1
|
Ibrahimová M, Jamriková V, Pavelková K, Bořecká K. Nucleocapsid Antibodies as an Optimal Serological Marker of SARS-CoV-2 Infection: A Longitudinal Study at the Thomayer University Hospital. J Clin Lab Anal 2025; 39:e25149. [PMID: 39760288 PMCID: PMC11821721 DOI: 10.1002/jcla.25149] [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: 02/20/2024] [Revised: 12/04/2024] [Accepted: 12/24/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND The longitudinal study was conducted over the initial 2 years of the COVID-19 pandemic, spanning from June 2020 to December 2022, in healthcare workers (HCWs) of the Thomayer University Hospital. A total of 3892 blood samples were collected and analyzed for total nucleocapsid (N) antibodies. The aim of the study was to evaluate the dynamics of N antibodies, their relationship to the PCR test, spike (S) antibodies, interferon-gamma, and prediction of reinfection with SARS-CoV-2. METHODS Blood collections were performed in three rounds, along with questionnaires addressing clinical symptoms of past infection, PCR testing, and vaccination. Antibody measurements included total N antibodies (Roche Diagnostics) and postvaccination S antibodies (Euroimmun). Cellular immunity was tested by interferon-gamma release assay (Euroimmun). RESULTS At the end of the study, 35.9% of HCWs were positive for N antibodies, and 39.5% of HCWs had either known PCR positivity or N antibodies or both. Ten percent of participants had no knowledge of a COVID-19 infection and 35% of positive individuals exhibited no symptoms. The values of positive antibodies decrease over a period of 6 months to 1 year, depending on the initial value, and their dynamics are highly variable. The study also demonstrated that the highest levels of spike antibodies and interferon-gamma occur during so-called hybrid immunity. CONCLUSION Nucleocapsid antibodies proved valuable in monitoring SARS-CoV-2 infection dynamics, and they may detect cases of SARS-CoV-2 infection missed by PCR tests. The study identified distinct patterns in antibody dynamics and protection of hybrid immunity during reinfection.
Collapse
Affiliation(s)
- Markéta Ibrahimová
- Laboratory of ImmunologyThomayer University HospitalPragueCzech Republic
| | - Vladislava Jamriková
- Department of Clinical BiochemistryThomayer University HospitalPragueCzech Republic
| | - Kateřina Pavelková
- Department of Hospital Epidemiology and Infection ControlThomayer University HospitalPragueCzech Republic
| | - Klára Bořecká
- Department of Clinical BiochemistryThomayer University HospitalPragueCzech Republic
| |
Collapse
|
2
|
Orosa Vázquez I, Díaz M, Zúñiga Rosales Y, Amada K, Chang J, Relova Hernández E, Tundidor Y, Roblejo Balbuena H, Monzón G, Torres Rives B, Noa Romero E, Carrillo Valdés D, Valdivia Álvarez I, Delahanty Fernández A, Díaz C, Solozabal J, Gil M, Sánchez B, Rojas G, Marcheco B, Carmenate T. Studying the Humoral Response against SARS-CoV-2 in Cuban COVID-19 Recovered Patients. J Immunol Res 2024; 2024:7112940. [PMID: 39359695 PMCID: PMC11446615 DOI: 10.1155/2024/7112940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/30/2024] [Accepted: 09/05/2024] [Indexed: 10/04/2024] Open
Abstract
Understanding the immune response generated by SARS-CoV-2 is critical for assessing efficient therapeutic protocols and gaining insights into the durability of protective immunity. The current work was aimed at studying the specific humoral responses against SARS-CoV-2 in Cuban COVID-19 convalescents. We developed suitable tools and methods based on ELISA methodology, for supporting this evaluation. Here, we describe the development of an ELISA for the quantification of anti-RBD IgG titers in a large number of samples and a similar test in the presence of NH4SCN as chaotropic agent for estimating the RBD specific antibody avidity. Additionally, a simple and rapid ELISA based on antibody-mediated blockage of the binding RBD-ACE2 was implemented for detecting, as a surrogate of conventional test, the levels of anti-RBD inhibitory antibodies in convalescent sera. In a cohort of 273 unvaccinated convalescents, we identified higher anti-RBD IgG titer (1 : 1,330, p < 0.0001) and higher levels of inhibitory antibodies blocking RBD-ACE2 binding (1 : 216, p < 0.05) among those who had recovered from severe illness. Our results suggest that disease severity, and not demographic features such as age, sex, and skin color, is the main determinant of the magnitude and neutralizing ability of the anti-RBD antibody response. An additional paired longitudinal assessment in 14 symptomatic convalescents revealed a decline in the antiviral antibody response and the persistence of neutralizing antibodies for at least 4 months after the onset of symptoms. Overall, SARS-CoV-2 infection elicits different levels of antibody response according to disease severity that declines over time and can be monitored using our homemade serological assays.
Collapse
Affiliation(s)
- Ivette Orosa Vázquez
- Center of Molecular Immunology, 15th Avenue and 216 Street, Siboney, Playa, Havana, Cuba
| | - Marianniz Díaz
- Center of Molecular Immunology, 15th Avenue and 216 Street, Siboney, Playa, Havana, Cuba
| | - Yaima Zúñiga Rosales
- National Center of Medical Genetics, 31st Avenue, N°3102 and 146 Street, Cubanacán, Playa, Havana, Cuba
| | - Klayris Amada
- Julio Trigo Hospital, km 7½ Calzada de Bejucal, Diez de Octubre, Havana, Cuba
| | - Janoi Chang
- Center of Molecular Immunology, 15th Avenue and 216 Street, Siboney, Playa, Havana, Cuba
| | | | - Yaima Tundidor
- Center of Molecular Immunology, 15th Avenue and 216 Street, Siboney, Playa, Havana, Cuba
| | - Hilda Roblejo Balbuena
- National Center of Medical Genetics, 31st Avenue, N°3102 and 146 Street, Cubanacán, Playa, Havana, Cuba
| | - Giselle Monzón
- National Center of Medical Genetics, 31st Avenue, N°3102 and 146 Street, Cubanacán, Playa, Havana, Cuba
| | - Bárbara Torres Rives
- National Center of Medical Genetics, 31st Avenue, N°3102 and 146 Street, Cubanacán, Playa, Havana, Cuba
| | - Enrique Noa Romero
- Research Center of Civil Defense, José de las Lajas, San, Mayabeque, Cuba
| | | | | | | | - Claudia Díaz
- Center of Molecular Immunology, 15th Avenue and 216 Street, Siboney, Playa, Havana, Cuba
| | - Joaquín Solozabal
- Center of Molecular Immunology, 15th Avenue and 216 Street, Siboney, Playa, Havana, Cuba
| | - Mileidys Gil
- Julio Trigo Hospital, km 7½ Calzada de Bejucal, Diez de Octubre, Havana, Cuba
| | - Belinda Sánchez
- Center of Molecular Immunology, 15th Avenue and 216 Street, Siboney, Playa, Havana, Cuba
| | - Gertrudis Rojas
- Center of Molecular Immunology, 15th Avenue and 216 Street, Siboney, Playa, Havana, Cuba
| | - Beatriz Marcheco
- National Center of Medical Genetics, 31st Avenue, N°3102 and 146 Street, Cubanacán, Playa, Havana, Cuba
| | - Tania Carmenate
- Center of Molecular Immunology, 15th Avenue and 216 Street, Siboney, Playa, Havana, Cuba
| |
Collapse
|
3
|
Lisi S, Malerba F, Quaranta P, Florio R, Vitaloni O, Monaca E, Bruni Ercole B, Bitonti AR, Del Perugia O, Mignanelli M, Perrera P, Sabbatella R, Raimondi F, Piazza CR, Moles A, Alfano C, Pistello M, Cattaneo A. Selection and characterization of human scFvs targeting the SARS-CoV-2 nucleocapsid protein isolated from antibody libraries of COVID-19 patients. Sci Rep 2024; 14:15864. [PMID: 38982108 PMCID: PMC11233501 DOI: 10.1038/s41598-024-66558-0] [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: 01/18/2024] [Accepted: 07/02/2024] [Indexed: 07/11/2024] Open
Abstract
In 2019, the novel SARS-CoV-2 coronavirus emerged in China, causing the pneumonia named COVID-19. At the beginning, all research efforts were focused on the spike (S) glycoprotein. However, it became evident that the nucleocapsid (N) protein is pivotal in viral replication, genome packaging and evasion of the immune system, is highly immunogenic, which makes it another compelling target for antibody development alongside the spike protein. This study focused on the construction of single chain fragments variable (scFvs) libraries from SARS-CoV-2-infected patients to establish a valuable, immortalized and extensive antibodies source. We used the Intracellular Antibody Capture Technology to select a panel of scFvs against the SARS-CoV-2 N protein. The whole panel of scFv was expressed and characterized both as intrabodies and recombinant proteins. ScFvs were then divided into 2 subgroups: those that exhibited high binding activity to N protein when expressed in yeast or in mammalian cells as intrabodies, and those purified as recombinant proteins, displaying affinity for recombinant N protein in the nanomolar range. This panel of scFvs against the N protein represents a novel platform for research and potential diagnostic applications.
Collapse
Affiliation(s)
- Simonetta Lisi
- Bio@SNS Laboratory, Scuola Normale Superiore, 56126, Pisa, Italy
| | - Francesca Malerba
- Fondazione EBRI (European Brain Research Institute) Rita Levi-Montalcini, 00161, Rome, Italy
| | - Paola Quaranta
- Retrovirus Centre, Department of Translational Research, University of Pisa, 56126, Pisa, Italy
- Virology Operative Unit, Pisa University Hospital, 56124, Pisa, Italy
| | - Rita Florio
- Fondazione EBRI (European Brain Research Institute) Rita Levi-Montalcini, 00161, Rome, Italy
| | - Ottavia Vitaloni
- Bio@SNS Laboratory, Scuola Normale Superiore, 56126, Pisa, Italy
| | - Elisa Monaca
- Structural Biology and Biophysics Unit, Fondazione Ri.MED, 90133, Palermo, Italy
| | - Bruno Bruni Ercole
- Fondazione EBRI (European Brain Research Institute) Rita Levi-Montalcini, 00161, Rome, Italy
| | | | - Olga Del Perugia
- Bio@SNS Laboratory, Scuola Normale Superiore, 56126, Pisa, Italy
| | | | - Paola Perrera
- Retrovirus Centre, Department of Translational Research, University of Pisa, 56126, Pisa, Italy
| | - Raffaele Sabbatella
- Structural Biology and Biophysics Unit, Fondazione Ri.MED, 90133, Palermo, Italy
| | | | - Carmen Rita Piazza
- Retrovirus Centre, Department of Translational Research, University of Pisa, 56126, Pisa, Italy
- Department of Medical Biotechnologies, University of Siena, 53100, Siena, Italy
| | - Anna Moles
- Genomnia Srl, 20091, Bresso, MI, Italy
- Institute of Biochemistry and Cell Biology, CNR, 80131, Napoli, Italy
| | - Caterina Alfano
- Structural Biology and Biophysics Unit, Fondazione Ri.MED, 90133, Palermo, Italy
| | - Mauro Pistello
- Retrovirus Centre, Department of Translational Research, University of Pisa, 56126, Pisa, Italy
- Virology Operative Unit, Pisa University Hospital, 56124, Pisa, Italy
| | - Antonino Cattaneo
- Bio@SNS Laboratory, Scuola Normale Superiore, 56126, Pisa, Italy.
- Fondazione EBRI (European Brain Research Institute) Rita Levi-Montalcini, 00161, Rome, Italy.
| |
Collapse
|
4
|
Panahi M, Teimoori A, Esmaeili S, Aminianfar H, Milani A, Hosseini SY, Esmaeili P, Biglari A, Baesi K. Stability of Neutralizing Antibody of PastoCoAd Vaccine Candidates against a Variant of Concern of SARS-CoV-2 in Animal Models. IRANIAN BIOMEDICAL JOURNAL 2024; 28:214-20. [PMID: 39044638 PMCID: PMC11444480 DOI: 10.61186/ibj.3980] [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: 07/25/2024]
Abstract
Background Since the beginning of the SARS-CoV-2 pandemic, there have been mutations caused by new SARS-CoV-2 variants, such as Alpha, Beta, Gamma, Delta, and Omicron, recognized as the variants of concern (VOC) worldwide. These variants can affect vaccine efficacy, disease control, and treatment effectiveness. The present study aimed to evaluate the levels of total and neutralizing antibodies produced by PastoCoAd vaccine candidates against the VOC strains at different time points. Methods Two vaccine candidates were employed against SARS-CoV-2 using adenoviral vectors: prime only (a mixture of rAd5-S and rAd5 RBD-N) and heterologous prime-boost (rAd5-S/SOBERANA vaccine). The immunogenicity of these vaccine candidates was assessed in mouse, rabbit, and hamster models using ELISA assay and virus neutralization antibody test. Results The immunogenicity results indicated a significant increase in both total and neutralizing antibodies titers in the groups receiving the vaccine candidates at various time points compared to the control group (p < 0.05). The results also showed that the PastoCoAd vaccine candidates Ad5 S & RBD-N and Ad5 S/SOBERANA could neutralize the VOC strains in the animal models. Conclusion The ability of vaccine candidate to neutralize the VOC strains in animal models by generating neutralizing antibodies at different time points may be attributed to the use of the platform based on the Adenoviral vector, the N proteins in the Ad5 S & RBD-N vaccine candidate, and the SOBERANA Plus booster in the Ad5 S/SOBERANA vaccine candidate.
Collapse
Affiliation(s)
- Mohammad Panahi
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Ali Teimoori
- Department of Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Saber Esmaeili
- National Reference Laboratory for Plague, Tularemia and Q Fever, Research Center for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Akanlu, Kabudar Ahang, Hamadan, Iran
- Department of Epidemiology and Biostatistics, Research Center for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | | | - Alireza Milani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Younes Hosseini
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parisa Esmaeili
- National Reference Laboratory for Plague, Tularemia and Q Fever, Research Center for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Akanlu, Kabudar Ahang, Hamadan, Iran
- Department of Epidemiology and Biostatistics, Research Center for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | | | - Kazem Baesi
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
5
|
Xu L, Abudunaibi B, Zeng Z, Zhao Y, Wang Y, Guo X, Zhang Y, Li T, Lu W, Tian W, Guo Z, Su C, Chen T. Relationship of various COVID-19 antibody titer with individual characteristics and prediction of future epidemic trend in Xiamen City, China. J Thorac Dis 2024; 16:2404-2420. [PMID: 38738254 PMCID: PMC11087623 DOI: 10.21037/jtd-23-1516] [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: 09/27/2023] [Accepted: 02/29/2024] [Indexed: 05/14/2024]
Abstract
Background Reinfection of coronavirus disease 2019 (COVID-19) has raised concerns about how reliable immunity from infection and vaccination is. With mass testing for the virus halted, understanding the current prevalence of COVID-19 is crucial. This study investigated 1,191 public health workers at the Xiamen Center for Disease Control, focusing on changes in antibody titers and their relationship with individual characteristics. Methods The study began by describing the epidemiological characteristics of the study participants. Multilinear regression (MLR) models were employed to explore the associations between individual attributes and antibody titers. Additionally, group-based trajectory models (GBTMs) were utilized to identify trajectories in antibody titer changes. To predict and simulate future epidemic trends and examine the correlation of antibody decay with epidemics, a high-dimensional transmission dynamics model was constructed. Results Analysis of epidemiological characteristics revealed significant differences in vaccination status between infected and non-infected groups (χ2=376.706, P<0.05). However, the distribution of antibody titers among the infected and vaccinated populations was not significantly different. The MLR model identified age as a common factor affecting titers of immunoglobulin G (IgG), immunoglobulin M (IgM), and neutralizing antibody (NAb), while other factors showed varying impacts. History of pulmonary disease and hospitalization influenced IgG titer, and factors such as gender, smoking, family history of pulmonary diseases, and hospitalization impacted NAb titers. Age was the sole determinant of IgM titers in this study. GBTM analysis indicated a "gradual decline type" trajectory for IgG (95.65%), while IgM and NAb titers remained stable over the study period. The high-dimensional transmission dynamics model predicted and simulated peak epidemic periods in Xiamen City, which correlated with IgG decay. Age-group-specific simulations revealed a higher incidence and infection rate among individuals aged 30-39 years during both the second and third peaks, followed by those aged 40-49, 50-59, 18-29, and 70-79 years. Conclusions Our study shows that antibody titer could be influenced by age, previous pulmonary diseases as well as smoking. Furthermore, the decline in IgG titers is consistent with epidemic trends. These findings emphasize the need for further exploration of these factors and the development of optimized self-protection countermeasures against reinfection.
Collapse
Affiliation(s)
- Liansheng Xu
- Xiamen Center for Disease Control and Prevention, Xiamen, China
| | - Buasiyamu Abudunaibi
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
| | - Zhiqi Zeng
- Respiratory Disease AI Laboratory on Epidemic Intelligence and Medical Big Data Instrument Applications, Macao University of Science and Technology, Macao, China
| | - Yunkang Zhao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
| | - Yao Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
| | - Xiaohao Guo
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
| | - Yidun Zhang
- Xiamen Center for Disease Control and Prevention, Xiamen, China
| | - Tao Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
| | - Wenkui Lu
- Xiamen Center for Disease Control and Prevention, Xiamen, China
| | - Weiliang Tian
- School of Medicine, Duke University, Durham, NC, USA
| | - Zhinan Guo
- Xiamen Center for Disease Control and Prevention, Xiamen, China
| | - Chenghao Su
- Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, China
| | - Tianmu Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
| |
Collapse
|
6
|
Lobaina Y, Chen R, Suzarte E, Ai P, Huerta V, Musacchio A, Silva R, Tan C, Martín A, Lazo L, Guillén-Nieto G, Yang K, Perera Y, Hermida L. The Nucleocapsid Protein of SARS-CoV-2, Combined with ODN-39M, Is a Potential Component for an Intranasal Bivalent Vaccine with Broader Functionality. Viruses 2024; 16:418. [PMID: 38543783 PMCID: PMC10976088 DOI: 10.3390/v16030418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 05/23/2024] Open
Abstract
Despite the rapid development of vaccines against COVID-19, they have important limitations, such as safety issues, the scope of their efficacy, and the induction of mucosal immunity. The present study proposes a potential component for a new generation of vaccines. The recombinant nucleocapsid (N) protein from the SARS-CoV-2 Delta variant was combined with the ODN-39M, a synthetic 39 mer unmethylated cytosine-phosphate-guanine oligodeoxynucleotide (CpG ODN), used as an adjuvant. The evaluation of its immunogenicity in Balb/C mice revealed that only administration by intranasal route induced a systemic cross-reactive, cell-mediated immunity (CMI). In turn, this combination was able to induce anti-N IgA in the lungs, which, along with the specific IgG in sera and CMI in the spleen, was cross-reactive against the nucleocapsid protein of SARS-CoV-1. Furthermore, the nasal administration of the N + ODN-39M preparation, combined with RBD Delta protein, enhanced the local and systemic immune response against RBD, with a neutralizing capacity. Results make the N + ODN-39M preparation a suitable component for a future intranasal vaccine with broader functionality against Sarbecoviruses.
Collapse
Affiliation(s)
- Yadira Lobaina
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou 425000, China; (Y.L.); (R.C.); (P.A.); (V.H.); (A.M.); (R.S.); (C.T.)
- R&D Department, Yongzhou Zhong Gu Biotechnology Co., Ltd., Yangjiaqiao Street, Lengshuitan District, Yongzhou 425000, China
| | - Rong Chen
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou 425000, China; (Y.L.); (R.C.); (P.A.); (V.H.); (A.M.); (R.S.); (C.T.)
- Yongzhou Development and Construction Investment Co., Ltd. (YDCI), Changfeng Industry Park, Yongzhou Economic and Technological Development Zone, No. 1 Liebao Road, Lengshuitan District, Yongzhou 425000, China
| | - Edith Suzarte
- CIGB: Research Department, Center for Genetic Engineering and Biotechnology, Havana 10600, Cuba; (E.S.); (A.M.); (L.L.); (G.G.-N.)
| | - Panchao Ai
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou 425000, China; (Y.L.); (R.C.); (P.A.); (V.H.); (A.M.); (R.S.); (C.T.)
- Yongzhou Development and Construction Investment Co., Ltd. (YDCI), Changfeng Industry Park, Yongzhou Economic and Technological Development Zone, No. 1 Liebao Road, Lengshuitan District, Yongzhou 425000, China
| | - Vivian Huerta
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou 425000, China; (Y.L.); (R.C.); (P.A.); (V.H.); (A.M.); (R.S.); (C.T.)
- CIGB: Research Department, Center for Genetic Engineering and Biotechnology, Havana 10600, Cuba; (E.S.); (A.M.); (L.L.); (G.G.-N.)
| | - Alexis Musacchio
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou 425000, China; (Y.L.); (R.C.); (P.A.); (V.H.); (A.M.); (R.S.); (C.T.)
- CIGB: Research Department, Center for Genetic Engineering and Biotechnology, Havana 10600, Cuba; (E.S.); (A.M.); (L.L.); (G.G.-N.)
| | - Ricardo Silva
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou 425000, China; (Y.L.); (R.C.); (P.A.); (V.H.); (A.M.); (R.S.); (C.T.)
- BCF: R&D Section, Representative Office BCF in China, Jingtai Tower, No. 24 Jianguomen Wai Street, Chaoyang District, Beijing 100022, China
| | - Changyuan Tan
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou 425000, China; (Y.L.); (R.C.); (P.A.); (V.H.); (A.M.); (R.S.); (C.T.)
- Yongzhou Development and Construction Investment Co., Ltd. (YDCI), Changfeng Industry Park, Yongzhou Economic and Technological Development Zone, No. 1 Liebao Road, Lengshuitan District, Yongzhou 425000, China
| | - Alejandro Martín
- CIGB: Research Department, Center for Genetic Engineering and Biotechnology, Havana 10600, Cuba; (E.S.); (A.M.); (L.L.); (G.G.-N.)
| | - Laura Lazo
- CIGB: Research Department, Center for Genetic Engineering and Biotechnology, Havana 10600, Cuba; (E.S.); (A.M.); (L.L.); (G.G.-N.)
| | - Gerardo Guillén-Nieto
- CIGB: Research Department, Center for Genetic Engineering and Biotechnology, Havana 10600, Cuba; (E.S.); (A.M.); (L.L.); (G.G.-N.)
| | - Ke Yang
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou 425000, China; (Y.L.); (R.C.); (P.A.); (V.H.); (A.M.); (R.S.); (C.T.)
- Yongzhou Development and Construction Investment Co., Ltd. (YDCI), Changfeng Industry Park, Yongzhou Economic and Technological Development Zone, No. 1 Liebao Road, Lengshuitan District, Yongzhou 425000, China
| | - Yasser Perera
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou 425000, China; (Y.L.); (R.C.); (P.A.); (V.H.); (A.M.); (R.S.); (C.T.)
- R&D Department, Yongzhou Zhong Gu Biotechnology Co., Ltd., Yangjiaqiao Street, Lengshuitan District, Yongzhou 425000, China
- CIGB: Research Department, Center for Genetic Engineering and Biotechnology, Havana 10600, Cuba; (E.S.); (A.M.); (L.L.); (G.G.-N.)
| | - Lisset Hermida
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Lengshuitan District, Yongzhou 425000, China; (Y.L.); (R.C.); (P.A.); (V.H.); (A.M.); (R.S.); (C.T.)
- Yongzhou Development and Construction Investment Co., Ltd. (YDCI), Changfeng Industry Park, Yongzhou Economic and Technological Development Zone, No. 1 Liebao Road, Lengshuitan District, Yongzhou 425000, China
- BCF: R&D Section, Representative Office BCF in China, Jingtai Tower, No. 24 Jianguomen Wai Street, Chaoyang District, Beijing 100022, China
| |
Collapse
|
7
|
Zheng G, Qiu G, Qian H, Shu Q, Xu J. Multifaceted role of SARS-CoV-2 structural proteins in lung injury. Front Immunol 2024; 15:1332440. [PMID: 38375473 PMCID: PMC10875085 DOI: 10.3389/fimmu.2024.1332440] [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: 11/02/2023] [Accepted: 01/22/2024] [Indexed: 02/21/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the third human coronavirus to cause acute respiratory distress syndrome (ARDS) and contains four structural proteins: spike, envelope, membrane, and nucleocapsid. An increasing number of studies have demonstrated that all four structural proteins of SARS-CoV-2 are capable of causing lung injury, even without the presence of intact virus. Therefore, the topic of SARS-CoV-2 structural protein-evoked lung injury warrants more attention. In the current article, we first synopsize the structural features of SARS-CoV-2 structural proteins. Second, we discuss the mechanisms for structural protein-induced inflammatory responses in vitro. Finally, we list the findings that indicate structural proteins themselves are toxic and sufficient to induce lung injury in vivo. Recognizing mechanisms of lung injury triggered by SARS-CoV-2 structural proteins may facilitate the development of targeted modalities in treating COVID-19.
Collapse
Affiliation(s)
| | - Guanguan Qiu
- Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Huifeng Qian
- Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Qiang Shu
- The Children’s Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Jianguo Xu
- Shaoxing Second Hospital, Shaoxing, Zhejiang, China
- The Children’s Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| |
Collapse
|
8
|
Jackson HK, Long HM, Yam‐Puc JC, Palmulli R, Haigh TA, Gerber PP, Lee JS, Matheson NJ, Young L, Trowsdale J, Lo M, Taylor GS, Thaventhiran JE, Edgar JR. Bioengineered small extracellular vesicles deliver multiple SARS-CoV-2 antigenic fragments and drive a broad immunological response. J Extracell Vesicles 2024; 13:e12412. [PMID: 38339765 PMCID: PMC10858312 DOI: 10.1002/jev2.12412] [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: 08/10/2023] [Revised: 12/22/2023] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
The COVID-19 pandemic highlighted the clear risk that zoonotic viruses pose to global health and economies. The scientific community responded by developing several efficacious vaccines which were expedited by the global need for vaccines. The emergence of SARS-CoV-2 breakthrough infections highlights the need for additional vaccine modalities to provide stronger, long-lived protective immunity. Here we report the design and preclinical testing of small extracellular vesicles (sEVs) as a multi-subunit vaccine. Cell lines were engineered to produce sEVs containing either the SARS-CoV-2 Spike receptor-binding domain, or an antigenic region from SARS-CoV-2 Nucleocapsid, or both in combination, and we tested their ability to evoke immune responses in vitro and in vivo. B cells incubated with bioengineered sEVs were potent activators of antigen-specific T cell clones. Mice immunised with sEVs containing both sRBD and Nucleocapsid antigens generated sRBD-specific IgGs, nucleocapsid-specific IgGs, which neutralised SARS-CoV-2 infection. sEV-based vaccines allow multiple antigens to be delivered simultaneously resulting in potent, broad immunity, and provide a quick, cheap, and reliable method to test vaccine candidates.
Collapse
Affiliation(s)
- Hannah K. Jackson
- Department of PathologyUniversity of CambridgeCambridgeUK
- Exosis, Inc. Palm BeachPalm BeachFloridaUSA
| | - Heather M. Long
- Institute of Immunology and ImmunotherapyUniversity of BirminghamBirminghamUK
| | | | | | - Tracey A. Haigh
- Institute of Immunology and ImmunotherapyUniversity of BirminghamBirminghamUK
| | - Pehuén Pereyra Gerber
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID)University of CambridgeCambridgeUK
- Department of MedicineUniversity of CambridgeCambridgeUK
| | - Jin S. Lee
- Department of PathologyUniversity of CambridgeCambridgeUK
| | - Nicholas J. