabling human angiotensin enzyme 2 (hACE2) receptor binding. However, the effect of spike-hACE2 binding on these important glycans remains poorly understood, and these changes could have implications in the development of drugs that inhibit viral entry. In this study, Gaussian accelerated molecular dynamics (GaMD) simulations of the hACE2-free and hACE2-bound spike protein are performed. Biophysical analyses were focused on the accessibility of three previously suggested druggable pockets underneath the three RBD subunits. A shielding effect by N234-linked glycans on the components of their adjacent pockets was observed. Although deshielding of central scaffold residues was observed in the hACE2-bound state, pocket A's accessibility was reduced due to an increase in NTDB-RBDB contacts, restricting entry into the pocket. For pocket B, changes in N234C and N343C expose the central scaffold residues in the bound state, increasing accessibility. In Pocket C, increased shielding due to N234A was found in the bound state, reducing accessibility. Despite these changes, the pockets remain accessible to ligands in both states and are still valid targets for drug development studies.
Collapse
Affiliation(s)
- Ronny L Cheng
- Institute of Chemistry, University of the Philippines Diliman, Quezon City, 1101, Philippines
| | - James Peter L Lim
- Institute of Chemistry, University of the Philippines Diliman, Quezon City, 1101, Philippines
| | - Myrnel A Fortuna
- Institute of Chemistry, University of the Philippines Diliman, Quezon City, 1101, Philippines
| | - Donnifer V Reyes
- Institute of Chemistry, University of the Philippines Diliman, Quezon City, 1101, Philippines
| | - Earl Adrian D R Hans
- Institute of Chemistry, University of the Philippines Diliman, Quezon City, 1101, Philippines
| | - Ricky B Nellas
- Institute of Chemistry, University of the Philippines Diliman, Quezon City, 1101, Philippines.
| |
Collapse
|
37
|
Jahantigh HR, Elsharkawy A, Guglani A, Arora K, Patterson LD, Kumar M. Neurobiological Alterations Induced by SARS-CoV-2: Insights from Variant-Specific Host Gene Expression Patterns in hACE2-Expressing Mice. Viruses 2025; 17:329. [PMID: 40143258 PMCID: PMC11946589 DOI: 10.3390/v17030329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
Since the onset of the COVID-19 pandemic, various severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) variants have emerged. Although the primary site of SARS-CoV-2 infection is the lungs, it can also affect the brain and induce neurological symptoms. However, the specific effects of different variants on the brain remain unclear. In this study, a whole-transcriptome analysis was conducted using the brain tissues of K18-hACE2 mice infected with the ancestral B.1 (Wuhan) variant and with major SARS-CoV-2 variants of concern, including B.1.1.7 (Alpha), B.1.351 (Beta), B.1.617.2 (Delta) and B.1.529 (Omicron). After sequencing, differential gene expression, gene ontology (GO) and genome pathway enrichment analyses were performed. An Immune Cell Abundance Identifier (ImmuCellAI) was used to identify the abundance of different cell populations. Additionally, RT-qPCR was used to validate the RNA-seq data. The viral load and hierarchical clustering analyses divided the samples into two different clusters with notable differences in gene expression at day 6 post-infection for all variants compared to the control group. GO and the Kyoto Encyclopedia of genes and genomes enrichment analyses revealed similar patterns of pathway enrichment for different variants. ImmuCellAI revealed the changes in immune cell populations, including the decrease in CD4+ T and B cell proportions and the increase in CD8+ T and dendritic cell proportions. A co-expression network analysis revealed that some genes, such as STAT1, interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α), were dysregulated in all variants. A RT-qPCR analysis for IL-6, CXCL10 and IRF7 further validated the RNA-seq analysis. In conclusion, this study provides, for the first time, an extensive transcriptome analysis of a K18-hACE2 mouse brain after infection with major SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Hamid Reza Jahantigh
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (H.R.J.); (A.E.); (A.G.); (K.A.); (L.D.P.)
| | - Amany Elsharkawy
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (H.R.J.); (A.E.); (A.G.); (K.A.); (L.D.P.)
- Center of Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Anchala Guglani
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (H.R.J.); (A.E.); (A.G.); (K.A.); (L.D.P.)
| | - Komal Arora
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (H.R.J.); (A.E.); (A.G.); (K.A.); (L.D.P.)
| | - Lila D. Patterson
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (H.R.J.); (A.E.); (A.G.); (K.A.); (L.D.P.)
| | - Mukesh Kumar
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (H.R.J.); (A.E.); (A.G.); (K.A.); (L.D.P.)
