1
|
Liang J, Liu X, Liu Z, Xu X, Sun Y, Chen Y, Xiao R, Wang Y. Nanozymes featuring a mesoporous silica shell for rapid enrichment and ultrasensitive lateral flow immunoassay of influenza A. Anal Chim Acta 2025; 1335:343474. [PMID: 39643324 DOI: 10.1016/j.aca.2024.343474] [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: 11/06/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Respiratory illnesses stemming from influenza A viruses represent a significant worldwide health concern. There is an immediate need for a rapid and sensitive method to detect influenza A viruses early, without requiring extra equipment. RESULTS Here, we established a lateral flow immunoassay (LFIA) for the detection of influenza A (Flu A) using a "three-in-one" multifunctional mesoporous Fe3O4@SiO2@Pt nanozymes (Fe3O4@MSiO2@Pt NZs) with excellent magnetic separation properties, colorimetric, and peroxidase-like (POD-like) activities. Effective enrichment of target Flu A in complex samples as well as greater loading of Pt particles by mesoporous structures with large specific surface area to enhance POD-like activity can significantly improve the detection sensitivity of the LFIA. After colorimetric amplification by Fe3O4@MSiO2@Pt tags catalysis, the qualitative and quantitative results of detection for Flu A nucleoprotein (Flu A-NP) were 0.01 and 0.0089 ng mL-1, respectively. This indicated a sensitivity approximately 100 times greater than commercially available colloidal Au nanoparticle (AuNP)-based LFIA strips. For detection of inactivated H1N1 virus, quantification can be as low as 33 copies mL-1. Moreover, it demonstrated high accuracy in pharyngeal swab sample simulation experiments. SIGNIFICANCE Therefore, the proposed platform based on Fe3O4@MSiO2@Pt NZs-LFIA offered a promising approach for point-of-care testing (POCT), enabling rapid and ultrasensitive diagnosis of Flu A.
Collapse
Affiliation(s)
- Jing Liang
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China; State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Xiaoxian Liu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Zhenzhen Liu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Xuanming Xu
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Yinuo Sun
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Yuan Chen
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Rui Xiao
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China.
| | - Yajie Wang
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
| |
Collapse
|
2
|
Huang T, Yu J, Zhang S, Teng D, Dai D, Zhu Y, Gao L. Immunogenicity and safety of concomitant administration of recombinant COVID-19 vaccine and quadrivalent inactivated influenza vaccine in Chinese adults: An open-label, randomized, controlled trial. Hum Vaccin Immunother 2024; 20:2330770. [PMID: 38602539 PMCID: PMC11017946 DOI: 10.1080/21645515.2024.2330770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/12/2024] [Indexed: 04/12/2024] Open
Abstract
The immunogenicity and safety of the concomitant administration of recombinant COVID-19 vaccine and quadrivalent inactivated influenza vaccine (Split Virion) (QIIV) in Chinese adults are unclear. In this open-label, randomized controlled trial, participants aged ≥ 18 years were recruited. Eligible healthy adults were randomly assigned (1:1) to receive QIIV at the same time as the first dose of COVID-19 vaccine (simultaneous-group) or 14 days after the second dose of COVID-19 vaccine (non-simultaneous-group). The primary outcome was to compare the difference in immunogenicity of QIIV (H1N1, H3N2, Yamagata, and Victoria) between the two groups. A total of 299 participants were enrolled, 149 in the simultaneous-group and 150 in the non-simultaneous-group. There were no significant differences in geometric mean titer (GMT) [H1N1: 386.4 (95%CI: 299.2-499.0) vs. 497.4 (95%CI: 377.5-655.3); H3N2: 66.9 (95%CI: 56.1-79.8) vs. 81.4 (95%CI: 67.9-97.5); Yamagata: 95.6 (95%CI: 79.0-115.8) vs. 74.3 (95%CI: 58.6-94.0); and Victoria: 48.5 (95%CI: 37.6-62.6) vs. 65.8 (95%CI: 49.0-88.4)] and seroconversion rate (H1N1: 87.5% vs. 90.1%; H3N2: 58.1% vs. 62.0%; Yamagata: 75.0% vs. 64.5%; and Victoria: 55.1% vs. 62.8%) of QIIV antibodies between the simultaneous and non-simultaneous groups. For the seroprotection rate of QIIV antibodies, a higher seroprotection rate of Yamagata antibody was observed only in the simultaneous-group than in the non-simultaneous-group [86.0% vs. 76.0%, p = .040]. In addition, no significant difference in adverse events was observed between the two groups (14.2% vs. 23.5%, p = .053). In conclusion, no immune interference or safety concerns were found for concomitant administration of COVID-19 vaccine with QIIV in adults aged ≥ 18 years.
Collapse
Affiliation(s)
- Tao Huang
- Department of Vaccine Clinical Observation Center, Hunan Provincial Center for Disease Control and Prevention, Changsha, Hunan, P.R. China
| | - Jun Yu
- Jiangsu GDK Biological Technology Co, Ltd, Taizhou, Jiangsu, P.R. China
| | - Siyuan Zhang
- Department of Vaccine Clinical Observation Center, Hunan Provincial Center for Disease Control and Prevention, Changsha, Hunan, P.R. China
| | - Dewei Teng
- Department of Vaccine Clinical Observation Center, Hunan Provincial Center for Disease Control and Prevention, Changsha, Hunan, P.R. China
| | - Defang Dai
- Department of Vaccine Clinical Observation Center, Hunan Provincial Center for Disease Control and Prevention, Changsha, Hunan, P.R. China
| | - Yinbiao Zhu
- Jiangsu GDK Biological Technology Co, Ltd, Taizhou, Jiangsu, P.R. China
| | - Lidong Gao
- Department of Vaccine Clinical Observation Center, Hunan Provincial Center for Disease Control and Prevention, Changsha, Hunan, P.R. China
| |
Collapse
|
3
|
Mishra S, Jain D, Dey AA, Nagaraja S, Srivastava M, Khatun O, Balamurugan K, Anand M, Ashok AK, Tripathi S, Ganji M, Kesavardhana S. Bat RNA viruses employ viral RHIMs orchestrating species-specific cell death programs linked to Z-RNA sensing and ZBP1-RIPK3 signaling. iScience 2024; 27:111444. [PMID: 39697597 PMCID: PMC11652944 DOI: 10.1016/j.isci.2024.111444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 01/06/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
RHIM is a protein motif facilitating the assembly of large signaling complexes triggering regulated cell death. A few DNA viruses employ viral RHIMs mimicking host RHIMs and counteract cell death by interacting with host RHIM-proteins to alleviate antiviral defenses. Whether RNA viruses operate such viral RHIMs remains unknown. Here, we identified viral RHIMs in Nsp13 of SARS-CoV-2 and other bat RNA viruses, providing the basis for bats as the hosts for their evolution. Nsp13 promoted viral RHIM and RNA-binding channel-dependent cell death. However, Nsp13 viral RHIM is more critical for human cell death than in bat-derived Tb1 Lu cells, suggesting species-specific regulation. Nsp13 showed RHIM-dependent interactions with ZBP1 and RIPK3, forming large complexes and promoting ZBP1-RIPK3 signaling-mediated cell death. Intriguingly, the SARS-CoV-2 genome consisted of Z-RNA-forming segments promoting Nsp13-dependent cell death. Our findings reveal the functional viral RHIMs of bat-originated RNA viruses regulating host cell death associated with ZBP1-RIPK3 signaling, indicating possible mechanisms of cellular damage and cytokine storm in bat-originated RNA virus infections.
Collapse
Affiliation(s)
- Sanchita Mishra
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Disha Jain
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Ayushi Amin Dey
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Sahana Nagaraja
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Mansi Srivastava
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Oyahida Khatun
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru 560012, India
- Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru 560012, India
| | - Keerthana Balamurugan
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Micky Anand
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Avinash Karkada Ashok
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Shashank Tripathi
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru 560012, India
- Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru 560012, India
| | - Mahipal Ganji
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Sannula Kesavardhana
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| |
Collapse
|
4
|
Savaş Şen Z, Aydın Teke T, Yalcinkaya R, Özdem S, Gümüşer Cinni R, Çinar HG, Öztürk Z, Özdemir G, Akçaboy M, Polat M, Kaman A, Öz FN. Comparison of clinical and laboratory characteristics of COVID-19 and influenza in hospitalized children. Eur J Clin Microbiol Infect Dis 2024; 43:2419-2427. [PMID: 39392555 DOI: 10.1007/s10096-024-04964-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 10/08/2024] [Indexed: 10/12/2024]
Abstract
PURPOSE COVID-19 and influenza infections have similar modes of transmission and clinical symptoms but have different prognoses and treatment methods; therefore, it is important to make a final diagnosis. Herein, we aimed to compare the demographic, clinical, and laboratory differences in hospitalized pediatric patients with COVID-19 and influenza. METHODS This retrospective study comprised patients with COVID-19 managed between March 2020 to February 2022, and patients with influenza managed between December 2017 to February 2022, at a tertiary care hospital. The clinical data and laboratory parameters were obtained from the medical records of the hospital. Pediatric intensive care unit (PICU) admission, need for oxygen support, and the mortality rates of the patients were recorded and compared statistically. RESULTS Overall, 107 patients with COVID-19 and 250 patients with influenza were included. Underlying chronic disease (UCD) rates were statistically higher in patients with COVID-19 (p < 0.001). When the symptoms were compared, fever, cough, and runny nose were more common in patients with influenza, and abdominal pain and rash were more common in patients with COVID-19 (p < 0.05). In patients with influenza, white blood cell count and absolute neutrophil count values were lower (p = 0.021 and p = 0.037, respectively), and aspartate aminotransferase and creatinine kinase values were higher (p = 0.007 and p < 0.001, respectively). PICU admission rates and oxygen support needs were similar in both groups (p > 0.05). When the virus was COVID-19, it had 7.8 times higher risk of mortality compared to influenza (p = 0.002). There were statistically significant risk for mortality when the virus was COVID-19, the risk of mortality was 6.9 times higher in those with UCD, 8.5 times higher in those with admission to PICU and 3.8 times higher in those with needing mechanical ventilation (MV) compared to when the virus was influenza (p = 0.004, p = 0.006 and p = 0.049, respectively). The mortality rate was higher in patients with COVID-19 (p = 0.007). CONCLUSION This study showed that COVID-19 might negatively affect the survival times and increase mortality rates, especially in children with an UCD, admitted to the PICU and in need of MV.
Collapse
Affiliation(s)
- Zeynep Savaş Şen
- Department of Pediatric Infectious Disease, Ankara Etlik Integrated Health Campus, Ankara, 06170, Turkey.
| | - Türkan Aydın Teke
- Department of Pediatric Infectious Disease, Ankara Etlik Integrated Health Campus, Ankara, 06170, Turkey
| | - Rumeysa Yalcinkaya
- Department of Pediatric Infectious Disease, Ankara Etlik Integrated Health Campus, Ankara, 06170, Turkey
| | - Suna Özdem
- Department of Pediatric Infectious Disease, Mersin City Hospital, Mersin, Turkey
| | - Rüveyda Gümüşer Cinni
- Department of Pediatric Infectious Disease, Ankara Etlik Integrated Health Campus, Ankara, 06170, Turkey
| | - Hasibe Gökçe Çinar
- Department of Pediatric Radiology, Ankara Etlik Integrated Health Campus, Ankara, Turkey
| | - Zeynelabidin Öztürk
- Division of Pediatric Intensive Care, Ankara Etlik Integrated Health Campus, Ankara, Turkey
| | - Göktuğ Özdemir
- Division of Pediatric Intensive Care, Ankara Etlik Integrated Health Campus, Ankara, Turkey
| | - Meltem Akçaboy
- Department of Pediatrics, Ankara Etlik Integrated Health Campus, Ankara, Turkey
| | - Meltem Polat
- Faculty of Medicine, Department of Pediatric Infectious Disease, Gazi University, Ankara, Turkey
| | - Ayşe Kaman
- Department of Pediatric Infectious Disease, Ankara Etlik Integrated Health Campus, Ankara, 06170, Turkey
| | - Fatma Nur Öz
- Department of Pediatric Infectious Disease, Ankara Etlik Integrated Health Campus, Ankara, 06170, Turkey
| |
Collapse
|
5
|
Hemavathi KN, Middha SK, Raju R, Pilankatta R, Keshava Prasad TS, Abhinand CS. Computational screening of phytocompounds from C. amboinicus identifies potential inhibitors of influenza A (H3N2) virus by targeting hemagglutinin. J Biomol Struct Dyn 2024:1-13. [PMID: 39520503 DOI: 10.1080/07391102.2024.2424940] [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: 10/26/2023] [Accepted: 03/22/2024] [Indexed: 11/16/2024]
Abstract
The H3N2 subtype of the influenza A virus continues to be a notable public health issue due to its association with seasonal epidemics and severe human morbidity. The constrained effectiveness of current antiviral medications, combined with the inevitable emergence of drug-resistant variants, mandates the exploration of innovative therapeutic approaches. This study focuses on the identification of phytocompounds from Coleus amboinicus with the potential to target hemagglutinin, viral protein involved in viral entry by binding to sialyl glycoconjugates receptors on the surface of host cells. Molecular docking studies were carried out to assess the efficacy of C. amboinicus phytocompounds with hemagglutinin receptor-binding site. The study revealed that among the 84 signature phytocompounds, isosalvianolic acid and salvianolic acid C showed the highest docking scores and favourable intermolecular interactions. Pharmacokinetic analysis and Pan-assay interference compounds (PAINS) filtering confirmed that isosalvianolic acid meets the criteria outlined in Lipinski's rule of five, exhibits favourable ADMET profiles and passes PAINS filters. Furthermore, the molecular dynamics simulations followed by radius of gyration (Rg), solvent accessible surface area (SASA), and MM-PBSA calculations for binding free energy, verified the stability of the docked complexes. Together, the study identifies isosalvianolic acid as a promising inhibitor of the H3N2 virus by binding to hemagglutinin, indicating its potential as a strategy for therapeutic intervention.
Collapse
Affiliation(s)
| | - Sushil Kumar Middha
- Department of Biotechnology, Maharani Lakshmi Ammanni College for Women, Bengaluru, India
| | - Rajesh Raju
- Center for Integrative Omics Data Science, Yenepoya (Deemed to be University), Mangalore, India
| | - Rajendra Pilankatta
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Kasaragod, India
| | | | - Chandran S Abhinand
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| |
Collapse
|
6
|
Ong YC, Tejo BA, Yap WB. An Immunoinformatic Approach for Identifying and Designing Conserved Multi-Epitope Vaccines for Coronaviruses. Biomedicines 2024; 12:2530. [PMID: 39595095 PMCID: PMC11592158 DOI: 10.3390/biomedicines12112530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/30/2024] [Accepted: 11/02/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES The COVID-19 pandemic caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus has exposed the vulnerabilities and unpreparedness of the global healthcare system in dealing with emerging zoonoses. In the past two decades, coronaviruses (CoV) have been responsible for three major viral outbreaks, and the likelihood of future outbreaks caused by these viruses is high and nearly inevitable. Therefore, effective prophylactic universal vaccines targeting multiple circulating and emerging coronavirus strains are warranted. METHODS This study utilized an immunoinformatic approach to identify evolutionarily conserved CD4+ (HTL) and CD8+ (CTL) T cells, and B-cell epitopes in the coronaviral spike (S) glycoprotein. RESULTS A total of 132 epitopes were identified, with the majority of them found to be conserved across the bat CoVs, pangolin CoVs, endemic coronaviruses, SARS-CoV-2, and Middle East respiratory syndrome coronavirus (MERS-CoV). Their peptide sequences were then aligned and assembled to identify the overlapping regions. Eventually, two major peptide assemblies were derived based on their promising immune-stimulating properties. CONCLUSIONS In this light, they can serve as lead candidates for universal coronavirus vaccine development, particularly in the search for pan-coronavirus multi-epitope universal vaccines that can confer protection against current and novel coronaviruses.
Collapse
Affiliation(s)
- Yu Chuan Ong
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| | - Bimo Ario Tejo
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, Serdang 43400, Malaysia;
| | - Wei Boon Yap
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
- One Health UKM, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| |
Collapse
|
7
|
Chandra A, Kesavardhana S. PANoptosis Regulation in Reservoir Hosts of Zoonotic Viruses. Viruses 2024; 16:1733. [PMID: 39599847 PMCID: PMC11599095 DOI: 10.3390/v16111733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/25/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024] Open
Abstract
Zoonotic viruses originating from reservoir hosts, such as bats and birds, often cause severe illness and outbreaks amongst humans. Upon zoonotic virus transmission, infected cells mount innate immune responses that include the activation of programmed cell death pathways to recruit innate immune cells to the site of infection and eliminate viral replication niches. Different inflammatory and non-inflammatory cell death pathways, such as pyroptosis, apoptosis, necroptosis, and PANoptosis can undergo concurrent activation in humans leading to mortality and morbidity during zoonosis. While controlled activation of PANoptosis is vital for viral clearance during infection and restoring tissue homeostasis, uncontrolled PANoptosis activation results in immunopathology during zoonotic virus infections. Intriguingly, animal reservoirs of zoonotic viruses, such as bats and birds, appear to have a unique immune tolerance adaptation, allowing them to host viruses without succumbing to disease. The mechanisms facilitating high viral tolerance in bats and birds are poorly understood. In this perspective review, we discuss the regulation of PANoptotic pathways in bats and birds and indicate how they co-exist with viruses with mild clinical signs and no immunopathology. Understanding the PANoptotic machinery of bats and birds may thus assist us in devising strategies to contain zoonotic outbreaks amongst humans.
Collapse
Affiliation(s)
| | - Sannula Kesavardhana
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru 560012, India
| |
Collapse
|
8
|
Muthukutty P, MacDonald J, Yoo SY. Combating Emerging Respiratory Viruses: Lessons and Future Antiviral Strategies. Vaccines (Basel) 2024; 12:1220. [PMID: 39591123 PMCID: PMC11598775 DOI: 10.3390/vaccines12111220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024] Open
Abstract
Emerging viral diseases, including seasonal illnesses and pandemics, pose significant global public health risks. Respiratory viruses, particularly coronaviruses and influenza viruses, are associated with high morbidity and mortality, imposing substantial socioeconomic burdens. This review focuses on the current landscape of respiratory viruses, particularly influenza and SARS-CoV-2, and their antiviral treatments. It also discusses the potential for pandemics and the development of new antiviral vaccines and therapies, drawing lessons from past outbreaks to inform future strategies for managing viral threats.
Collapse
Affiliation(s)
| | | | - So Young Yoo
- Institute of Nanobio Convergence, Pusan National University, Busan 46241, Republic of Korea; (P.M.); (J.M.)
| |
Collapse
|
9
|
Cinatl J, Wass MN, Michaelis M. Multiple mechanisms enable broad-spectrum activity of the Pelargonium sidoides root extract EPs 7630 against acute respiratory tract infections. Front Pharmacol 2024; 15:1455870. [PMID: 39469622 PMCID: PMC11513585 DOI: 10.3389/fphar.2024.1455870] [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: 06/27/2024] [Accepted: 09/30/2024] [Indexed: 10/30/2024] Open
Abstract
There is clinical evidence showing that the Pelargonium sidoides root extract EPs 7630 is a safe and effective treatment for a range of acute infectious respiratory illnesses. Moreover, EPs 7630 has been shown to reduce the use of antibiotics, which is important in the context of rising antibiotic resistance levels. A wide range of mechanisms appears to contribute to the beneficial effects of EPs 7630, e.g. antibacterial, antiviral, immunomodulatory, and epithelial barrier effects. This broad spectrum of pharmacological activities seems to enable the clinical activity of EPs 7630 against multiple respiratory infections. In particular, the combination of antiviral and immunomodulatory effects may enable EPs 7630 to tackle acute viral respiratory infections both in early stages of the disease process, which are driven by virus replication, as well as in later stages, which are caused by an overshooting immune response. Hence, EPs 7630 is a prime example of a plant extract with evidence-based clinical efficacy, including a solid understanding of the underlying mechanisms of action. The example of EPs 7630 demonstrates that plant extracts have a potential role as evidence-based clinical treatments and that they deserve pre-clinical and clinical testing and investigation in the same way as any other drug class.
Collapse
Affiliation(s)
- Jindrich Cinatl
- Interdisciplinary Laboratory for Tumour and Virus Research, Dr Petra Joh Research Institute, Frankfurt am Main, Germany
| | - Mark N. Wass
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Martin Michaelis
- Interdisciplinary Laboratory for Tumour and Virus Research, Dr Petra Joh Research Institute, Frankfurt am Main, Germany
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| |
Collapse
|
10
|
Creusat F, Jouan Y, Gonzalez L, Barsac E, Ilango G, Lemoine R, Soulard D, Hankard A, Boisseau C, Guillon A, Lin Q, de Amat Herbozo C, Sencio V, Winter N, Sizaret D, Trottein F, Si-Tahar M, Briard B, Mallevaey T, Faveeuw C, Baranek T, Paget C. IFN-γ primes bone marrow neutrophils to acquire regulatory functions in severe viral respiratory infections. SCIENCE ADVANCES 2024; 10:eadn3257. [PMID: 39392875 PMCID: PMC11468905 DOI: 10.1126/sciadv.adn3257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 09/11/2024] [Indexed: 10/13/2024]
Abstract
Neutrophil subsets endowed with regulatory/suppressive properties are widely regarded as deleterious immune cells that can jeopardize antitumoral response and/or antimicrobial resistance. Here, we describe a sizeable fraction of neutrophils characterized by the expression of programmed death-ligand 1 (PD-L1) in biological fluids of humans and mice with severe viral respiratory infections (VRI). Biological and transcriptomic approaches indicated that VRI-driven PD-L1+ neutrophils are endowed with potent regulatory functions and reduced classical antimicrobial properties, as compared to their PD-L1- counterpart. VRI-induced regulatory PD-L1+ neutrophils were generated remotely in the bone marrow in an IFN-γ-dependent manner and were quickly mobilized into the inflamed lungs where they fulfilled their maturation. Neutrophil depletion and PD-L1 blockade during experimental VRI resulted in higher mortality, increased local inflammation, and reduced expression of resolving factors. These findings suggest that PD-L1+ neutrophils are important players in disease tolerance by mitigating local inflammation during severe VRI and that they may constitute relevant targets for future immune interventions.
Collapse
Affiliation(s)
- Florent Creusat
- INSERM, Centre d’Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France
- Université de Tours, Faculté de Médecine de Tours, Tours, France
| | - Youenn Jouan
- INSERM, Centre d’Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France
- Université de Tours, Faculté de Médecine de Tours, Tours, France
- Service de Médecine Intensive et Réanimation, CHRU de Tours, Tours, France
- Service de Chirurgie Cardiaque et de Réanimation Chirurgicale Cardio-Vasculaire, CHRU de Tours, Tours, France
| | - Loïc Gonzalez
- INSERM, Centre d’Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France
- Université de Tours, Faculté de Médecine de Tours, Tours, France
| | - Emilie Barsac
- INSERM, Centre d’Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France
- Université de Tours, Faculté de Médecine de Tours, Tours, France
| | - Guy Ilango
- INSERM, Centre d’Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France
- Université de Tours, Faculté de Médecine de Tours, Tours, France
| | - Roxane Lemoine
- Université de Tours, Faculté de Médecine de Tours, Tours, France
- Cytometry and Single-cell Immunobiology Core Facility, University of Tours, Tours, France
| | - Daphnée Soulard
- Centre d’Infection et d’Immunité de Lille, Inserm U1019, CNRS UMR 8204, Université de Lille, CHU Lille- Institut Pasteur de Lille, 59000 Lille, France
| | - Antoine Hankard
- INSERM, Centre d’Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France
- Université de Tours, Faculté de Médecine de Tours, Tours, France
| | - Chloé Boisseau
- INSERM, Centre d’Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France
- Université de Tours, Faculté de Médecine de Tours, Tours, France
| | - Antoine Guillon
- INSERM, Centre d’Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France
- Université de Tours, Faculté de Médecine de Tours, Tours, France
- Service de Médecine Intensive et Réanimation, CHRU de Tours, Tours, France
| | - Qiaochu Lin
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | | | - Valentin Sencio
- Centre d’Infection et d’Immunité de Lille, Inserm U1019, CNRS UMR 8204, Université de Lille, CHU Lille- Institut Pasteur de Lille, 59000 Lille, France
| | - Nathalie Winter
- INRAe (Institut National de la Recherche pour l'Agriculture, l'Alimentation et l’Environnement), Université de Tours, ISP, 37380 Nouzilly, France
| | - Damien Sizaret
- INSERM, Centre d’Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France
- Université de Tours, Faculté de Médecine de Tours, Tours, France
- Service d’Anatomie et Cytologie Pathologiques, CHRU de Tours, Tours, France
| | - François Trottein
- Centre d’Infection et d’Immunité de Lille, Inserm U1019, CNRS UMR 8204, Université de Lille, CHU Lille- Institut Pasteur de Lille, 59000 Lille, France
| | - Mustapha Si-Tahar
- INSERM, Centre d’Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France
- Université de Tours, Faculté de Médecine de Tours, Tours, France
| | - Benoit Briard
- INSERM, Centre d’Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France
- Université de Tours, Faculté de Médecine de Tours, Tours, France
| | - Thierry Mallevaey
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Christelle Faveeuw
- Centre d’Infection et d’Immunité de Lille, Inserm U1019, CNRS UMR 8204, Université de Lille, CHU Lille- Institut Pasteur de Lille, 59000 Lille, France
| | - Thomas Baranek
- INSERM, Centre d’Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France
- Université de Tours, Faculté de Médecine de Tours, Tours, France
| | - Christophe Paget
- INSERM, Centre d’Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France
- Université de Tours, Faculté de Médecine de Tours, Tours, France
| |
Collapse
|
11
|
Navanandan N, Jackson ND, Hamlington KL, Everman JL, Pruesse E, Secor EA, Stewart Z, Diener K, Hardee I, Edid A, Sulbaran H, Mistry RD, Florin TA, Yoder AC, Moore CM, Szefler SJ, Liu AH, Seibold MA. Viral Determinants of Childhood Asthma Exacerbation Severity and Treatment Response. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024:S2213-2198(24)00946-2. [PMID: 39368548 DOI: 10.1016/j.jaip.2024.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/30/2024] [Accepted: 09/18/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND Although respiratory viruses are common triggers of asthma exacerbations, the influence of viral infection characteristics on exacerbation presentation and treatment response in the pediatric emergency department (ED) is unclear. OBJECTIVE To assess viral infection characteristics of children experiencing ED asthma exacerbations and to test their associations with severity and treatment response. METHODS This is a prospective study of children, aged 4 to 18 years, who received standard ED asthma exacerbation treatment with inhaled bronchodilators and systemic corticosteroids. Nasal swabs collected for viral metagenomic analyses determined virus presence, load, and species. Outcomes included exacerbation severity (Pediatric Asthma Severity [PAS] score, clinician impression, and vital signs) and treatment response (discharge home without needing additional asthma therapies). RESULTS Of 107 children, 47% had moderate/severe exacerbations by PAS and 64% demonstrated treatment response. Viral metagenomic analysis on nasal swabs from 73 children detected virus in 86%, with 10 different species identified, primarily rhinovirus A (RV-A), RV-C, and enterovirus D68. Exacerbations involving RV-A were milder (odds ratio [OR] = 0.25; 95% confidence interval [CI] = 0.07-0.83) and tended to be more responsive to treatment than non-RV-A infections, whereas exacerbations involving enterovirus D68 were more severe (OR = 8.3; 95% CI = 1.3-164.7) and had no treatment response association. Viral load was not associated with treatment response but exhibited a strong linear relationship with heart rate (rpartial = 0.48), respiratory rate (rpartial = 0.25), and oxygen saturation (rpartial = -0.25), indicative of severity. CONCLUSIONS The majority of ED asthma exacerbations are triggered by respiratory viruses. Viral species are associated with severity and treatment response, suggesting that early pathogen detection could inform ED treatment decisions. Additional studies are needed to identify differences in pathobiology underlying exacerbations triggered by different viral species, and how to effectively treat these heterogeneous exacerbations.
Collapse
Affiliation(s)
- Nidhya Navanandan
- Section of Emergency Medicine, Department of Pediatrics, Children's Hospital Colorado, University of Colorado, Aurora, Colo.
| | - Nathan D Jackson
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colo
| | - Katharine L Hamlington
- Section of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital Colorado, University of Colorado, Aurora, Colo
| | - Jamie L Everman
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colo
| | - Elmar Pruesse
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colo
| | - Elizabeth A Secor
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colo
| | - Zoe Stewart
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colo
| | - Katrina Diener
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colo
| | - Isabel Hardee
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Alec Edid
- Section of Emergency Medicine, Department of Pediatrics, Children's Hospital Colorado, University of Colorado, Aurora, Colo
| | - Helio Sulbaran
- Section of Emergency Medicine, Department of Pediatrics, Children's Hospital Colorado, University of Colorado, Aurora, Colo
| | - Rakesh D Mistry
- Section of Pediatric Emergency Medicine, Department of Pediatrics, Yale University School of Medicine, New Haven, Conn
| | - Todd A Florin
- Division of Pediatric Emergency Medicine, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Angela C Yoder
- Colorado School of Public Health, University of Colorado Anschutz, Aurora, Colo
| | - Camille M Moore
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colo; Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Stanley J Szefler
- Section of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital Colorado, University of Colorado, Aurora, Colo
| | - Andrew H Liu
- Section of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital Colorado, University of Colorado, Aurora, Colo
| | - Max A Seibold
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colo; Department of Pediatrics, National Jewish Health, Denver, Colo; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colo
| |
Collapse
|
12
|
Shang C, Yu J, Zou S, Li H, Cao B. Functional evaluation of TMEM176B and its predictive role for severe respiratory viral infection through integrated analysis of single-cell and bulk RNA-sequencing. J Med Virol 2024; 96:e29954. [PMID: 39377494 DOI: 10.1002/jmv.29954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/05/2024] [Accepted: 09/26/2024] [Indexed: 10/09/2024]
Abstract
Transmembrane protein 176B (TMEM176B), localized mainly on the endosomal membrane, has been reported as an immune regulatory factor in malignant diseases. However, the biological function of this molecule remains undetermined during respiratory viral infections. To investigate the functions and prognostic value of this gene, six gene sets were selected from the Gene Expression Omnibus database for research. First, the function of TMEM176B and its co-expressed genes were evaluated at different levels (cell, peripheral blood, lung tissue). Afterwards, a machine learning algorithm was utilized to analyze the relationship between TMEM176B and its interacting genes with prognosis. After importance evaluation and variable screening, a prognostic model was established. Finally, the reliability of the model was further verified through external data sets. In vitro experiments were conducted to validate the function of TMEM176B. TMEM176B and its co-expressed genes are involved in multiple processes such as inflammasome activation, myeloid immune cell development, and immune cell infiltration. Machine learning further screened 27 interacting gene modules including TMEM176B as prognostic models for severe respiratory viral infections, with the area under the ROC curve (AUCs) of 0.986 and 0.905 in derivation and external validation sets, respectively. We further confirmed that viral load as well as NLRP3 activation and cell death were significantly enhanced in TMEM176B-/- THP-1-differentiated macrophages via in vitro experiments. Our study revealed that TMEM176B is involved in a wide range of biological functions in respiratory viral infections and has potential prognostic value, which is expected to bring new insights into the clinical management of severe respiratory viral infection hosts.
Collapse
Affiliation(s)
- Congcong Shang
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Jiapei Yu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Shumei Zou
- National Institute for Viral Disease Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory for Medical Virology, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hui Li
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Bin Cao
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China
- Department of Pulmonary and Critical Care Medicine, Clinical Center for Pulmonary Infections, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Song J, Zhang C, Fu S, Xu X. Visualized lateral flow assay for logic determination of co-existing viral RNA fragments. Biosens Bioelectron 2024; 261:116519. [PMID: 38917515 DOI: 10.1016/j.bios.2024.116519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/16/2024] [Accepted: 06/21/2024] [Indexed: 06/27/2024]
Abstract
Different types of pathogenic viruses that have common transmission path can be co-infected, inducing distinct disease procession in comparison to that infection of one. Also, in the post COVID-19 time, more types of respiratory infectious virus are becoming prevalent and are concurrent. Those bring an urgent need for detection of co-existing viruses. Here, we propose a visualized lateral flow assay for logic determination of co-existing viral RNA fragments. In the presence of specific viral RNA inputs, DNAzyme is de-blocked according to defined logic, and catalyzes the hydrolysis of hairpin-structural substrate. One of cleaved substrates contains DNAzyme domain to realize dual signal amplification, which obtains copious of the other cleaved substrates. The cleaved substrates act as linking strands for bridging DNA-modified gold nanoparticles onto lateral flow strip to induce coloration on test line. "AND", "OR" and "INHIBIT" controlled lateral flow assays are respectively demonstrated for co-existing viral RNA detection, and the visual results can be obtained by the same kind of prepared strip, without need of re-fabricating strips according to logic systems. The work provides a flexible, convenient, visual and logic-processing strategy for simultaneous analysis of co-existing viruses.
Collapse
Affiliation(s)
- Juanjuan Song
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China
| | - Chuhao Zhang
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China
| | - Siting Fu
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China
| | - Xiaowen Xu
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China.
| |
Collapse
|
14
|
Kollath DR, Grill FJ, Itogawa AN, Fabio-Braga A, Morales MM, Shepardson KM, Bryant ML, Yi J, Ramsey ML, Luberto ET, Celona KR, Keim PS, Settles EW, Lake D, Barker BM. Developing a Coccidioides posadasii and SARS-CoV-2 Co-infection Model in the K18-hACE2 Transgenic Mouse. COMMUNICATIONS MEDICINE 2024; 4:186. [PMID: 39349727 PMCID: PMC11442577 DOI: 10.1038/s43856-024-00610-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 09/16/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Early reports showed that patients with COVID-19 had recrudescence of previously resolved coccidioidomycosis (Valley fever, VF), and there were indications that coinfection had more severe outcomes. We therefore investigated serial infection of Coccidioides posadasii and SARS-CoV-2 in a K18-hACE2 mouse model to assess disease outcomes. METHODS In our model, we challenged K18-hACE2 mice sequentially with a sub-lethal dose of SARS-CoV-2 and 24 hours later with low virulence strain of Coccidioides posadasii, and vice versa, compared to mice that only received a single infection challenge. We performed survival and pathogenesis mouse studies as well as looked at the systemic immune response differences between treatment groups. RESULTS Here we show that co-infected groups have a more severe disease progression as well as a decrease in survival. Importantly, results differ depending on the SARS-CoV-2 variant (WA-1, Delta, or Omicron) and infection timing (SARS-CoV-2 first, C. posadasii second or vice versa). We find that groups that are infected with the virus first had a decrease in survival, increased morbidity and weight loss, increased fungal and viral burdens, differences in immune responses, and the amount and size of fungal spherules. We also find that groups coinfected with C. posadasii first have a decrease fungal burden and inflammatory responses. CONCLUSIONS This is the first in vivo model investigation of a coinfection of SARS-CoV-2 and Coccidioides. Because of the potential for increased severity of disease in a coinfection, we suggest populations that live in areas of high coccidioidomycosis endemicity may experience higher incidence of complicated disease progression with COVID-19.
Collapse
Affiliation(s)
- Daniel R Kollath
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | | | - Ashley N Itogawa
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | - Ana Fabio-Braga
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | - Matthew M Morales
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | - Kelly M Shepardson
- University of California, Merced, Department of Molecular Cell Biology, Merced, CA, USA
| | - Mitchell L Bryant
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | - Jinhee Yi
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | - Marieke L Ramsey
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | - Emily T Luberto
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | - Kimberly R Celona
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | - Paul S Keim
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | - Erik W Settles
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | - Douglas Lake
- School of Life Sciences at Arizona State University, Tempe, AZ, USA
| | - Bridget M Barker
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA.
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, USA.
| |
Collapse
|
15
|
Cai N, Zhan X, Zhang Q, Di H, Chen C, Hu Y, Yan X. Red Blood Cell-Derived Small Extracellular Vesicles Inhibit Influenza Virus through Surface-Displayed Sialic Acids. Angew Chem Int Ed Engl 2024:e202413946. [PMID: 39275883 DOI: 10.1002/anie.202413946] [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: 07/24/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 09/16/2024]
Abstract
Disrupting the conserved multivalent binding of hemagglutinin (HA) on influenza A virus (IAV) to sialic acids (SAs) on the host cell membrane offers a robust strategy to block viral attachment and infection, irrespective of antigenic evolution or drug resistance. In this study, we exploit red blood cell-derived small extracellular vesicles (RBC sEVs) as nanodecoys by harnessing their high abundance of surface-displayed SAs to interact with IAV through multivalent HA-SA interactions. This high-avidity binding inhibits viral adhesion to the cell surface, effectively preventing both attachment and infection in a dose-dependent manner. Notably, enzymatic removal of SAs from RBC sEVs significantly diminishes their anti-IAV efficacy. Our findings indicate that RBC sEVs possess intrinsic anti-IAV properties due to their native multivalent SAs and hold considerable promise as antiviral therapeutics.
Collapse
Affiliation(s)
- Niangui Cai
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Xiaozhen Zhan
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Qingyuan Zhang
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Haonan Di
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Chen Chen
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Yunyun Hu
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Xiaomei Yan
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| |
Collapse
|
16
|
Lv L, Chen Y, Zhao B. Pathogen shape: Implication on pathogenicity via respiratory deposition. ENVIRONMENT INTERNATIONAL 2024; 191:108978. [PMID: 39197372 DOI: 10.1016/j.envint.2024.108978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/06/2024] [Accepted: 08/21/2024] [Indexed: 09/01/2024]
Abstract
The shape of environmental aerosols contributes to the discrepancy in their dynamic behavior compared to spherical particles, which have received inadequate consideration. We reported deposition patterns of aerosols and aerosol-transmissible pathogens in real human respiratory systems, taking into account their actual shape, using a validated computational-based model. We found that the shape of the aerosols significantly influenced its deposits and accessibility within the respiratory system, significantly in the tracheobronchial region. As an example, we estimated that over 180 % of differences in deposits in the trachea and bronchi were attributable to pathogens shape, inferring the underlying pathogenicity difference of these regions. These findings, capturing the spatial heterogeneity of pathogens and aerosols deposition in human respiratory system, have major implication for understanding the evolution of aerosol-related disease.
Collapse
Affiliation(s)
- Lipeng Lv
- Department of Building Science, School of Architecture, Tsinghua University, Beijing 100084, China
| | - Yahong Chen
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing 100084, China
| | - Bin Zhao
- Department of Building Science, School of Architecture, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory of Indoor Air Quality Evaluation and Control, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
17
|
Macht A, Huang Y, Reinert LS, Grass V, Lohmer K, Aristizabal Prada ET, Babel E, Semmler A, Zhang W, Wegner A, Lichtenegger-Hartl E, Haas S, Hasenpusch G, Meyer S, Paludan SR, Pichlmair A, Rudolph C, Langenickel T. Mucosal IFNλ1 mRNA-based immunomodulation effectively reduces SARS-CoV-2 induced mortality in mice. EMBO Rep 2024; 25:3777-3788. [PMID: 39060455 PMCID: PMC11387833 DOI: 10.1038/s44319-024-00216-4] [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: 01/16/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
RNA vaccines elicit protective immunity against SARS-CoV-2, but the use of mRNA as an antiviral immunotherapeutic is unexplored. Here, we investigate the activity of lipidoid nanoparticle (LNP)-formulated mRNA encoding human IFNλ1 (ETH47), which is a critical driver of innate immunity at mucosal surfaces protecting from viral infections. IFNλ1 mRNA administration promotes dose-dependent protein translation, induction of interferon-stimulated genes without relevant signs of unspecific immune stimulation, and dose-dependent inhibition of SARS-CoV-2 replication in vitro. Pulmonary administration of IFNλ1 mRNA in mice results in a potent reduction of virus load, virus-induced body weight loss and significantly increased survival. These data support the development of inhaled administration of IFNλ1 mRNA as a potential prophylactic option for individuals exposed to SARS-CoV-2 or at risk suffering from COVID-19. Based on the broad antiviral activity of IFNλ1 regardless of virus or variant, this approach might also be utilized for other respiratory viral infections or pandemic preparedness.
Collapse
Affiliation(s)
| | - Yiqi Huang
- Institute of Virology, Technical University of Munich, Munich, Germany
| | - Line S Reinert
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Vincent Grass
- Institute of Virology, Technical University of Munich, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | - Søren R Paludan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Andreas Pichlmair
- Institute of Virology, Technical University of Munich, Munich, Germany
| | | | | |
Collapse
|
18
|
Uematsu T, Takai-Todaka R, Haga K, Kobayashi H, Imajima M, Kobayashi N, Katayama K, Hanaki H. Pharmacological effect of cepharanthine on SARS-CoV-2-induced disease in a Syrian hamster model. J Infect Chemother 2024:S1341-321X(24)00237-X. [PMID: 39197667 DOI: 10.1016/j.jiac.2024.08.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/07/2024] [Accepted: 08/26/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), remains a global public health threat. Although several effective vaccines and therapeutics have been developed, continuous emergence of new variants necessitates development of drugs with different mechanisms of action. Recent studies indicate that cepharanthine, a chemical derivative purified from Stephania cepharantha, inhibits SARS-CoV-2 replication in vitro. METHODS This study examined the in vivo effects of cepharanthine using a Syrian hamster SARS-CoV-2 infection model. To evaluate the prophylactic and therapeutic effects, cepharanthine was intranasally administered before or after SARS-CoV-2 infection. Effects were assessed by monitoring body weight changes, lung pathology, lung viral load, and inflammatory response in the lungs. RESULTS Pre-infection administration of cepharanthine resulted in less weight loss, reduced virus titers, alleviated histopathological severity, and decreased lung inflammation. Furthermore, post-infection administration of cepharanthine also exhibited therapeutic effects. CONCLUSIONS This study demonstrated that both prophylactic and therapeutic administration of cepharanthine reduces the pathogenesis of COVID-19 in a Syrian hamster SARS-CoV-2 infection model. Our findings suggest that cepharanthine is a potential therapeutic agent against COVID-19.
Collapse
Affiliation(s)
- Takayuki Uematsu
- Biomedical Laboratory, Division of Biomedical Research, Kitasato University Medical Center, Arai, Kitamoto, Saitama, Japan.
| | - Reiko Takai-Todaka
- Laboratory of Viral Infection Control, Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, Japan.
| | - Kei Haga
- Laboratory of Viral Infection Control, Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, Japan.
| | - Hideyuki Kobayashi
- Tokyo New Drug Research Laboratories, Pharmaceutical Business Unit, Kowa Company, Ltd., Tokyo, Japan.
| | - Makiko Imajima
- Tokyo New Drug Research Laboratories, Pharmaceutical Business Unit, Kowa Company, Ltd., Tokyo, Japan.
| | - Noritada Kobayashi
- Biomedical Laboratory, Division of Biomedical Research, Kitasato University Medical Center, Arai, Kitamoto, Saitama, Japan.
| | - Kazuhiko Katayama
- Laboratory of Viral Infection Control, Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, Japan.
| | - Hideaki Hanaki
- Infection Control Research Center, Ōmura Satoshi Memorial Institute, Kitasato University, Tokyo, Japan.
| |
Collapse
|
19
|
Yang D, Almanzar N, Xia J, Udit S, Yeung ST, Khairallah C, Hoagland DA, Umans BD, Sarden N, Erdogan O, Baalbaki N, Beekmayer-Dhillon A, Lee J, Meerschaert KA, Liberles SD, Yipp BG, Franklin RA, Khanna KM, Baral P, Haber AL, Chiu IM. Vagal TRPV1 + sensory neurons regulate myeloid cell dynamics and protect against influenza virus infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.21.609013. [PMID: 39229208 PMCID: PMC11370462 DOI: 10.1101/2024.08.21.609013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Influenza viruses are a major global cause of morbidity and mortality. Vagal TRPV1 + nociceptive sensory neurons, which innervate the airways, are known to mediate defenses against harmful agents. However, their function in lung antiviral defenses remains unclear. Our study reveals that both systemic and vagal-specific ablation of TRPV1 + nociceptors reduced survival in mice infected with influenza A virus (IAV), despite no significant changes in viral burden or weight loss. Mice lacking nociceptors showed exacerbated lung pathology and elevated levels of pro-inflammatory cytokines. The increased mortality was not attributable to the loss of the TRPV1 ion channel or neuropeptides CGRP or substance P. Immune profiling through flow cytometry and single-cell RNA sequencing identified significant nociceptor deficiency-mediated changes in the lung immune landscape, including an expansion of neutrophils and monocyte-derived macrophages. Transcriptional analysis revealed impaired interferon signaling in these myeloid cells and an imbalance in distinct neutrophil sub-populations in the absence of nociceptors. Furthermore, anti-GR1-mediated depletion of myeloid cells during IAV infection significantly improved survival, underscoring a role of nociceptors in preventing pathogenic myeloid cell states that contribute to IAV-induced mortality. One Sentence Summary : TRPV1 + neurons facilitate host survival from influenza A virus infection by controlling myeloid cell responses and immunopathology.
Collapse
|
20
|
Rajanala K, Upadhyay AK. Vaccines for Respiratory Viruses-COVID and Beyond. Vaccines (Basel) 2024; 12:936. [PMID: 39204059 PMCID: PMC11360283 DOI: 10.3390/vaccines12080936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
The COVID-19 (coronavirus disease 2019) pandemic had an extensive impact on global morbidity and mortality. Several other common respiratory viruses, such as the influenza virus and respiratory syncytial virus (RSV), are endemic or epidemic agents causing acute respiratory infections that are easily transmissible and pose a significant threat to communities due to efficient person-to-person transmission. These viruses can undergo antigenic variation through genetic mutations, resulting in the emergence of novel strains or variants, thereby diminishing the effectiveness of current vaccines, and necessitating ongoing monitoring and adjustment of vaccine antigens. As the virus-specific immunity is maintained only for several weeks or months after the infection, there is an emergent need to develop effective and durable vaccines. Additionally, specific populations, such as elderly or immunocompromised individuals, may exhibit reduced immune responses to respiratory viruses, posing significant challenges to develop vaccines that elicit durable and potent immunity. We present a comprehensive review of the molecular mechanisms underlying the pathogenesis and virulence of common respiratory viruses, such as RSV, influenza virus, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We discuss several vaccine approaches that are under development. A thorough understanding of the current strategies and the challenges encountered during the vaccine development process can lead to the advancement of effective next-generation vaccines.
Collapse
|
21
|
Huang L, Tang W, He L, Li M, Lin X, Hu A, Huang X, Wu Z, Wu Z, Chen S, Hu Y. Engineered probiotic Escherichia coli elicits immediate and long-term protection against influenza A virus in mice. Nat Commun 2024; 15:6802. [PMID: 39122688 PMCID: PMC11315933 DOI: 10.1038/s41467-024-51182-3] [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: 02/09/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Influenza virus infection remains a major global health problem and requires a universal vaccine with broad protection against different subtypes as well as a rapid-response vaccine to provide immediate protection in the event of an epidemic outbreak. Here, we show that intranasal administration of probiotic Escherichia coli Nissle 1917 activates innate immunity in the respiratory tract and provides immediate protection against influenza virus infection within 1 day. Based on this vehicle, a recombinant strain is engineered to express and secret five tandem repeats of the extracellular domain of matrix protein 2 from different influenza virus subtypes. Intranasal vaccination with this strain induces durable humoral and mucosal responses in the respiratory tract, and provides broad protection against the lethal challenge of divergent influenza viruses in female BALB/c mice. Our findings highlight a promising delivery platform for developing mucosal vaccines that provide immediate and sustained protection against respiratory pathogens.
Collapse
Affiliation(s)
- Ling Huang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei Tang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Lina He
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Mengke Li
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xian Lin
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- Hubei JiangXia Laboratory, Wuhan, 430071, China
| | - Ao Hu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xindi Huang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Zhouyu Wu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhiyong Wu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shiyun Chen
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Yangbo Hu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
- Hubei JiangXia Laboratory, Wuhan, 430071, China.
| |
Collapse
|
22
|
Moore S, Gopichandran K, Sevier E, Gamare S, Almuntashiri S, Ramírez G, Regino N, Jiménez-Alvarez L, Cruz-Lagunas A, Rodriguez-Reyna TS, Zuñiga J, Owen CA, Wang X, Zhang D. Club Cell Secretory Protein-16 (CC16) as a Prognostic Biomarker for COVID-19 and H1N1 Viral Infections. Diagnostics (Basel) 2024; 14:1720. [PMID: 39202207 PMCID: PMC11353392 DOI: 10.3390/diagnostics14161720] [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: 05/22/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) and H1N1 viruses are inflammatory lung pathogens that can lead to acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). ALI/ARDS are still life-threatening diseases in critically ill patients with 30-40% mortality in the last decade. Currently, there are no laboratory tests for the early diagnosis or prognosis of ALI/ARDS. Club cell secretory protein (CC16) has been investigated as a potential biomarker of lung epithelial damage in various lung diseases. In this study, we evaluated whether plasma CC16 reflects the severity of COVID-19 and H1N1 infections. The plasma CC16 levels showed no significant differences between H1N1 and COVID-19 groups (p = 0.09). Among all subjects, CC16 levels were significantly higher in non-survivors than in survivors (p = 0.001). Upon the area under the receiver operating characteristic (AUROC) analysis, CC16 had an acceptable value to distinguish survivors and non-survivors (p = 0.002). In the COVID-19 group, plasma CC16 levels moderately correlated with the Acute Physiology and Chronic Health Evaluation II (APACHE II) score (r = 0.374, p = 0.003) and Sequential Organ Failure Assessment (SOFA) score (r = 0.474, p < 0.001). In the H1N1 group, a positive correlation was observed between the CC16 levels and hospital length of stay (r = 0.311, p = 0.022). Among all the patients, weak correlations between plasma CC16 levels with the SOFA score (r = 0.328, p < 0.001) and hospital length of stay (r = 0.310, p < 0.001) were observed. Thus, circulating CC16 might reflect the severity of COVID-19 and H1N1 infections.
Collapse
Affiliation(s)
- Shane Moore
- Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
| | - Keerthana Gopichandran
- Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
| | - Elizabeth Sevier
- Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
| | - Siddhika Gamare
- Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
| | - Sultan Almuntashiri
- Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
- Department of Clinical Pharmacy, College of Pharmacy, University of Hail, Hail 55473, Saudi Arabia
| | - Gustavo Ramírez
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Tlalpan 4502, Mexico City 14080, Mexico
| | - Nora Regino
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Tlalpan 4502, Mexico City 14080, Mexico
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Mexico City 14380, Mexico
| | - Luis Jiménez-Alvarez
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Tlalpan 4502, Mexico City 14080, Mexico
| | - Alfredo Cruz-Lagunas
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Tlalpan 4502, Mexico City 14080, Mexico
| | - Tatiana S. Rodriguez-Reyna
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City 14080, Mexico
| | - Joaquin Zuñiga
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Tlalpan 4502, Mexico City 14080, Mexico
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Mexico City 14380, Mexico
| | - Caroline A. Owen
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xiaoyun Wang
- Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
| | - Duo Zhang
- Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
23
|
Baral B, Saini V, Kandpal M, Kundu P, Dixit AK, Parmar HS, Meena AK, Trivedi P, Jha HC. The interplay of co-infections in shaping COVID-19 severity: Expanding the scope beyond SARS-CoV-2. J Infect Public Health 2024; 17:102486. [PMID: 39002466 DOI: 10.1016/j.jiph.2024.102486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 06/17/2024] [Accepted: 06/23/2024] [Indexed: 07/15/2024] Open
Abstract
High mortality has been reported in severe cases of COVID-19. Emerging reports suggested that the severity is not only due to SARS-CoV-2 infection, but also due to coinfections by other pathogens exhibiting symptoms like COVID-19. During the COVID-19 pandemic, simultaneous respiratory coinfections with various viral (Retroviridae, Flaviviridae, Orthomyxoviridae, and Picoviridae) and bacterial (Mycobacteriaceae, Mycoplasmataceae, Enterobacteriaceae and Helicobacteraceae) families have been observed. These pathogens intensify disease severity by potentially augmenting SARSCoV-2 replication, inflammation, and modulation of signaling pathways. Coinfection emerges as a critical determinant of COVID-19 severity, principally instigated by heightened pro-inflammatory cytokine levels, as cytokine storm. Thereby, in co-infection scenario, the severity is also driven by the modulation of inflammatory signaling pathways by both pathogens possibly associated with interleukin, interferon, and cell death exacerbating the severity. In the current review, we attempt to understand the role of co- infections by other pathogens and their involvement in the severity of COVID-19.
Collapse
Affiliation(s)
- Budhadev Baral
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India
| | - Vaishali Saini
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India
| | - Meenakshi Kandpal
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India
| | - Pratik Kundu
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India
| | - Amit Kumar Dixit
- Central Ayurveda Research Institute, 4-CN Block, Sector -V, Bidhannagar, Kolkata 700 091, India
| | - Hamendra Singh Parmar
- School of Biotechnology, Devi Ahilya Vishwavidyalaya, Takshashila Campus, Indore, Madhya Pradesh 452001, India
| | - Ajay Kumar Meena
- Regional Ayurveda Research Institute, Gwalior, Amkhoh, Gwalior, Madhya Pradesh 474001, India
| | - Pankaj Trivedi
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Hem Chandra Jha
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India; Centre for Rural Development and Technology, Indian Institute of Technology Indore, Simrol, Indore 453552, Madhya Pradesh, India.
| |
Collapse
|
24
|
Sibille G, Mannino G, Frasson I, Pavan M, Luganini A, Salata C, Maffei ME, Gribaudo G. The Novel A-Type Proanthocyanidin-Rich Phytocomplex SP4™ Acts as a Broad-Spectrum Antiviral Agent against Human Respiratory Viruses. Int J Mol Sci 2024; 25:7370. [PMID: 39000477 PMCID: PMC11242173 DOI: 10.3390/ijms25137370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
The appearance of new respiratory virus infections in humans with epidemic or pandemic potential has underscored the urgent need for effective broad-spectrum antivirals (BSAs). Bioactive compounds derived from plants may provide a natural source of new BSA candidates. Here, we investigated the novel phytocomplex formulation SP4™ as a candidate direct-acting BSA against major current human respiratory viruses, including coronaviruses and influenza viruses. SP4™ inhibited the in vitro replication of SARS-CoV-2, hCoV-OC43, hCoV-229E, Influenza A and B viruses, and respiratory syncytial virus in the low-microgram range. Using hCoV-OC43 as a representative respiratory virus, most of the antiviral activity of SP4™ was observed to stem primarily from its dimeric A-type proanthocyanidin (PAC-A) component. Further investigations of the mechanistic mode of action showed SP4™ and its PAC-A-rich fraction to prevent hCoV-OC43 from attaching to target cells and exert virucidal activity. This occurred through their interaction with the spike protein of hCoV-OC43 and SARS-CoV-2, thereby interfering with spike functions and leading to the loss of virion infectivity. Overall, these findings support the further development of SP4™ as a candidate BSA of a natural origin for the prevention of human respiratory virus infections.
Collapse
Affiliation(s)
- Giulia Sibille
- Microbiology and Virology Unit, Department of Life Sciences and Systems Biology, University of Torino, Via Accademia Albertina 13, 10123 Turin, Italy; (G.S.); (M.P.); (A.L.)
| | - Giuseppe Mannino
- Plant Physiology Unit, Department of Life Sciences and Systems Biology, University of Torino, Via Quarello 15/a, 10135 Torino, Italy; (G.M.); (M.E.M.)
| | - Ilaria Frasson
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (I.F.); (C.S.)
| | - Marta Pavan
- Microbiology and Virology Unit, Department of Life Sciences and Systems Biology, University of Torino, Via Accademia Albertina 13, 10123 Turin, Italy; (G.S.); (M.P.); (A.L.)
| | - Anna Luganini
- Microbiology and Virology Unit, Department of Life Sciences and Systems Biology, University of Torino, Via Accademia Albertina 13, 10123 Turin, Italy; (G.S.); (M.P.); (A.L.)
| | - Cristiano Salata
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (I.F.); (C.S.)
| | - Massimo E. Maffei
- Plant Physiology Unit, Department of Life Sciences and Systems Biology, University of Torino, Via Quarello 15/a, 10135 Torino, Italy; (G.M.); (M.E.M.)
| | - Giorgio Gribaudo
- Microbiology and Virology Unit, Department of Life Sciences and Systems Biology, University of Torino, Via Accademia Albertina 13, 10123 Turin, Italy; (G.S.); (M.P.); (A.L.)
| |
Collapse
|
25
|
Rizvi ZA, Sadhu S, Dandotiya J, Sharma P, Binayke A, Singh V, Das V, Khatri R, Kumar R, Samal S, Kalia M, Awasthi A. SARS-CoV-2 infection induces thymic atrophy mediated by IFN-γ in hACE2 transgenic mice. Eur J Immunol 2024; 54:e2350624. [PMID: 38655818 DOI: 10.1002/eji.202350624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/26/2024]
Abstract
Pathogenic infections cause thymic atrophy, perturb thymic T-cell development, and alter immunological response. Previous studies reported dysregulated T-cell function and lymphopenia in coronavirus disease-19 (COVID-19). However, immunopathological changes in the thymus associated with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection have not been elucidated. Here, we report that SARS-CoV-2 infects thymocytes, and induces CD4+CD8+ (double positive; DP) T-cell apoptosis leading to thymic atrophy and loss of peripheral TCR repertoire in K18-hACE2 transgenic mice. Infected thymus led to increased CD44+CD25- T-cells, indicating an early arrest in the T-cell maturation pathway. Thymic atrophy was notably higher in male hACE2-Tg mice than in females and involved an upregulated de-novo synthesis pathway of thymic glucocorticoid. Further, IFN-γ was crucial for thymic atrophy, as anti-IFN-γ -antibody neutralization blunted thymic involution. Therapeutic use of Remdesivir also rescued thymic atrophy. While the Omicron variant and its sub-lineage BA.5 variant caused marginal thymic atrophy, the delta variant of SARS-CoV-2 exhibited severe thymic atrophy characterized by severely depleted DP T-cells. Recently characterized broadly SARS-CoV-2 neutralizing monoclonal antibody P4A2 was able to rescue thymic atrophy and restore the thymic maturation pathway of T-cells. Together, we report SARS-CoV-2-associated thymic atrophy resulting from impaired T-cell maturation pathway which may contribute to dyregulated T cell response during COVID-19.
Collapse
Affiliation(s)
- Zaigham Abbas Rizvi
- Immuno-biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Srikanth Sadhu
- Immuno-biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Jyotsna Dandotiya
- Immuno-biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Puja Sharma
- Regional Centre Biotechnology, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Akshay Binayke
- Immuno-biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Virendra Singh
- Immuno-biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Vinayaka Das
- Immuno-biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Ritika Khatri
- Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Rajesh Kumar
- Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Sweety Samal
- Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Manjula Kalia
- Regional Centre Biotechnology, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| | - Amit Awasthi
- Immuno-biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana, India
| |
Collapse
|
26
|
Akalu YT, Patel RS, Taft J, Canas-Arranz R, Richardson A, Buta S, Martin-Fernandez M, Sazeides C, Pearl RL, Mainkar G, Kurland AP, Geltman R, Rosberger H, Kang DD, Kurian AA, Kaur K, Altman J, Dong Y, Johnson JR, Zhangi L, Lim JK, Albrecht RA, García-Sastre A, Rosenberg BR, Bogunovic D. Broad-spectrum RNA antiviral inspired by ISG15 -/- deficiency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600468. [PMID: 38979204 PMCID: PMC11230275 DOI: 10.1101/2024.06.24.600468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Type I interferons (IFN-I) are cytokines with potent antiviral and inflammatory capacities. IFN-I signaling drives the expression of hundreds of IFN-I stimulated genes (ISGs), whose aggregate function results in the control of viral infection. A few of these ISGs are tasked with negatively regulating the IFN-I response to prevent overt inflammation. ISG15 is a negative regulator whose absence leads to persistent, low-grade elevation of ISG expression and concurrent, self-resolving mild autoinflammation. The limited breadth and low-grade persistence of ISGs expressed in ISG15 deficiency are sufficient to confer broad-spectrum antiviral resistance. Inspired by ISG15 deficiency, we have identified a nominal collection of 10 ISGs that recapitulate the broad antiviral potential of the IFN-I system. The expression of the 10 ISG collection in an IFN-I non-responsive cell line increased cellular resistance to Zika, Vesicular Stomatitis, Influenza A (IAV), and SARS-CoV-2 viruses. A deliverable prophylactic formulation of this syndicate of 10 ISGs significantly inhibited IAV PR8 replication in vivo in mice and protected hamsters against a lethal SARS-CoV-2 challenge, suggesting its potential as a broad-spectrum antiviral against many current and future emerging viral pathogens. One-Sentence Summary Human inborn error of immunity-guided discovery and development of a broad-spectrum RNA antiviral therapy.
Collapse
|
27
|
Hodel KVS, Fiuza BSD, Conceição RS, Aleluia ACM, Pitanga TN, Fonseca LMDS, Valente CO, Minafra-Rezende CS, Machado BAS. Pharmacovigilance in Vaccines: Importance, Main Aspects, Perspectives, and Challenges-A Narrative Review. Pharmaceuticals (Basel) 2024; 17:807. [PMID: 38931474 PMCID: PMC11206969 DOI: 10.3390/ph17060807] [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: 04/24/2024] [Revised: 05/29/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Pharmacovigilance plays a central role in safeguarding public health by continuously monitoring the safety of vaccines, being critical in a climate of vaccine hesitancy, where public trust is paramount. Pharmacovigilance strategies employed to gather information on adverse events following immunization (AEFIs) include pre-registration data, media reports, clinical trials, and societal reporting. Early detection of AEFIs during clinical trials is crucial for thorough safety analysis and preventing serious reactions once vaccines are deployed. This review highlights the importance of societal reporting, encompassing contributions from community members, healthcare workers, and pharmaceutical companies. Technological advancements such as quick response (QR) codes can facilitate prompt AEFI reporting. While vaccines are demonstrably safe, the possibility of adverse events necessitates continuous post-marketing surveillance. However, underreporting remains a challenge, underscoring the critical role of public engagement in pharmacovigilance. This narrative review comprehensively examines and synthesizes key aspects of virus vaccine pharmacovigilance, with special considerations for specific population groups. We explore applicable legislation, the spectrum of AEFIs associated with major vaccines, and the unique challenges and perspectives surrounding pharmacovigilance in this domain.
Collapse
Affiliation(s)
- Katharine Valéria Saraiva Hodel
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), SENAI CIMATEC University Center, Salvador 41650-010, Bahia State, Brazil
| | - Bianca Sampaio Dotto Fiuza
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), SENAI CIMATEC University Center, Salvador 41650-010, Bahia State, Brazil
| | - Rodrigo Souza Conceição
- Department of Medicine, College of Pharmacy, Federal University of Bahia, Salvador 40170-115, Bahia State, Brazil
| | - Augusto Cezar Magalhães Aleluia
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), SENAI CIMATEC University Center, Salvador 41650-010, Bahia State, Brazil
- Department of Natural Sciences, Southwestern Bahia State University (UESB), Campus Vitória da Conquista, Vitória da Conquista 45031-300, Bahia State, Brazil
| | - Thassila Nogueira Pitanga
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), SENAI CIMATEC University Center, Salvador 41650-010, Bahia State, Brazil
- Laboratory for Research in Genetics and Translational Hematology, Gonçalo Moniz Institute, FIOCRUZ-BA, Salvador 40296-710, Bahia State, Brazil
| | - Larissa Moraes dos Santos Fonseca
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), SENAI CIMATEC University Center, Salvador 41650-010, Bahia State, Brazil
| | - Camila Oliveira Valente
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), SENAI CIMATEC University Center, Salvador 41650-010, Bahia State, Brazil
| | | | - Bruna Aparecida Souza Machado
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), SENAI CIMATEC University Center, Salvador 41650-010, Bahia State, Brazil
| |
Collapse
|
28
|
Carvajal JJ, García-Castillo V, Cuellar SV, Campillay-Véliz CP, Salazar-Ardiles C, Avellaneda AM, Muñoz CA, Retamal-Díaz A, Bueno SM, González PA, Kalergis AM, Lay MK. New insights into the pathogenesis of SARS-CoV-2 during and after the COVID-19 pandemic. Front Immunol 2024; 15:1363572. [PMID: 38911850 PMCID: PMC11190347 DOI: 10.3389/fimmu.2024.1363572] [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: 12/31/2023] [Accepted: 04/24/2024] [Indexed: 06/25/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the respiratory distress condition known as COVID-19. This disease broadly affects several physiological systems, including the gastrointestinal, renal, and central nervous (CNS) systems, significantly influencing the patient's overall quality of life. Additionally, numerous risk factors have been suggested, including gender, body weight, age, metabolic status, renal health, preexisting cardiomyopathies, and inflammatory conditions. Despite advances in understanding the genome and pathophysiological ramifications of COVID-19, its precise origins remain elusive. SARS-CoV-2 interacts with a receptor-binding domain within angiotensin-converting enzyme 2 (ACE2). This receptor is expressed in various organs of different species, including humans, with different abundance. Although COVID-19 has multiorgan manifestations, the main pathologies occur in the lung, including pulmonary fibrosis, respiratory failure, pulmonary embolism, and secondary bacterial pneumonia. In the post-COVID-19 period, different sequelae may occur, which may have various causes, including the direct action of the virus, alteration of the immune response, and metabolic alterations during infection, among others. Recognizing the serious adverse health effects associated with COVID-19, it becomes imperative to comprehensively elucidate and discuss the existing evidence surrounding this viral infection, including those related to the pathophysiological effects of the disease and the subsequent consequences. This review aims to contribute to a comprehensive understanding of the impact of COVID-19 and its long-term effects on human health.
Collapse
Affiliation(s)
- Jonatan J. Carvajal
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
| | - Valeria García-Castillo
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
| | - Shelsy V. Cuellar
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
| | | | - Camila Salazar-Ardiles
- Center for Research in Physiology and Altitude Medicine (FIMEDALT), Biomedical Department, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - Andrea M. Avellaneda
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
- Department of Basic Sciences, Faculty of Sciences, Universidad Santo Tomás, Antofagasta, Chile
| | - Christian A. Muñoz
- Research Center in Immunology and Biomedical Biotechnology of Antofagasta (CIIBBA), University of Antofagasta, Antofagasta, Chile
- Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
- Millennium Institute on Immunology and Immunotherapy, Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - Angello Retamal-Díaz
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
- Research Center in Immunology and Biomedical Biotechnology of Antofagasta (CIIBBA), University of Antofagasta, Antofagasta, Chile
- Millennium Institute on Immunology and Immunotherapy, Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Margarita K. Lay
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
- Research Center in Immunology and Biomedical Biotechnology of Antofagasta (CIIBBA), University of Antofagasta, Antofagasta, Chile
- Millennium Institute on Immunology and Immunotherapy, Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| |
Collapse
|
29
|
da Cunha Lima EA, Toledo LV, Correia MDL, de Almeida Pereira D, Caetano RO, Faria TB, de Castro Moura C, Krempser P, Braga LM. Effect of a Non-pharmacological Intervention on Vaccine-related Pain: Randomized Clinical Trial. Pain Manag Nurs 2024:S1524-9042(24)00154-1. [PMID: 38772758 DOI: 10.1016/j.pmn.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 04/03/2024] [Accepted: 04/07/2024] [Indexed: 05/23/2024]
Abstract
BACKGROUND Acute pain during vaccine administration is an expected event; however, some strategies, such as the use of high frequency vibration devices associated with cryotherapy, may minimize it. AIM Evaluate the effect of high frequency vibration associated with cryotherapy on the level of pain related to the administration of influenza vaccine by intramuscular route in adults. METHOD A randomized clinical trial was conducted with 350 adults who received the influenza vaccine. Participants allocated to the intervention group used a high-frequency vibration device associated with cryotherapy during vaccination, and those in the control group administered the vaccine according to the service routine. RESULTS Mean self-reported pain after vaccination of participants who used the device was lower (1.5 ±1.7) when compared to those who did not use it (1.9 ±1.9) (p = .041). There was an association of higher levels of pain after vaccination with participants in the control group (p = .011), females (p = .042), with higher level of pain expected with vaccination (p < .001) and higher level of anxiety before (p < .001) and after vaccination (p = .001). CONCLUSIONS The use of high frequency vibration associated with cryotherapy has been shown to be a viable non-pharmacological intervention for the reduction of pain associated with influenza vaccination in adults.
Collapse
Affiliation(s)
| | - Luana Vieira Toledo
- Department of Medicine and Nursing. Federal University of Viçosa, Viçosa, Brazil
| | | | | | | | | | | | - Paula Krempser
- Child Department and Public Health, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Luciene Muniz Braga
- Department of Medicine and Nursing. Federal University of Viçosa, Viçosa, Brazil
| |
Collapse
|
30
|
Sen P, Zhang Z, Sakib S, Gu J, Li W, Adhikari BR, Motsenyat A, L'Heureux-Hache J, Ang JC, Panesar G, Salena BJ, Yamamura D, Miller MS, Li Y, Soleymani L. High-Precision Viral Detection Using Electrochemical Kinetic Profiling of Aptamer-Antigen Recognition in Clinical Samples and Machine Learning. Angew Chem Int Ed Engl 2024; 63:e202400413. [PMID: 38458987 DOI: 10.1002/anie.202400413] [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: 01/07/2024] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 03/10/2024]
Abstract
High-precision viral detection at point of need with clinical samples plays a pivotal role in the diagnosis of infectious diseases and the control of a global pandemic. However, the complexity of clinical samples that often contain very low viral concentrations makes it a huge challenge to develop simple diagnostic devices that do not require any sample processing and yet are capable of meeting performance metrics such as very high sensitivity and specificity. Herein we describe a new single-pot and single-step electrochemical method that uses real-time kinetic profiling of the interaction between a high-affinity aptamer and an antigen on a viral surface. This method generates many data points per sample, which when combined with machine learning, can deliver highly accurate test results in a short testing time. We demonstrate this concept using both SARS-CoV-2 and Influenza A viruses as model viruses with specifically engineered high-affinity aptamers. Utilizing this technique to diagnose COVID-19 with 37 real human saliva samples results in a sensitivity and specificity of both 100 % (27 true negatives and 10 true positives, with 0 false negative and 0 false positive), which showcases the superb diagnostic precision of this method.
Collapse
Affiliation(s)
- Payel Sen
- Department of Engineering Physics, McMaster University, Canada
| | - Zijie Zhang
- Department of Biochemistry and Biomedical Sciences, McMaster University, Canada
| | - Sadman Sakib
- Department of Engineering Physics, McMaster University, Canada
| | - Jimmy Gu
- Department of Biochemistry and Biomedical Sciences, McMaster University, Canada
| | - Wantong Li
- Department of Engineering Physics, McMaster University, Canada
| | | | - Ariel Motsenyat
- Department of Integrated Biomedical Engineering and Health Sciences, McMaster University, Canada
| | | | - Jann C Ang
- Department of Biochemistry and Biomedical Sciences, McMaster University, Canada
- McMaster Immunology Research Centre, McMaster University, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Canada
| | - Gurpreet Panesar
- Department of Biochemistry and Biomedical Sciences, McMaster University, Canada
| | | | - Debora Yamamura
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Canada
| | - Matthew S Miller
- Department of Biochemistry and Biomedical Sciences, McMaster University, Canada
- McMaster Immunology Research Centre, McMaster University, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Canada
| | - Yingfu Li
- Department of Biochemistry and Biomedical Sciences, McMaster University, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Canada
- School of Biomedical Engineering, McMaster University, Canada
| | - Leyla Soleymani
- Department of Engineering Physics, McMaster University, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Canada
- School of Biomedical Engineering, McMaster University, Canada
| |
Collapse
|
31
|
Swart IC, Van Gelder W, De Haan CAM, Bosch BJ, Oliveira S. Next generation single-domain antibodies against respiratory zoonotic RNA viruses. Front Mol Biosci 2024; 11:1389548. [PMID: 38784667 PMCID: PMC11111979 DOI: 10.3389/fmolb.2024.1389548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
The global impact of zoonotic viral outbreaks underscores the pressing need for innovative antiviral strategies, particularly against respiratory zoonotic RNA viruses. These viruses possess a high potential to trigger future epidemics and pandemics due to their high mutation rate, broad host range and efficient spread through airborne transmission. Recent pandemics caused by coronaviruses and influenza A viruses underscore the importance of developing targeted antiviral strategies. Single-domain antibodies (sdAbs), originating from camelids, also known as nanobodies or VHHs (Variable Heavy domain of Heavy chain antibodies), have emerged as promising tools to combat current and impending zoonotic viral threats. Their unique structure, coupled with attributes like robustness, compact size, and cost-effectiveness, positions them as strong alternatives to traditional monoclonal antibodies. This review describes the pivotal role of sdAbs in combating respiratory zoonotic viruses, with a primary focus on enhancing sdAb antiviral potency through optimization techniques and diverse administration strategies. We discuss both the promises and challenges within this dynamically growing field.
Collapse
Affiliation(s)
- Iris C. Swart
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Virology Section, Infectious Diseases and Immunology Division, Department Biomolecular Health Sciences, Faculty Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Willem Van Gelder
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Cornelis A. M. De Haan
- Virology Section, Infectious Diseases and Immunology Division, Department Biomolecular Health Sciences, Faculty Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Berend-Jan Bosch
- Virology Section, Infectious Diseases and Immunology Division, Department Biomolecular Health Sciences, Faculty Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Sabrina Oliveira
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Pharmaceutics, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
32
|
Kim MJ, Haizan I, Ahn MJ, Park DH, Choi JH. Recent Advances in Lateral Flow Assays for Viral Protein Detection with Nanomaterial-Based Optical Sensors. BIOSENSORS 2024; 14:197. [PMID: 38667190 PMCID: PMC11048458 DOI: 10.3390/bios14040197] [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: 03/15/2024] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024]
Abstract
Controlling the progression of contagious diseases is crucial for public health management, emphasizing the importance of early viral infection diagnosis. In response, lateral flow assays (LFAs) have been successfully utilized in point-of-care (POC) testing, emerging as a viable alternative to more traditional diagnostic methods. Recent advancements in virus detection have primarily leveraged methods such as reverse transcription-polymerase chain reaction (RT-PCR), reverse transcription-loop-mediated isothermal amplification (RT-LAMP), and the enzyme-linked immunosorbent assay (ELISA). Despite their proven effectiveness, these conventional techniques are often expensive, require specialized expertise, and consume a significant amount of time. In contrast, LFAs utilize nanomaterial-based optical sensing technologies, including colorimetric, fluorescence, and surface-enhanced Raman scattering (SERS), offering quick, straightforward analyses with minimal training and infrastructure requirements for detecting viral proteins in biological samples. This review describes the composition and mechanism of and recent advancements in LFAs for viral protein detection, categorizing them into colorimetric, fluorescent, and SERS-based techniques. Despite significant progress, developing a simple, stable, highly sensitive, and selective LFA system remains a formidable challenge. Nevertheless, an advanced LFA system promises not only to enhance clinical diagnostics but also to extend its utility to environmental monitoring and beyond, demonstrating its potential to revolutionize both healthcare and environmental safety.
Collapse
Affiliation(s)
- Min Jung Kim
- School of Chemical Engineering, Clean Energy Research Center, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si 54896, Jeollabuk-do, Republic of Korea; (M.J.K.); (D.-H.P.)
| | - Izzati Haizan
- Department of Bioprocess Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si 54896, Jeollabuk-do, Republic of Korea;
| | - Min Ju Ahn
- Department of Biotechnology, Jeonbuk National University, 79 Gobongro, Iksan-si 54596, Jeollabuk-do, Republic of Korea;
| | - Dong-Hyeok Park
- School of Chemical Engineering, Clean Energy Research Center, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si 54896, Jeollabuk-do, Republic of Korea; (M.J.K.); (D.-H.P.)
| | - Jin-Ha Choi
- School of Chemical Engineering, Clean Energy Research Center, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si 54896, Jeollabuk-do, Republic of Korea; (M.J.K.); (D.-H.P.)
- Department of Bioprocess Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si 54896, Jeollabuk-do, Republic of Korea;
| |
Collapse
|
33
|
Sim JK, Lee HS, Yang J, Gwack J, Kim BI, Cha JO, Min KH, Lee YS. Comparative Analysis of Clinical Outcomes Using Propensity Score Matching: Coronavirus Disease 2019 vs. Seasonal Influenza in Korea. J Korean Med Sci 2024; 39:e128. [PMID: 38622937 PMCID: PMC11018986 DOI: 10.3346/jkms.2024.39.e128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 03/18/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND The advent of the omicron variant and the formulation of diverse therapeutic strategies marked a new epoch in the realm of coronavirus disease 2019 (COVID-19). Studies have compared the clinical outcomes between COVID-19 and seasonal influenza, but such studies were conducted during the early stages of the pandemic when effective treatment strategies had not yet been developed, which limits the generalizability of the findings. Therefore, an updated evaluation of the comparative analysis of clinical outcomes between COVID-19 and seasonal influenza is requisite. METHODS This study used data from the severe acute respiratory infection surveillance system of South Korea. We extracted data for influenza patients who were infected between 2018 and 2019 and COVID-19 patients who were infected in 2021 (pre-omicron period) and 2022 (omicron period). Comparisons of outcomes were conducted among the pre-omicron, omicron, and influenza cohorts utilizing propensity score matching. The adjusted covariates in the propensity score matching included age, sex, smoking, and comorbidities. RESULTS The study incorporated 1,227 patients in the pre-omicron cohort, 1,948 patients in the omicron cohort, and 920 patients in the influenza cohort. Following propensity score matching, 491 patients were included in each respective group. Clinical presentations exhibited similarities between the pre-omicron and omicron cohorts; however, COVID-19 patients demonstrated a higher prevalence of dyspnea and pulmonary infiltrates compared to their influenza counterparts. Both COVID-19 groups exhibited higher in-hospital mortality and longer hospital length of stay than the influenza group. The omicron group showed no significant improvement in clinical outcomes compared to the pre-omicron group. CONCLUSION The omicron group did not demonstrate better clinical outcomes than the pre-omicron group, and exhibited significant disease severity compared to the influenza group. Considering the likely persistence of COVID-19 infections, it is imperative to sustain comprehensive studies and ongoing policy support for the virus to enhance the prognosis for individuals affected by COVID-19.
Collapse
Affiliation(s)
- Jae Kyeom Sim
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Hye Sun Lee
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, Korea
| | - Juyeon Yang
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, Korea
| | - Jin Gwack
- Division of Infectious Disease Control, Bureau of Infectious Disease Policy, Korea Disease Control and Prevention Agency (KDCA), Cheongju, Korea
| | - Bryan Inho Kim
- Division of Infectious Disease Control, Bureau of Infectious Disease Policy, Korea Disease Control and Prevention Agency (KDCA), Cheongju, Korea
| | - Jeong-Ok Cha
- Division of Infectious Disease Control, Bureau of Infectious Disease Policy, Korea Disease Control and Prevention Agency (KDCA), Cheongju, Korea
| | - Kyung Hoon Min
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Young Seok Lee
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Korea University Guro Hospital, Seoul, Korea.
| |
Collapse
|
34
|
Zeng Y, Gan X, Xu Z, Hu X, Hu C, Ma H, Tu H, Chai B, Yang C, Hu S, Chai Y. AIEgens-enhanced rapid sensitive immunofluorescent assay for SARS-CoV-2 with digital microfluidics. Anal Chim Acta 2024; 1298:342398. [PMID: 38462346 DOI: 10.1016/j.aca.2024.342398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 02/14/2024] [Accepted: 02/20/2024] [Indexed: 03/12/2024]
Abstract
BACKGROUND Sensitive and rapid antigen detection is critical for the diagnosis and treatment of infectious diseases, but conventional ELISAs including chemiluminescence-based assays are limited in sensitivity and require many operation steps. Fluorescence immunoassays are fast and convenient but often show limited sensitivity and dynamic range. RESULTS To address the need, an aggregation-induced emission fluorgens (AIEgens) enhanced immunofluorescent assay with beads-based quantification on the digital microfluidic (DMF) platform was developed. Portable DMF devices and chips with small electrodes were fabricated, capable of manipulating droplets within 100 nL and boosting the reaction efficiency. AIEgen nanoparticles (NPs) with high fluorescence and photostability were synthesized to enhance the test sensitivity and detection range. The integration of AIEgen probes, transparent DMF chip design, and the large magnetic beads (10 μm) as capture agents enabled rapid and direct image-taking and signal calculation of the test result. The performance of this platform was demonstrated by point-of-care quantification of SARS-CoV-2 nucleocapsid (N) protein. Within 25 min, a limit of detection of 5.08 pg mL-1 and a limit of quantification of 8.91 pg mL-1 can be achieved using <1 μL sample. The system showed high reproducibility across the wide dynamic range (10-105 pg mL-1), with the coefficient of variance ranging from 2.6% to 9.8%. SIGNIFICANCE This rapid, sensitive AIEgens-enhanced immunofluorescent assay on the DMF platform showed simplified reaction steps and improved performance, providing insight into the small-volume point-of-care testing of different biomarkers in research and clinical applications.
Collapse
Affiliation(s)
- Yuping Zeng
- Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, China.
| | - Xiangyu Gan
- Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, China.
| | - Zhourui Xu
- Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, China.
| | - Xiaoxiang Hu
- Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, China.
| | - Chenxuan Hu
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China.
| | - Hanbin Ma
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China; Guangdong ACXEL Micro & Nano Tech Co., Ltd, Foshan, Guangdong province, China.
| | - Hangjia Tu
- Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, China.
| | - Bao Chai
- Department of Dermatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China; Department of Dermatology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518052, China.
| | - Chengbin Yang
- Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, China.
| | - Siyi Hu
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China.
| | - Yujuan Chai
- Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, China.
| |
Collapse
|
35
|
Luong QXT, Hoang PT, Lee Y, Ayun RQ, Na K, Park S, Lin C, Ho PT, Lee TK, Lee S. An RNA-hydrolyzing recombinant minibody prevents both influenza A virus and coronavirus in co-infection models. Sci Rep 2024; 14:8472. [PMID: 38605110 PMCID: PMC11009316 DOI: 10.1038/s41598-024-52810-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 01/23/2024] [Indexed: 04/13/2024] Open
Abstract
With the lifting of COVID-19 non-pharmaceutical interventions, the resurgence of common viral respiratory infections was recorded in several countries worldwide. It facilitates viral co-infection, further burdens the already over-stretched healthcare systems. Racing to find co-infection-associated efficacy therapeutic agents need to be rapidly established. However, it has encountered numerous challenges that necessitate careful investigation. Here, we introduce a potential recombinant minibody-associated treatment, 3D8 single chain variable fragment (scFv), which has been developed as a broad-spectrum antiviral drug that acts via its nucleic acid catalytic and cell penetration abilities. In this research, we demonstrated that 3D8 scFv exerted antiviral activity simultaneously against both influenza A viruses (IAVs) and coronaviruses in three established co-infection models comprising two types of coronaviruses [beta coronavirus-human coronavirus OC43 (hCoV-OC43) and alpha coronavirus-porcine epidemic diarrhea virus (PEDV)] in Vero E6 cells, two IAVs [A/Puerto Rico/8/1934 H1N1 (H1N1/PR8) and A/X-31 (H3N2/X-31)] in MDCK cells, and a combination of coronavirus and IAV (hCoV-OC43 and adapted-H1N1) in Vero E6 cells by a statistically significant reduction in viral gene expression, proteins level, and approximately around 85%, 65%, and 80% of the progeny of 'hCoV-OC43-PEDV', 'H1N1/PR8-H3N2/X-31', and 'hCoV-OC43-adapted-H1N1', respectively, were decimated in the presence of 3D8 scFv. Taken together, we propose that 3D8 scFv is a promising broad-spectrum drug for treatment against RNA viruses in co-infection.
Collapse
Affiliation(s)
- Quynh Xuan Thi Luong
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Korea
| | - Phuong Thi Hoang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Korea
| | - Yongjun Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Korea
| | | | - Kyungho Na
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Korea
| | - Seonhyeon Park
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Korea
| | - Chengmin Lin
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Korea
| | - Phuong Thi Ho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Korea
| | - Taek-Kyun Lee
- Ecological Risk Research Department, Korea Institute of Ocean Science & Technology, Geoje, 53201, Korea.
| | - Sukchan Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Korea.
| |
Collapse
|
36
|
Raya S, Malla B, Shrestha S, Sthapit N, Kattel H, Sharma ST, Tuladhar R, Maharjan R, Takeda T, Kitajima M, Tandukar S, Haramoto E. Quantification of multiple respiratory viruses in wastewater in the Kathmandu Valley, Nepal: Potential implications of wastewater-based epidemiology for community disease surveillance in developing countries. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 920:170845. [PMID: 38340866 DOI: 10.1016/j.scitotenv.2024.170845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/04/2024] [Accepted: 02/07/2024] [Indexed: 02/12/2024]
Abstract
Despite being the major cause of death, clinical surveillance of respiratory viruses at the community level is very passive, especially in developing countries. This study focused on the surveillance of three respiratory viruses [severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), influenza A virus (IFV-A), and respiratory syncytial virus (RSV)] in the Kathmandu Valley, Nepal, by implication of wastewater-based epidemiology (WBE). Fifty-one untreated wastewater samples were from two wastewater treatment plants (WWTPs) between April and October 2022. Among eight combinations of the pre-evaluated methods, the combination of concentration by simple centrifugation, pretreatment by DNA/RNA Shield (Zymo Research), and extraction by the QIAamp Viral RNA Mini Kit (QIAGEN) showed the best performance for detecting respiratory viruses. Using this method with a one-step reverse transcription-quantitative polymerase chain reaction (RT-qPCR), SARS-CoV-2 RNA was successfully detected from both WWTPs (positive ratio, 100 % and 81 %) at concentrations of 5.6 ± 0.6 log10 copies/L from each WWTP. Forty-six SARS-CoV-2 RNA-positive samples were further tested for three mutation site-specific one-step RT-qPCR (L452R, T478K, and E484A/G339D), where G339D/E484A mutations were frequently detected in both WWTPs (96 %). IFV-A RNA was more frequently detected in WWTP A (84 %) compared to WWTP B (38 %). RSV RNA was also detected in both WWTPs (28 % and 8 %, respectively). This is the first study on detecting IFV-A and RSV in wastewater in Nepal, showing the applicability and importance of WBE for respiratory viruses in developing countries where clinical data are lacking.
Collapse
Affiliation(s)
- Sunayana Raya
- Department of Engineering, University of Yamanashi, 4-3-11 Takeda, Kofu, Yamanashi 400-8511, Japan
| | - Bikash Malla
- Interdisciplinary Center for River Basin Environment, University of Yamanashi, 4-3-11 Takeda, Kofu, Yamanashi 400-8511, Japan
| | - Sadhana Shrestha
- Interdisciplinary Center for River Basin Environment, University of Yamanashi, 4-3-11 Takeda, Kofu, Yamanashi 400-8511, Japan
| | - Niva Sthapit
- Interdisciplinary Center for River Basin Environment, University of Yamanashi, 4-3-11 Takeda, Kofu, Yamanashi 400-8511, Japan
| | - Hari Kattel
- Department of Microbiology, Tribhuvan University Teaching Hospital, Kathmandu, Nepal
| | - Sangita Tara Sharma
- Department of Microbiology, Tribhuvan University Teaching Hospital, Kathmandu, Nepal
| | - Reshma Tuladhar
- Department of Microbiology, Tribhuvan University, Kathmandu, Nepal
| | - Rabin Maharjan
- Department of Civil Engineering, Institute of Engineering, Tribhuvan University, Lalitpur, Nepal
| | - Tomoko Takeda
- Department of Earth and Planetary Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| | - Masaaki Kitajima
- Division of Environmental Engineering, Hokkaido University, North 13 West 8, Kita-ku, Sapporo, Hokkaido 060-8628, Japan.
| | | | - Eiji Haramoto
- Interdisciplinary Center for River Basin Environment, University of Yamanashi, 4-3-11 Takeda, Kofu, Yamanashi 400-8511, Japan.
| |
Collapse
|
37
|
Arunachalam AB. Vaccines Induce Homeostatic Immunity, Generating Several Secondary Benefits. Vaccines (Basel) 2024; 12:396. [PMID: 38675778 PMCID: PMC11053716 DOI: 10.3390/vaccines12040396] [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: 02/27/2024] [Revised: 03/28/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
The optimal immune response eliminates invading pathogens, restoring immune equilibrium without inflicting undue harm to the host. However, when a cascade of immunological reactions is triggered, the immune response can sometimes go into overdrive, potentially leading to harmful long-term effects or even death. The immune system is triggered mostly by infections, allergens, or medical interventions such as vaccination. This review examines how these immune triggers differ and why certain infections may dysregulate immune homeostasis, leading to inflammatory or allergic pathology and exacerbation of pre-existing conditions. However, many vaccines generate an optimal immune response and protect against the consequences of pathogen-induced immunological aggressiveness, and from a small number of unrelated pathogens and autoimmune diseases. Here, we propose an "immuno-wave" model describing a vaccine-induced "Goldilocks immunity", which leaves fine imprints of both pro-inflammatory and anti-inflammatory milieus, derived from both the innate and the adaptive arms of the immune system, in the body. The resulting balanced, 'quiet alert' state of the immune system may provide a jump-start in the defense against pathogens and any associated pathological inflammatory or allergic responses, allowing vaccines to go above and beyond their call of duty. In closing, we recommend formally investigating and reaping many of the secondary benefits of vaccines with appropriate clinical studies.
Collapse
Affiliation(s)
- Arun B Arunachalam
- Analytical Sciences, R&D Sanofi Vaccines, 1 Discovery Dr., Swiftwater, PA 18370, USA
| |
Collapse
|
38
|
Gupta N, Silke J. Blocking cell death limits lung damage and inflammation from influenza. Nature 2024; 628:726-727. [PMID: 38600194 DOI: 10.1038/d41586-024-00910-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
|
39
|
Cilloniz C, Dy-Agra G, Pagcatipunan RS, Torres A. Viral Pneumonia: From Influenza to COVID-19. Semin Respir Crit Care Med 2024; 45:207-224. [PMID: 38228165 DOI: 10.1055/s-0043-1777796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Respiratory viruses are increasingly recognized as a cause of community-acquired pneumonia (CAP). The implementation of new diagnostic technologies has facilitated their identification, especially in vulnerable population such as immunocompromised and elderly patients and those with severe cases of pneumonia. In terms of severity and outcomes, viral pneumonia caused by influenza viruses appears similar to that caused by non-influenza viruses. Although several respiratory viruses may cause CAP, antiviral therapy is available only in cases of CAP caused by influenza virus or respiratory syncytial virus. Currently, evidence-based supportive care is key to managing severe viral pneumonia. We discuss the evidence surrounding epidemiology, diagnosis, management, treatment, and prevention of viral pneumonia.
Collapse
Affiliation(s)
- Catia Cilloniz
- Hospital Clinic of Barcelona, IDIBAPS, CIBERESA, Barcelona, Spain
- Faculty of Health Sciences, Continental University, Huancayo, Peru
| | - Guinevere Dy-Agra
- Institute of Pulmonary Medicine, St Luke's Medical Center-Global City, Taguig, Metro Manila, Philippines
| | - Rodolfo S Pagcatipunan
- Institute of Pulmonary Medicine, St Luke's Medical Center-Global City, Taguig, Metro Manila, Philippines
| | - Antoni Torres
- Hospital Clinic of Barcelona, IDIBAPS, CIBERESA, Barcelona, Spain
- School of Medicine, University of Barcelona, Barcelona, Spain
| |
Collapse
|
40
|
Li H, Khoa ND, Kuga K, Ito K. In silico identification of viral loads in cough-generated droplets - Seamless integrated analysis of CFPD-HCD-EWF. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 246:108073. [PMID: 38341896 DOI: 10.1016/j.cmpb.2024.108073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/19/2024] [Accepted: 02/07/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND AND OBJECTIVE Respiratory diseases caused by respiratory viruses have significantly threatened public health worldwide. This study presents a comprehensive approach to predict viral dynamics and the generation of stripped droplets within the mucus layer of the respiratory tract during coughing using a larynx-trachea-bifurcation (LTB) model. METHODS This study integrates computational fluid-particle dynamics (CFPD), host-cell dynamics (HCD), and the Eulerian wall film (EWF) model to propose a potential means for seamless integrated analysis. The verified CFPD-HCD coupling model based on a 3D-shell model was used to characterize the severe acute respiratory syndrome, coronavirus 2 (SARS-CoV-2) dynamics in the LTB mucus layer, whereas the EWF model was employed to account for the interfacial fluid to explore the generation mechanism and trace the origin site of droplets exhaled during a coughing event of an infected host. RESULTS The results obtained using CFPD delineated the preferential deposition sites for droplets in the laryngeal and tracheal regions. Thus, the analysis of the HCD model showed that the viral load increased rapidly in the laryngeal region during the peak of infection, whereas there was a growth delay in the tracheal region (up to day 8 after infection). After two weeks of infection, the high viral load gradually migrated towards the glottic region. Interestingly, the EWF model demonstrated a high concentration of exhaled droplets originating from the larynx. The coupling technique indicated a concurrent high viral load in the mucus layer and site of origin of the exhaled droplets. CONCLUSIONS This interdisciplinary research underscores the seamless analysis from initial exposure to virus-laden droplets, the dynamics of viral infection in the LTB mucus layer, and the re-emission from the coughing activities of an infected host. Our efforts aimed to address the complex challenges at the intersection of viral dynamics and respiratory health, which can contribute to a more detailed understanding and targeted prevention of respiratory diseases.
Collapse
Affiliation(s)
- Hanyu Li
- Faculty of Engineering Sciences, Kyushu University, 6-1 Kasuga-koen, Kasuga, Fukuoka 816-8580, Japan
| | - Nguyen Dang Khoa
- Interdisciplinary Graduate School of Engineering Sciences, Kyushu University, Japan.
| | - Kazuki Kuga
- Faculty of Engineering Sciences, Kyushu University, 6-1 Kasuga-koen, Kasuga, Fukuoka 816-8580, Japan
| | - Kazuhide Ito
- Faculty of Engineering Sciences, Kyushu University, 6-1 Kasuga-koen, Kasuga, Fukuoka 816-8580, Japan
| |
Collapse
|
41
|
Gautam A, Boyd DF, Nikhar S, Zhang T, Siokas I, Van de Velde LA, Gaevert J, Meliopoulos V, Thapa B, Rodriguez DA, Cai KQ, Yin C, Schnepf D, Beer J, DeAntoneo C, Williams RM, Shubina M, Livingston B, Zhang D, Andrake MD, Lee S, Boda R, Duddupudi AL, Crawford JC, Vogel P, Loch C, Schwemmle M, Fritz LC, Schultz-Cherry S, Green DR, Cuny GD, Thomas PG, Degterev A, Balachandran S. Necroptosis blockade prevents lung injury in severe influenza. Nature 2024; 628:835-843. [PMID: 38600381 PMCID: PMC11151938 DOI: 10.1038/s41586-024-07265-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/01/2024] [Indexed: 04/12/2024]
Abstract
Severe influenza A virus (IAV) infections can result in hyper-inflammation, lung injury and acute respiratory distress syndrome1-5 (ARDS), for which there are no effective pharmacological therapies. Necroptosis is an attractive entry point for therapeutic intervention in ARDS and related inflammatory conditions because it drives pathogenic lung inflammation and lethality during severe IAV infection6-8 and can potentially be targeted by receptor interacting protein kinase 3 (RIPK3) inhibitors. Here we show that a newly developed RIPK3 inhibitor, UH15-38, potently and selectively blocked IAV-triggered necroptosis in alveolar epithelial cells in vivo. UH15-38 ameliorated lung inflammation and prevented mortality following infection with laboratory-adapted and pandemic strains of IAV, without compromising antiviral adaptive immune responses or impeding viral clearance. UH15-38 displayed robust therapeutic efficacy even when administered late in the course of infection, suggesting that RIPK3 blockade may provide clinical benefit in patients with IAV-driven ARDS and other hyper-inflammatory pathologies.
Collapse
Affiliation(s)
- Avishekh Gautam
- Center for Immunology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - David F Boyd
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
- Department of Host-Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA, USA
| | - Sameer Nikhar
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, USA
| | - Ting Zhang
- Center for Immunology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Ioannis Siokas
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
| | - Lee-Ann Van de Velde
- Department of Host-Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Jessica Gaevert
- Department of Host-Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Victoria Meliopoulos
- Department of Host-Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Bikash Thapa
- Center for Immunology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Diego A Rodriguez
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Kathy Q Cai
- Center for Immunology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Chaoran Yin
- Center for Immunology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Daniel Schnepf
- Institute of Virology Department for Medical Microbiology and Hygiene, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julius Beer
- Institute of Virology Department for Medical Microbiology and Hygiene, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Carly DeAntoneo
- Center for Immunology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Riley M Williams
- Center for Immunology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Maria Shubina
- Center for Immunology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Brandi Livingston
- Department of Host-Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Dingqiang Zhang
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
| | - Mark D Andrake
- Center for Immunology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Seungheon Lee
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, USA
| | - Raghavender Boda
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, USA
| | - Anantha L Duddupudi
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, USA
| | - Jeremy Chase Crawford
- Department of Host-Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Peter Vogel
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Martin Schwemmle
- Institute of Virology Department for Medical Microbiology and Hygiene, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Stacey Schultz-Cherry
- Department of Host-Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Douglas R Green
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Gregory D Cuny
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, USA.
| | - Paul G Thomas
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA.
- Department of Host-Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA.
| | - Alexei Degterev
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA.
| | | |
Collapse
|
42
|
Wang T, Wang Y, Yao Y. Gut microbe guides alveolar macrophages to fight flu. Cell Host Microbe 2024; 32:296-298. [PMID: 38484707 DOI: 10.1016/j.chom.2024.02.007] [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: 02/08/2024] [Revised: 02/10/2024] [Accepted: 02/12/2024] [Indexed: 03/19/2024]
Abstract
The intestinal microbiota is associated with defense against respiratory viral infections. In this issue of Cell Host & Microbe, Ngo and colleagues1 show that intestinal commensal segmented filamentous bacteria reprogram alveolar macrophages with improved influenza-viral-neutralizing and phagocytic functions while maintaining inflammatory anergy to better protect the lung.
Collapse
Affiliation(s)
- Tao Wang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Yanling Wang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Yushi Yao
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; Liangzhu Laboratory, Hangzhou, Zhejiang 310023, China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang 310003, China.
| |
Collapse
|
43
|
Zheng Y, Li Y, Li M, Wang R, Jiang Y, Zhao M, Lu J, Li R, Li X, Shi S. COVID-19 cooling: Nanostrategies targeting cytokine storm for controlling severe and critical symptoms. Med Res Rev 2024; 44:738-811. [PMID: 37990647 DOI: 10.1002/med.21997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 08/16/2023] [Accepted: 10/29/2023] [Indexed: 11/23/2023]
Abstract
As severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants continue to wreak havoc worldwide, the "Cytokine Storm" (CS, also known as the inflammatory storm) or Cytokine Release Syndrome has reemerged in the public consciousness. CS is a significant contributor to the deterioration of infected individuals. Therefore, CS control is of great significance for the treatment of critically ill patients and the reduction of mortality rates. With the occurrence of variants, concerns regarding the efficacy of vaccines and antiviral drugs with a broad spectrum have grown. We should make an effort to modernize treatment strategies to address the challenges posed by mutations. Thus, in addition to the requirement for additional clinical data to monitor the long-term effects of vaccines and broad-spectrum antiviral drugs, we can use CS as an entry point and therapeutic target to alleviate the severity of the disease in patients. To effectively combat the mutation, new technologies for neutralizing or controlling CS must be developed. In recent years, nanotechnology has been widely applied in the biomedical field, opening up a plethora of opportunities for CS. Here, we put forward the view of cytokine storm as a therapeutic target can be used to treat critically ill patients by expounding the relationship between coronavirus disease 2019 (COVID-19) and CS and the mechanisms associated with CS. We pay special attention to the representative strategies of nanomaterials in current neutral and CS research, as well as their potential chemical design and principles. We hope that the nanostrategies described in this review provide attractive treatment options for severe and critical COVID-19 caused by CS.
Collapse
Affiliation(s)
- Yu Zheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuke Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mao Li
- Health Management Centre, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, China
| | - Rujing Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuhong Jiang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Mengnan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rui Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sanjun Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
44
|
MILANO GIOVANNA, MARCHI SERENA, VICENTI ILARIA, BIBA CAMILLA, FIASCHI LIA, TROMBETTA CLAUDIAMARIA, LAZZERI GIACOMO, MONTOMOLI EMANUELE, MANINI ILARIA. SARS-CoV-2 and influenza virus coinfections in the Tuscan population during the 2021/2022 influenza season. JOURNAL OF PREVENTIVE MEDICINE AND HYGIENE 2024; 65:E11-E16. [PMID: 38706768 PMCID: PMC11066830 DOI: 10.15167/2421-4248/jpmh2024.65.1.3179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/13/2024] [Indexed: 05/07/2024]
Abstract
Introduction The 2021/2022 influenza season was not characterised by a well-defined incidence peak. As reported by the Italian National Institute of Health, a high value of incidence of influenza cases was recorded in week 13, but it was still lower than in other influenza seasons. This abnormal circulation was probably due to relaxation of the COVID-19 pandemic restriction measures, such as social distancing, smart-working, home leaning and the use of masks, which greatly reduced the circulation of respiratory-transmitted viruses, including human respiratory syncytial virus (HRSV). The symptoms of SARS-CoV-2 and influenza are quite similar, sharing the human-to-human transmission route via respiratory droplets. Methods The aim of this study was to estimate the rate of coinfection with influenza viruses and/or HRSV in SARS-CoV-2-positive subjects (N = 940) in a population of central Italy during the 2021/2022 season. Results A total of 54 cases of coinfection were detected during the study period, 51 cases (5.4%) of SARS-CoV-2 and influenza virus and three cases (0.3%) of SARS-CoV-2 and HRSV coinfection. Conclusions These results highlight the importance of continuous monitoring of the circulation of influenza virus and other respiratory viruses in the context of the COVID-19 pandemic.
Collapse
Affiliation(s)
- GIOVANNA MILANO
- Department of Life Science, University of Siena, Siena, Italy
| | - SERENA MARCHI
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - ILARIA VICENTI
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - CAMILLA BIBA
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - LIA FIASCHI
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - CLAUDIA MARIA TROMBETTA
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- Interuniversity Research Center on Influenza and other Transmissible Infections
| | - GIACOMO LAZZERI
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- Interuniversity Research Center on Influenza and other Transmissible Infections
| | - EMANUELE MONTOMOLI
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- Interuniversity Research Center on Influenza and other Transmissible Infections
- VisMederi S.r.l., Siena, Italy
| | - ILARIA MANINI
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- Interuniversity Research Center on Influenza and other Transmissible Infections
| |
Collapse
|
45
|
Priyanka, Mohan B, Poonia E, Kumar S, Virender, Singh C, Xiong J, Liu X, Pombeiro AJL, Singh G. COVID-19 Virus Structural Details: Optical and Electrochemical Detection. J Fluoresc 2024; 34:479-500. [PMID: 37382834 DOI: 10.1007/s10895-023-03307-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/12/2023] [Indexed: 06/30/2023]
Abstract
The increasing viral species have ruined people's health and the world's economy. Therefore, it is urgent to design bio-responsive materials to provide a vast platform for detecting a different family's passive or active virus. One can design a reactive functional unit for that moiety based on the particular bio-active moieties in viruses. Nanomaterials as optical and electrochemical biosensors have enabled better tools and devices to develop rapid virus detection. Various material science platforms are available for real-time monitoring and detecting COVID-19 and other viral loads. In this review, we discuss the recent advances of nanomaterials in developing the tools for optical and electrochemical sensing COVID-19. In addition, nanomaterials used to detect other human viruses have been studied, providing insights for developing COVID-19 sensing materials. The basic strategies for nanomaterials develop as virus sensors, fabrications, and detection performances are studied. Moreover, the new methods to enhance the virus sensing properties are discussed to provide a gateway for virus detection in variant forms. The study will provide systematic information and working of virus sensors. In addition, the deep discussion of structural properties and signal changes will offer a new gate for researchers to develop new virus sensors for clinical applications.
Collapse
Affiliation(s)
- Priyanka
- Department of Chemistry and Centre of Advanced Studies, Panjab University, Chandigarh, 160014, India
| | - Brij Mohan
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Av. RoviscoPais, 1049-001, Lisbon, Portugal.
| | - Ekta Poonia
- Department of Chemistry, Deenbandhu Chhotu Ram University of Science & Technology, Murthal, Sonepat, 131039, Haryana, India
| | - Sandeep Kumar
- School of Science, Harbin Institute of Technology (Shenzhen), Shenzhen, 518055, China
| | - Virender
- Department of Chemistry, Kurukshetra University, Kurukshetra, 136119, Haryana, India
| | - Charan Singh
- Department of Pharmaceutical Sciences, School of Sciences, Hemvati Nandan Bahuguna Garhwal University (A Central University), Srinagar, Uttarakhand, 246174, India
| | - Jichuan Xiong
- Electronic and Optical Engineering, Nanjing University of Science and Technology, Nanjing, 210094, People's Republic of China
| | - Xuefeng Liu
- Electronic and Optical Engineering, Nanjing University of Science and Technology, Nanjing, 210094, People's Republic of China
| | - Armando J L Pombeiro
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Av. RoviscoPais, 1049-001, Lisbon, Portugal
| | - Gurjaspreet Singh
- Department of Chemistry and Centre of Advanced Studies, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
46
|
Rochowski MT, Jayathilake K, Balcerak JM, Selvan MT, Gunasekara S, Miller C, Rudd JM, Lacombe VA. Impact of Delta SARS-CoV-2 Infection on Glucose Metabolism: Insights on Host Metabolism and Virus Crosstalk in a Feline Model. Viruses 2024; 16:295. [PMID: 38400070 PMCID: PMC10893195 DOI: 10.3390/v16020295] [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: 01/10/2024] [Revised: 02/07/2024] [Accepted: 02/10/2024] [Indexed: 02/25/2024] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) causes enhanced mortality in people with metabolic and cardiovascular diseases. Other highly infectious RNA viruses have demonstrated dependence on glucose transport and utilization, so we hypothesized that SARS-CoV-2 infection could lead to alterations in cellular and whole-body glucose metabolism. Twenty-four healthy domestic cats were intratracheally inoculated with B.1.617.2 (delta) SARS-CoV-2 and samples were collected at 4- and 12-days post-inoculation (dpi). Blood glucose and circulating cortisol concentrations were elevated at 4 and 12 dpi. Serum insulin concentration was statistically significantly decreased, while angiotensin 2 concentration was elevated at 12 dpi. SARS-CoV-2 RNA was detected in the pancreas and skeletal muscle at low levels; however, no change in the number of insulin-producing cells or proinflammatory cytokines was observed in the pancreas of infected cats through 12 dpi. SARS-CoV-2 infection statistically significantly increased GLUT protein expression in both the heart and lungs, correlating with increased AMPK expression. In brief, SARS-CoV-2 increased blood glucose concentration and cardio-pulmonary GLUT expression through an AMPK-dependent mechanism, without affecting the pancreas, suggesting that SARS-CoV-2 induces the reprogramming of host glucose metabolism. A better understanding of host cell metabolism and virus crosstalk could lead to the discovery of novel metabolic therapeutic targets for patients affected by COVID-19.
Collapse
Affiliation(s)
- Matthew T. Rochowski
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA; (M.T.R.)
- Harold Hamm Diabetes Center, Oklahoma City, OK 73104, USA
| | - Kaushalya Jayathilake
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA; (M.T.R.)
| | - John-Michael Balcerak
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA; (M.T.R.)
| | - Miruthula Tamil Selvan
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA; (M.T.S.); (S.G.); (C.M.); (J.M.R.)
| | - Sachithra Gunasekara
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA; (M.T.S.); (S.G.); (C.M.); (J.M.R.)
| | - Craig Miller
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA; (M.T.S.); (S.G.); (C.M.); (J.M.R.)
| | - Jennifer M. Rudd
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA; (M.T.S.); (S.G.); (C.M.); (J.M.R.)
| | - Véronique A. Lacombe
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA; (M.T.R.)
- Harold Hamm Diabetes Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
47
|
Ao J, Ma AX, Li J, Wang CY, Fu DD, Du L, Yu C, Liu SL, Wang ZG, Pang DW. Real-Time Dissection of the Exosome Pathway for Influenza Virus Infection. ACS NANO 2024; 18:4507-4519. [PMID: 38270127 DOI: 10.1021/acsnano.3c11309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
Exosomes play an important role in the spread of viral infections and immune escape. However, the exact ability and mechanisms by which exosomes produced during viral infections (vExos) infect host cells are still not fully understood. In this study, we developed a dual-color exosome labeling strategy that simultaneously labels the external and internal structures of exosomes with quantum dots to enable in situ monitoring of the transport process of vExos in live cells using the single-particle tracking technique. Our finding revealed that vExos contains the complete influenza A virus (IAV) genome and viral ribonucleoprotein complexes (vRNPs) proteins but lacks viral envelope proteins. Notably, these vExos have the ability to infect cells and produce progeny viruses. We also found that vExos are transported in three stages, slow-fast-slow, and move to the perinuclear region via microfilaments and microtubules. About 30% of internalized vExos shed the external membrane and release the internal vRNPs into the cytoplasm by fusion with endolysosomes. This study suggested that vExos plays a supporting role in IAV infection by assisting with IAV propagation in a virus-independent manner. It emphasizes the need to consider the infectious potential of vExos and draws attention to the potential risk of exosomes produced by viral infections.
Collapse
Affiliation(s)
- Jian Ao
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, People's Republic of China
| | - Ai-Xin Ma
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
| | - Jing Li
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, People's Republic of China
| | - Chun-Yu Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
| | - Dan-Dan Fu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, People's Republic of China
| | - Lei Du
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, People's Republic of China
| | - Cong Yu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
| | - Shu-Lin Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
| | - Zhi-Gang Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
| | - Dai-Wen Pang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, People's Republic of China
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
| |
Collapse
|
48
|
Chung PYJ, Dhillon SK, Simoens C, Cuypers L, Laenen L, Bonde J, Corbisier P, Buttinger G, Cocuzza CE, Van Gucht S, Van Ranst M, Arbyn M. Assessment of the clinical and analytical performance of three Seegene Allplex SARS-CoV-2 assays within the VALCOR framework. Microbiol Spectr 2024; 12:e0239723. [PMID: 38189291 PMCID: PMC10846132 DOI: 10.1128/spectrum.02397-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 12/05/2023] [Indexed: 01/09/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic demonstrated the need for accurate diagnostic testing for the early detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Although the pandemic has ended, accurate assays are still needed to monitor viral spread at national levels and beyond through population and wastewater surveillance. To enhance early detection, SARS-CoV-2 assays should have high diagnostic accuracy and should be validated to assure accurate results. Three distinct SARS-CoV-2 assays were evaluated with clinical samples using the VALCOR (VALidation of SARS-CORona Virus-2 assays) framework, with the TaqPath COVID-19 assay (ThermoFisher Scientific, USA) as a comparator. We evaluated clinical sensitivity, specificity, limit of detection (LOD), and overall concordance between comparator and three index Allplex SARS-CoV-2 assays (Seegene, South Korea): Allplex-SC2, Allplex-SC2Fast (Fast PCR), and Allplex-SC2FabR (SARS-CoV-2/FluA/FluB/respiratory syncytial virus). Analytical performance and LOD of index assays were assessed using a dilution series of three synthetic SARS-CoV-2 sequence reference materials (RMs). Ninety SARS-CoV-2 positives and 90 SARS-CoV-2 negatives were tested. All Allplex assays had 100.0% sensitivity (95%CI = 95.9%-100.0%). Allplex-SC2 and Allplex-SC2Fast assays had 97.8% specificity (95%CI = 92.3%-99.7%) and 98.9% overall concordance [κ = 0.978 (95%CI = 0.947-1.000)]. Allplex-SC2FabR assay showed 100.0% specificity (95%CI = 95.9%-100.0%) and 100.0% overall concordance [κ = 1.000 (95%CI = 1.000-1.000)]. LOD assessment of index assays revealed detection down to 2.61 × 102 copies/mL in clinical samples, while the analytical LOD was 9.00 × 102 copies/mL. In conclusion, the evaluation of the three Seegene Allplex SARS-CoV-2 assays showed high sensitivity and specificity and an overall good assay concordance with the comparator. The assays showed low analytical LOD using RM and even a slightly lower LOD in clinical samples. Non-overlapping target gene sequences between SARS-CoV-2 assays and RMs emphasize the need for aligning targeted sequences of diagnostic assays and RMs.IMPORTANCEThe coronavirus disease 2019 pandemic has a significant impact on global public health, economies, and societies. As shown through the first phases of the pandemic, accurate and timely diagnosis is crucial for disease control, prevention, and monitoring. Though the pandemic phase of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has concluded, diagnostic assays remain in demand to monitor SARS-CoV-2 at the individual patient level, regionally, and nationally, as well as to remain an infectious disease preparedness instrument to monitor any new SARS-CoV-2 dissemination across borders using population and wastewater surveillance. The anticipation by WHO and central health care policy entities such as the Center for Disease Control, EMA, and multiple national health authorities is that SARS-CoV-2 will reside as an endemic respiratory disease for years to come. The key strategic consideration is hence shifting from combating a pandemic situation with a high number of patients to instead allowing precise diagnostics of suspected patients with the intention of correct management in a low-prevalence setting.
Collapse
Affiliation(s)
- Pui Yan Jenny Chung
- Unit of Cancer Epidemiology, Belgian Cancer Centre, Sciensano, Brussels, Belgium
| | - Sharonjit K. Dhillon
- Unit of Cancer Epidemiology, Belgian Cancer Centre, Sciensano, Brussels, Belgium
| | - Cindy Simoens
- Unit of Cancer Epidemiology, Belgian Cancer Centre, Sciensano, Brussels, Belgium
| | - Lize Cuypers
- Department of Laboratory Medicine, National Reference Centre for Respiratory Pathogens, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Clinical Microbiology, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Lies Laenen
- Department of Laboratory Medicine, National Reference Centre for Respiratory Pathogens, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Clinical Microbiology, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Jesper Bonde
- Molecular Pathology Laboratory, Department of Pathology, AHH-Hvidovre Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | - Clementina E. Cocuzza
- Laboratory of Clinical Microbiology and Virology, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | | | - Marc Van Ranst
- Laboratory of Clinical and Epidemiological Virology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Marc Arbyn
- Unit of Cancer Epidemiology, Belgian Cancer Centre, Sciensano, Brussels, Belgium
- Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, University of Ghent, Ghent, Belgium
| |
Collapse
|
49
|
Di Pietro C, Haberman AM, Lindenbach BD, Smith PC, Bruscia EM, Allore HG, Vander Wyk B, Tyagi A, Zeiss CJ. Prior Influenza Infection Mitigates SARS-CoV-2 Disease in Syrian Hamsters. Viruses 2024; 16:246. [PMID: 38400021 PMCID: PMC10891789 DOI: 10.3390/v16020246] [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: 01/11/2024] [Revised: 01/28/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Seasonal infection rates of individual viruses are influenced by synergistic or inhibitory interactions between coincident viruses. Endemic patterns of SARS-CoV-2 and influenza infection overlap seasonally in the Northern hemisphere and may be similarly influenced. We explored the immunopathologic basis of SARS-CoV-2 and influenza A (H1N1pdm09) interactions in Syrian hamsters. H1N1 given 48 h prior to SARS-CoV-2 profoundly mitigated weight loss and lung pathology compared to SARS-CoV-2 infection alone. This was accompanied by the normalization of granulocyte dynamics and accelerated antigen-presenting populations in bronchoalveolar lavage and blood. Using nasal transcriptomics, we identified a rapid upregulation of innate and antiviral pathways induced by H1N1 by the time of SARS-CoV-2 inoculation in 48 h dual-infected animals. The animals that were infected with both viruses also showed a notable and temporary downregulation of mitochondrial and viral replication pathways. Quantitative RT-PCR confirmed a decrease in the SARS-CoV-2 viral load and lower cytokine levels in the lungs of animals infected with both viruses throughout the course of the disease. Our data confirm that H1N1 infection induces rapid and transient gene expression that is associated with the mitigation of SARS-CoV-2 pulmonary disease. These protective responses are likely to begin in the upper respiratory tract shortly after infection. On a population level, interaction between these two viruses may influence their relative seasonal infection rates.
Collapse
Affiliation(s)
- Caterina Di Pietro
- Department of Pediatrics, Yale School of Medicine, New Haven, CT 06519, USA; (C.D.P.); (E.M.B.)
| | - Ann M. Haberman
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06519, USA;
| | - Brett D. Lindenbach
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06519, USA;
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06519, USA;
| | - Peter C. Smith
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06519, USA;
| | - Emanuela M. Bruscia
- Department of Pediatrics, Yale School of Medicine, New Haven, CT 06519, USA; (C.D.P.); (E.M.B.)
| | - Heather G. Allore
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06519, USA; (H.G.A.); (B.V.W.)
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06519, USA
| | - Brent Vander Wyk
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06519, USA; (H.G.A.); (B.V.W.)
| | - Antariksh Tyagi
- Department of Genetics, Yale Center for Genome Analysis, New Haven, CT 06519, USA;
| | - Caroline J. Zeiss
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06519, USA;
| |
Collapse
|
50
|
Chen Z, Yuan Y, Hu Q, Zhu A, Chen F, Li S, Guan X, Lv C, Tang T, He Y, Cheng J, Zheng J, Hu X, Zhao J, Zhao J, Sun J. SARS-CoV-2 immunity in animal models. Cell Mol Immunol 2024; 21:119-133. [PMID: 38238440 PMCID: PMC10806257 DOI: 10.1038/s41423-023-01122-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 01/25/2024] Open
Abstract
The COVID-19 pandemic, which was caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a worldwide health crisis due to its transmissibility. SARS-CoV-2 infection results in severe respiratory illness and can lead to significant complications in affected individuals. These complications encompass symptoms such as coughing, respiratory distress, fever, infectious shock, acute respiratory distress syndrome (ARDS), and even multiple-organ failure. Animal models serve as crucial tools for investigating pathogenic mechanisms, immune responses, immune escape mechanisms, antiviral drug development, and vaccines against SARS-CoV-2. Currently, various animal models for SARS-CoV-2 infection, such as nonhuman primates (NHPs), ferrets, hamsters, and many different mouse models, have been developed. Each model possesses distinctive features and applications. In this review, we elucidate the immune response elicited by SARS-CoV-2 infection in patients and provide an overview of the characteristics of various animal models mainly used for SARS-CoV-2 infection, as well as the corresponding immune responses and applications of these models. A comparative analysis of transcriptomic alterations in the lungs from different animal models revealed that the K18-hACE2 and mouse-adapted virus mouse models exhibited the highest similarity with the deceased COVID-19 patients. Finally, we highlighted the current gaps in related research between animal model studies and clinical investigations, underscoring lingering scientific questions that demand further clarification.
Collapse
Affiliation(s)
- Zhao Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Yaochang Yuan
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Qingtao Hu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, China
| | - Airu Zhu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Fenghua Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Shu Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Xin Guan
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Chao Lv
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Tian Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Yiyun He
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Jinling Cheng
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Jie Zheng
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Xiaoyu Hu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Jingxian Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China.
- Guangzhou National Laboratory, Guangzhou, Guangdong, 510005, China.
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China.
- Guangzhou National Laboratory, Guangzhou, Guangdong, 510005, China.
- Shanghai Institute for Advanced Immunochemical Studies, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, the Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, 518005, China.
| | - Jing Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China.
| |
Collapse
|