- Center of Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
38
|
Ghasemitarei M, Taeb H, Ghorbi T, Yusupov M, Ala-Nissila T, Bogaerts A. The effect of cysteine oxidation on conformational changes of SARS-CoV-2 spike protein using atomistic simulations. Sci Rep 2025; 15:6890. [PMID: 40011543 PMCID: PMC11865280 DOI: 10.1038/s41598-025-90918-z] [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: 12/23/2024] [Accepted: 02/17/2025] [Indexed: 02/28/2025] Open
Abstract
The SARS-CoV-2 Spike (S) protein plays a central role in viral entry into host cells, making it a key target for therapeutic interventions. Oxidative stress, often triggered during viral infections, can cause oxidation of cysteine in this protein. Here we investigate the impact of cysteine oxidation, specifically the formation of cysteic acid, on the conformational dynamics of the SARS-CoV-2 S protein using atomistic simulations. In particular, we examine how cysteine oxidation influences the transitions of the S protein's receptor-binding domain (RBD) between "down" (inaccessible) and "up" (accessible) states, which are critical for host cell receptor engagement. Using solvent-accessible surface area (SASA) analysis, we identify key cysteine residues susceptible to oxidation. The results of targeted molecular dynamics (TMD) and umbrella sampling (US) simulations reveal that oxidation reduces the energy barrier for RBD transitions by approximately 30 kJ mol-1, facilitating conformational changes and potentially enhancing viral infectivity. Furthermore, we analyze the interactions between oxidized cysteine residues and glycans, as well as alterations in hydrogen bonds and salt bridges. Our results show that oxidation disrupts normal RBD dynamics, influencing the energy landscape of conformational transitions. Our work provides novel insights into the role of cysteine oxidation in modulating the structural dynamics of the SARS-CoV-2 S protein, highlighting potential targets for antiviral strategies aimed at reducing oxidative stress or modifying post-translational changes. These findings contribute to a deeper understanding of viral infectivity and pathogenesis under oxidative conditions.
Collapse
Affiliation(s)
- Maryam Ghasemitarei
- Department of Applied Physics, Aalto University, P.O. Box 15600, 00076, Aalto, Espoo, Finland.
| | - Hoda Taeb
- Department of Physics, Simon Fraser University, Burnaby, Canada
| | - Tayebeh Ghorbi
- Laboratory of Experimental Biophysics, Centre for Advanced Technologies, 100174, Tashkent, Uzbekistan
| | - Maksudbek Yusupov
- Institute of Fundamental and Applied Research, National Research University TIIAME, 100000, Tashkent, Uzbekistan
- Department of Information Technologies, Tashkent International University of Education, 100207, Tashkent, Uzbekistan
| | - Tapio Ala-Nissila
- Department of Applied Physics, Aalto University, P.O. Box 15600, 00076, Aalto, Espoo, Finland
- Interdisciplinary Centre for Mathematical Modelling and Department of Mathematical Sciences, Loughborough University, Loughborough, Leicestershire, LE11 3TU, UK
| | - Annemie Bogaerts
- Research Group PLASMANT, Department of Chemistry, University of Antwerp, 2610, Antwerp, Belgium
| |
Collapse
|
39
|
Pitsillou E, El-Osta A, Hung A, Karagiannis TC. Epimaps of the SARS-CoV-2 Receptor-Binding Domain Mutational Landscape: Insights into Protein Stability, Epitope Prediction, and Antibody Binding. Biomolecules 2025; 15:301. [PMID: 40001604 PMCID: PMC11853434 DOI: 10.3390/biom15020301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/02/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants poses an ongoing threat to the efficacy of vaccines and therapeutic antibodies. Mutations predominantly affect the receptor-binding domain (RBD) of the spike protein, which mediates viral entry. The RBD is also a major target of monoclonal antibodies that were authorised for use during the pandemic. In this study, an in silico approach was used to investigate the mutational landscape of SARS-CoV-2 RBD variants, including currently circulating Omicron subvariants. A total of 40 single-point mutations were assessed for their potential effect on protein stability and dynamics. Destabilising effects were predicted for mutations such as L455S and F456L, while stabilising effects were predicted for mutations such as R346T. Conformational B-cell epitope predictions were subsequently performed for wild-type (WT) and variant RBDs. Mutations from SARS-CoV-2 variants were located within the predicted epitope residues and the epitope regions were found to correspond to the sites targeted by therapeutic antibodies. Furthermore, homology models of the RBD of SARS-CoV-2 variants were generated and were utilised for protein-antibody docking. The binding characteristics of 10 monoclonal antibodies against WT and 14 SARS-CoV-2 variants were evaluated. Through evaluating the binding affinities, interactions, and energy contributions of RBD residues, mutations that were contributing to viral evasion were identified. The findings from this study provide insight into the structural and molecular mechanisms underlying neutralising antibody evasion. Future antibody development could focus on broadly neutralising antibodies, engineering antibodies with enhanced binding affinity, and targeting spike protein regions beyond the RBD.
Collapse
MESH Headings
- SARS-CoV-2/genetics
- SARS-CoV-2/immunology
- SARS-CoV-2/chemistry
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/metabolism
- Humans
- Protein Stability
- Epitopes, B-Lymphocyte/immunology
- Epitopes, B-Lymphocyte/chemistry
- Epitopes, B-Lymphocyte/genetics
- Antibodies, Viral/immunology
- Mutation
- COVID-19/virology
- COVID-19/immunology
- Protein Domains
- Antibodies, Neutralizing/immunology
- Antibodies, Monoclonal/immunology
- Protein Binding
- Epitopes/immunology
- Epitopes/genetics
Collapse
Affiliation(s)
- Eleni Pitsillou
- Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC 3053, Australia
- School of Science, STEM College, RMIT University, Melbourne, VIC 3001, Australia
| | - Assam El-Osta
- Epigenetics in Human Health and Disease Program, Baker Heart and Diabetes Institute, 75 Commercial Road, Prahran, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville, VIC 3010, Australia
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
- Hong Kong Institute of Diabetes and Obesity, Prince of Wales Hospital, The Chinese University of Hong Kong, 3/F Lui Che Woo Clinical Sciences Building, 30–32 Ngan Shing Street, Sha Tin, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
- Biomedical Laboratory Science, Department of Technology, Faculty of Health, University College Copenhagen, 2200 Copenhagen, Denmark
| | - Andrew Hung
- School of Science, STEM College, RMIT University, Melbourne, VIC 3001, Australia
| | - Tom C. Karagiannis
- Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC 3053, Australia
- Epigenetics in Human Health and Disease Program, Baker Heart and Diabetes Institute, 75 Commercial Road, Prahran, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville, VIC 3010, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
40
|
Li R, Zhang J, Ren L. A Meta-Analysis of the Impact of Using Angiotensin-Converting Enzyme Inhibitors (ACEIs) or Angiotensin II Receptor Blockers (ARBs) on Mortality, Severity, and Healthcare Resource Utilization in Patients with COVID-19. Adv Respir Med 2025; 93:4. [PMID: 39996621 PMCID: PMC11852372 DOI: 10.3390/arm93010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 01/26/2025] [Indexed: 02/26/2025]
Abstract
OBJECTIVE The primary objective of this study is to explore the potential link between the utilization of angiotensin-converting enzyme inhibitors (ACEIs) or angiotensin II receptor blockers (ARBs) and its impact on mortality, disease severity, and healthcare resource utilization in individuals diagnosed with COVID-19. We aim to establish a solid theoretical foundation for safe and effective clinical medications. METHODS We conducted a comprehensive search of various databases, including CNKI, PubMed, Science, Cell, Springer, Nature, Web of Science, and Embase. We also traced the literature of the included studies to ensure a thorough analysis of the available evidence. After applying a set of inclusion and exclusion criteria, we ultimately included a total of 41 articles in our analysis. To determine the overall effect size for dichotomous variables, we used the Mantel-Haenszel odds ratio in random effect models. For continuous variables, we calculated the inverse variance SMD using random effect models. To assess the outcomes and heterogeneity, we considered p-values (p < 0.05) and I2 values for all outcomes. We performed multivariate and univariate meta-regression analyses using the maximum likelihood approach with the CMA 3.0 software. RESULTS The results of our analysis indicated that the use of ACEIs or ARBs did not significantly influence mortality (OR = 1.10, 95% CI 0.83-1.46, p = 0.43, I2 = 84%), severity (OR = 0.99, 95% CI 0.68-1.45, p = 0.98, I2 = 84%), or healthcare resource utilization (SMD = 0.03, 95% CI 0.06-0.12, p = 0.54, I2 = 37%) in patients with COVID-19 compared to those not taking ACEIs or ARBs. The multivariate meta-regression analysis model explained 63%, 31%, and 100% of the sources of heterogeneity for the three outcome indicators. CONCLUSIONS The use of ACEIs and ARBs is not significantly correlated with mortality, severity, or healthcare resource utilization in patients with COVID-19, indicating safe clinical use of the medications.
Collapse
Affiliation(s)
| | | | - Liang Ren
- Department of Forensic Medicine, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430032, China; (R.L.)
| |
Collapse
|
41
|
Burkova EE, Bakhno IA. Sequences in the Cytoplasmic Tail Contribute to the Intracellular Trafficking and the Cell Surface Localization of SARS-CoV-2 Spike Protein. Biomolecules 2025; 15:280. [PMID: 40001583 PMCID: PMC11853650 DOI: 10.3390/biom15020280] [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: 12/04/2024] [Revised: 01/09/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Spike protein is a surface glycoprotein of the SARS-CoV-2 coronavirus, providing interaction of the coronavirus with angiotensin-converting enzyme 2 (ACE2) on the host cell. The cytoplasmic tail of the S protein plays an important role in an intracellular transport and translocation of the glycoprotein to the plasma membrane. The cytoplasmic domain of the S protein contains binding sites for COPI, COPII, and SNX27, which are required for the intracellular trafficking of this glycoprotein. In addition, the cytoplasmic domain of the S protein contains S-palmitoylation sites. S-palmitoylation increases the hydrophobicity of the S protein by regulating its transport to the plasma membrane. The cytoplasmic tail of the S protein has a signaling sequence that provides interaction with the ERM family proteins, which may mediate communication between the cell membrane and the actin cytoskeleton. This review examines the role of the cytoplasmic tail of the SARS-CoV-2 S protein in its intracellular transport and translocation to the plasma membrane. Understanding these processes is necessary not only for the development of vaccines based on mRNA or adenovirus vectors encoding the full-length spike (S) protein, but also for the therapy of the new coronavirus infection (COVID-19).
Collapse
Affiliation(s)
- Evgeniya E. Burkova
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia;
| | | |
Collapse
|
42
|
Eriksson M, Larsson A. Avian Antibodies as Potential Therapeutic Tools. Antibodies (Basel) 2025; 14:18. [PMID: 39982233 PMCID: PMC11843883 DOI: 10.3390/antib14010018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/17/2025] [Accepted: 02/13/2025] [Indexed: 02/22/2025] Open
Abstract
Immunoglobulin Y (IgY) is the primary antibody found in the eggs of chicken (Gallus domesticus), allowing for large-scale antibody production with high titers, making them cost-effective antibody producers. IgY serves as a valuable alternative to mammalian antibodies typically used in immunodiagnostics and immunotherapy. Compared to mammalian antibodies, IgY offers several biochemical advantages, and its straightforward purification from egg yolk eliminates the need for invasive procedures like blood collection, reducing stress in animals. Due to the evolutionary differences between birds and mammals, chicken antibodies can bind to a broader range of epitopes on mammalian proteins than their mammalian counterparts. Studies have shown that chicken antibodies bind 3-5 times more effectively to rabbit IgG than swine antibodies, enhancing the signal in immunological assays. Additionally, IgY does not interact with rheumatoid factors or human anti-mouse IgG antibodies (HAMA), helping to minimize interference from these factors. IgY obtained from egg yolk of hens immunized against Pseudomonas aeruginosa has been used in patients suffering from cystic fibrosis and chronic pulmonary colonization with this bacterium. Furthermore, IgY has been used to counteract streptococcus mutans in the oral cavity and for the treatment of enteral infections in both humans and animals. However, the use of avian antibodies is limited to pulmonary, enteral, or topical application and should, due to immunogenicity, not be used for systemic administration. Thus, IgY expands the range of strategies available for combating pathogens in medicine, as a promising candidate both as an alternative to antibiotics and as a valuable tool in research and diagnostics.
Collapse
Affiliation(s)
- Mats Eriksson
- Department of Surgical Sciences, Section of Anaesthesiology and Intensive Care, Uppsala University, SE-751 85 Uppsala, Sweden
- NOVA Medical School, New University of Lisbon, 1099-085 Lisbon, Portugal
| | - Anders Larsson
- Department of Medical Sciences, Section of Clinical Chemistry, Uppsala University, SE-751 85 Uppsala, Sweden;
| |
Collapse
|
43
|
Arias-Arias JL, Monturiol-Gross L, Corrales-Aguilar E. A Live-Cell Imaging-Based Fluorescent SARS-CoV-2 Neutralization Assay by Antibody-Mediated Blockage of Receptor Binding Domain-ACE2 Interaction. BIOTECH 2025; 14:10. [PMID: 39982277 PMCID: PMC11843899 DOI: 10.3390/biotech14010010] [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: 12/12/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 02/22/2025] Open
Abstract
Neutralization assays have become an important tool since the beginning of the COVID-19 pandemic for testing vaccine responses and therapeutic antibodies as well as for monitoring humoral immunity to SARS-CoV-2 in epidemiological studies. The spike glycoprotein (S) present on the viral surface contains a receptor binding domain (RBD) that recognizes the angiotensin-converting enzyme 2 receptor (ACE2) in host cells, allowing virus entry. The gold standard for determining SARS-CoV-2 neutralizing antibodies is the plaque reduction neutralization test (PRNT), which relies on live-virus replication performed exclusively in biosafety level 3 (BSL-3) laboratories. Here, we report the development of a surrogate live-cell imaging-based fluorescent SARS-CoV-2 neutralization assay, applicable to BSL-1 or BSL-2 laboratories, by antibody-mediated blockage of the interaction between recombinant RBD with overexpressed ACE2 receptor in a genetically modified HEK 293T stable cell line. Our approach was able to detect neutralizing antibodies both in COVID-19-positive human serum samples and polyclonal equine formulations against SARS-CoV-2. This new cell-based surrogate neutralization assay represents a virus-free fluorescence imaging alternative to the reported approaches, which can be used to detect antibody-neutralizing capabilities toward SARS-CoV-2. This assay could also be extrapolated in the future to other established and emergent viral agents.
Collapse
Affiliation(s)
- Jorge L. Arias-Arias
- Centro de Investigación en Enfermedades Tropicales (CIET), Facultad de Microbiología, Universidad de Costa Rica, San José 11501-2060, Costa Rica;
- Dulbecco Lab Studio, Residencial Lisboa 2G, Alajuela 20102, Costa Rica
| | - Laura Monturiol-Gross
- Instituto Clodomiro Picado (ICP), Facultad de Microbiología, Universidad de Costa Rica, San José 11501-2060, Costa Rica;
| | - Eugenia Corrales-Aguilar
- Centro de Investigación en Enfermedades Tropicales (CIET), Facultad de Microbiología, Universidad de Costa Rica, San José 11501-2060, Costa Rica;
| |
Collapse
|
44
|
Jiang Y, Sanyal M, Hussein NA, Baghdasaryan A, Zhang M, Wang F, Ren F, Li J, Zhu G, Meng Y, Adamska JZ, Mellins E, Dai H. A SARS-CoV-2 vaccine on an NIR-II/SWIR emitting nanoparticle platform. SCIENCE ADVANCES 2025; 11:eadp5539. [PMID: 39919189 PMCID: PMC11804919 DOI: 10.1126/sciadv.adp5539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 01/07/2025] [Indexed: 02/09/2025]
Abstract
The COVID-19 pandemic caused a global health crisis that resulted in millions of deaths. Effective vaccines have played central roles in curtailing the pandemic. Here, we developed a down-converting near-infrared IIb (NIR-IIb; 1500 to 1700 nanometers) luminescent, pure NaErF4@NaYF4 rare-earth nanoparticle (pEr) as vaccine carriers. The pEr nanoparticles were coated with three layers of cross-linked biocompatible polymers (pEr-P3; ~55 nanometers) and conjugated to the receptor binding domain (RBD) of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein. Upon subcutaneous injection of the pEr-P3-RBD nanovaccine in mice, in vivo NIR-IIb imaging revealed active vaccine trafficking and migration to lymph nodes through lymphatic vessels. Two doses of the adjuvant-free vaccine elicited long-lasting (>7 months) high titers of serum viral neutralization antibody and anti-RBD immunoglobulin G, along with robust RBD-specific germinal center B cells and T follicular helper cells. We devised in vivo NIR-II molecular imaging of RBD-specific cells in lymph nodes, opening noninvasive assessments of vaccine-elicited immune responses longitudinally.
Collapse
Affiliation(s)
- Yingying Jiang
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
- Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong SAR
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU-SIRI, Shenzhen, P. R. China
| | - Mrinmoy Sanyal
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biochemistry, School of Medicine, Stanford University, Stanford, CA, USA
| | - Noor A. Hussein
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Ani Baghdasaryan
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Mengzhen Zhang
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Feifei Wang
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU-SIRI, Shenzhen, P. R. China
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Fuqiang Ren
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Jiachen Li
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Guanzhou Zhu
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Yifan Meng
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Julia Zofia Adamska
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | | | - Hongjie Dai
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
- Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong SAR
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU-SIRI, Shenzhen, P. R. China
| |
Collapse
|
45
|
Doi H, Nakano T, Sakakura K, Akisawa K, Okuwaki K, Hirano Y, Yamamoto E, Yasuoka K, Ohshima S, Katagiri T, Mochizuki Y. Large-Scale FMO-MP2 Calculations of the Spike Protein Droplet Model. J Comput Chem 2025; 46:e70052. [PMID: 39894970 PMCID: PMC11788466 DOI: 10.1002/jcc.70052] [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: 11/29/2024] [Revised: 01/05/2025] [Accepted: 01/17/2025] [Indexed: 02/04/2025]
Abstract
The spike protein of SARS-CoV-2 is a challenging target for theoretical approaches. Here we report a benchmark calculation of the spike protein droplet model by the fragment molecular orbital (FMO) at the second-order Møller-Plesset perturbation (MP2) level on the supercomputer Fugaku. One hundred structure samples from molecular dynamics (MD) simulations were used for both the closed and open forms of this protein (PDB IDs 6XLU and 6XM0 respectively). The number of total fragments is about 20,000, and the job time per structure was about 2 h on 8 racks of Fugaku.
Collapse
Affiliation(s)
- Hideo Doi
- Department of Chemistry and Research Center for Smart Molecules, Faculty of ScienceRikkyo UniversityTokyoJapan
| | - Tatsuya Nakano
- Department of HPC SupportResearch Organization for Information Science and TechnologyKobeJapan
| | | | - Kazuki Akisawa
- Department of Chemistry and Research Center for Smart Molecules, Faculty of ScienceRikkyo UniversityTokyoJapan
| | - Koji Okuwaki
- Department of Chemistry and Research Center for Smart Molecules, Faculty of ScienceRikkyo UniversityTokyoJapan
- JSOL Corp., KUDAN‐KAIKAN TERRACETokyoJapan
| | - Yoshinori Hirano
- Department of Mechanical EngineeringKeio UniversityYokohamaKanagawaJapan
| | - Eiji Yamamoto
- Department of System Design EngineeringKeio UniversityYokohamaKanagawaJapan
| | - Kenji Yasuoka
- Department of Mechanical EngineeringKeio UniversityYokohamaKanagawaJapan
| | - Satoshi Ohshima
- Research Institute for Information Technology, Kyushu UniversityFukuokaJapan
| | | | - Yuji Mochizuki
- Department of Chemistry and Research Center for Smart Molecules, Faculty of ScienceRikkyo UniversityTokyoJapan
- Institute of Industrial Science, the University of TokyoTokyoJapan
| |
Collapse
|
46
|
Lu Y, Li A, Shen F, He W, Yu S, Zhao Y, Feng X, Li M, Ouyang S, Zheng Y, Pang W. Recombinant protein HR212 targeting heptad repeat 2 domain in spike protein S2 subunit elicits broad-spectrum neutralizing antibodies against SARS-CoV-2 and its variants. MedComm (Beijing) 2025; 6:e70088. [PMID: 39931737 PMCID: PMC11808191 DOI: 10.1002/mco2.70088] [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: 06/09/2024] [Revised: 12/22/2024] [Accepted: 01/07/2025] [Indexed: 02/13/2025] Open
Abstract
SARS-CoV-2 variants are under continuous emergence carry numerous mutations within S1 subunit in spike protein and have escaped neutralization through many currently used vaccines and antibodies. The development of next-generation vaccines is a continuing and long-term need. In our prior research, the recombinant protein vaccine HR121 targeting the heptad repeat (HR) 1 domain of S2 subunit was constructed, which could evoke highly broad-spectrum neutralizing antibodies in vivo and confer efficient protective effect on several SARS-CoV-2 variants within multiple animal models. Compared with HR1, HR2 domain shows a more conservative degree within SARS-CoV-2 and related coronaviruses. Here, we designed a recombinant protein HR212 consisting of HR2-linker1-HR1-linker2-HR2. HR212 showed a high affinity with HR1 and was functionally analogous to HR2 within fusion intermediate in S2 subunit. Immunizing rabbits using HR212-mediated high nAbs for 28 pseudotyped SARS-CoV-2 variants, like currently circulating variants, such as BA.2.86 and JN.1. Transfer of rabbit anti-HR212 sera or immunization with HR212 offered efficient protective effect on SARS-CoV-2 ancestral strain and Omicron BA.2 variant infections of Syrian golden hamsters. According to our results, HR2 domain of S2 subunit is the novel target that can be used to develop broad-spectrum vaccines to resist SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Ying Lu
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ‐CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingYunnanChina
- University of Chinese Academy of SciencesBeijingChina
- Department of Biochemistry and Molecular BiologyFaculty of Basic Medical ScienceKunming Medical UniversityKunmingYunnanChina
| | - An‐Qi Li
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ‐CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingYunnanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Fan Shen
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ‐CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingYunnanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Wen‐Qiang He
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ‐CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingYunnanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Shu‐Heng Yu
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ‐CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingYunnanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yan‐Bo Zhao
- The Key Laboratory of Innate Immune Biology of Fujian Province, Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal UniversityFuzhouChina
| | - Xiao‐Li Feng
- Kunming National High‐level Biosafety Research Center for Non‐human Primates, Center for Biosafety Mega‐Science, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingYunnanChina
| | - Ming‐Hua Li
- Kunming National High‐level Biosafety Research Center for Non‐human Primates, Center for Biosafety Mega‐Science, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingYunnanChina
| | - Songying Ouyang
- The Key Laboratory of Innate Immune Biology of Fujian Province, Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal UniversityFuzhouChina
| | - Yong‐Tang Zheng
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ‐CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingYunnanChina
- University of Chinese Academy of SciencesBeijingChina
- EterniVax Biomedical IncShanghaiChina
| | - Wei Pang
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ‐CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingYunnanChina
- EterniVax Biomedical IncShanghaiChina
- Department of Pathogen Biology and ImmunologyFaculty of Basic Medical ScienceYunnan Provincial Key Laboratory of Public Health and Biosafety, Kunming Medical UniversityKunmingChina
| |
Collapse
|
47
|
Pozdnyakova V, Weber B, Cheng S, Ebinger JE. Review of Immunologic Manifestations of COVID-19 Infection and Vaccination. Rheum Dis Clin North Am 2025; 51:111-121. [PMID: 39550100 DOI: 10.1016/j.rdc.2024.09.001] [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] [Indexed: 11/18/2024]
Abstract
We herein summarize currently available and clinically relevant information regarding the human immune responses to SARS-CoV-2 infection and vaccination, in relation to COVID-19 outcomes with a focus on acute respiratory distress syndrome (ARDS) and myocarditis.
Collapse
Affiliation(s)
- Valeriya Pozdnyakova
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, D4005, Los Angeles, CA 90048, USA
| | - Brittany Weber
- Carl J. and Ruth Shapiro Cardiovascular Center, Brigham and Women's Hospital, 70 Francis Street, Boston, MA 02115, USA
| | - Susan Cheng
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, 127 South Vicente Boulevard, Suite A3100, Los Angeles, CA 90048, USA
| | - Joseph E Ebinger
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, 127 South Vicente Boulevard, Suite A3100, Los Angeles, CA 90048, USA.
| |
Collapse
|
48
|
Kumar A, Dutt M, Dehury B, Sganzerla Martinez G, Swan CL, Kelvin AA, Richardson CD, Kelvin DJ. Inhibition potential of natural flavonoids against selected omicron (B.1.19) mutations in the spike receptor binding domain of SARS-CoV-2: a molecular modeling approach. J Biomol Struct Dyn 2025; 43:1068-1082. [PMID: 38115191 PMCID: PMC11716671 DOI: 10.1080/07391102.2023.2291165] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/09/2023] [Indexed: 12/21/2023]
Abstract
The omicron (B.1.19) variant of contagious severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is considered a variant of concern (VOC) due to its increased transmissibility and highly infectious nature. The spike receptor-binding domain (RBD) is a hotspot of mutations and is regarded as a prominent target for screening drug candidates owing to its crucial role in viral entry and immune evasion. To date, no effective therapy or antivirals have been reported; therefore, there is an urgent need for rapid screening of antivirals. An extensive molecular modelling study has been performed with the primary goal to assess the inhibition potential of natural flavonoids as inhibitors against RBD from a manually curated library. Out of 40 natural flavonoids, five natural flavonoids, namely tomentin A (-8.7 kcal/mol), tomentin C (-8.6 kcal/mol), hyperoside (-8.4 kcal/mol), catechin gallate (-8.3 kcal/mol), and corylifol A (-8.2 kcal/mol), have been considered as the top-ranked compounds based on their binding affinity and molecular interaction profiling. The state-of-the-art molecular dynamics (MD) simulations of these top-ranked compounds in complex with RBD exhibited stable dynamics and structural compactness patterns on 200 nanoseconds. Additionally, complexes of these molecules demonstrated favorable free binding energies and affirmed the docking and simulation results. Moreover, the post-simulation validation of these interacted flavonoids using principal component analysis (PCA) revealed stable interaction patterns with RBD. The integrated results suggest that tomentin A, tomentin C, hyperoside, catechin gallate, and corylifol A might be effective against the emerging variants of SARS-CoV-2 and should be further evaluated using in-vitro and in-vivo experiments.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Anuj Kumar
- Laboratory of Immunity, Shantou University Medical College, Shantou, China
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, Canada
- Department of Paediatrics, IWK Health Center, Canadian Centre for Vaccinology (CCfV), Halifax, Canada
| | - Mansi Dutt
- Laboratory of Immunity, Shantou University Medical College, Shantou, China
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, Canada
- Department of Paediatrics, IWK Health Center, Canadian Centre for Vaccinology (CCfV), Halifax, Canada
| | - Budheswar Dehury
- Bioinformatics Division, ICMR-Regional Medical Research Centre, Bhubaneswar, India
| | - Gustavo Sganzerla Martinez
- Laboratory of Immunity, Shantou University Medical College, Shantou, China
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, Canada
- Department of Paediatrics, IWK Health Center, Canadian Centre for Vaccinology (CCfV), Halifax, Canada
| | - Cynthia L. Swan
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, Canada
| | - Alyson A. Kelvin
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, Canada
| | - Christopher D. Richardson
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, Canada
- Department of Paediatrics, IWK Health Center, Canadian Centre for Vaccinology (CCfV), Halifax, Canada
| | - David J. Kelvin
- Laboratory of Immunity, Shantou University Medical College, Shantou, China
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, Canada
- Department of Paediatrics, IWK Health Center, Canadian Centre for Vaccinology (CCfV), Halifax, Canada
| |
Collapse
|
49
|
Simpson J, Kasson PM. Structural prediction of chimeric immunogen candidates to elicit targeted antibodies against betacoronaviruses. PLoS Comput Biol 2025; 21:e1012812. [PMID: 39908344 PMCID: PMC11809852 DOI: 10.1371/journal.pcbi.1012812] [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: 10/01/2024] [Revised: 02/10/2025] [Accepted: 01/20/2025] [Indexed: 02/07/2025] Open
Abstract
Betacoronaviruses pose an ongoing pandemic threat. Antigenic evolution of the SARS-CoV-2 virus has shown that much of the spontaneous antibody response is narrowly focused rather than broadly neutralizing against even SARS-CoV-2 variants, let alone future threats. One way to overcome this is by focusing the antibody response against better-conserved regions of the viral spike protein. This has been demonstrated empirically in prior work, but we posit that systematic design tools will further potentiate antigenic focusing approaches. Here, we present a design approach to predict stable chimeras between SARS-CoV-2 and other coronaviruses, creating synthetic spike proteins that display a desired conserved region, in this case S2, and vary other regions. We leverage AlphaFold to predict chimeric structures and create a new metric for scoring chimera stability based on AlphaFold outputs. We evaluated 114 candidate spike chimeras using this approach. Top chimeras were further evaluated using molecular dynamics simulation as an intermediate validation technique, showing good stability compared to low-scoring controls. Experimental testing of five predicted-stable and two predicted-unstable chimeras confirmed 5/7 predictions, with one intermediate result. This demonstrates the feasibility of the underlying approach, which can be used to design custom immunogens to focus the immune response against a desired viral glycoprotein epitope.
Collapse
Affiliation(s)
- Jamel Simpson
- Program in Biophysics and Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Peter M. Kasson
- Program in Biophysics and Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Departments of Chemistry and Biochemistry and Biomedical Engineering, Georgia Institute of Technology, Atlanta, GeorgiaUnited States of America
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
50
|
Doroud D, Daneshi M, Kazemi-Lomedash F, Eftekhari Z. Comprehensive review of preclinical evaluation strategies for COVID-19 vaccine candidates: assessing immunogenicity, toxicology, and safety profiles. IRANIAN JOURNAL OF MICROBIOLOGY 2025; 17:1-18. [PMID: 40330066 PMCID: PMC12049746 DOI: 10.18502/ijm.v17i1.17796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Following the worldwide spread of Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), there is a vital requirement for safe and effective vaccines against Coronavirus disease 2019 (COVID-19). Therefore, several vaccine-candidate platforms have been designed, tested, and developed. Based on guidelines, preclinical studies are recommended to assess the safety and potency of COVID-19 vaccines in appropriate in vitro and in vivo settings. These studies provide essential information to describe the potential toxic properties of a vaccine and the formulation of vaccine agents during the preclinical trial phase. In toxicology studies, several factors must be considered, such as the appropriate animal species and strains, dosing timetable, mode of administration, time of sampling for biochemistry and antibody evaluation, and necropsy. Pharmacokinetic/ biodistribution studies are not usually required for infectious disease prophylaxis vaccines unless the vaccine contains a novel substance. Evaluating their biodistribution is crucial for newly developed vaccines, such as lipid nanoparticles -messenger RNA (LNP-mRNA), DNA, and Viral vectors in non-replicated (VVnr), or recombinant virus vaccines. The review highlights the importance of preclinical studies in assessing the safety and efficacy of vaccine candidates. This guidance is essential for researchers and manufacturers to design effective vaccines that can progress to clinical trials safely.
Collapse
Affiliation(s)
- Delaram Doroud
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Mojtaba Daneshi
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo,USA
| | - Fatemeh Kazemi-Lomedash
- Biotechnology Research Center, Department of Biotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Zohre Eftekhari
- Biotechnology Research Center, Department of Biotechnology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|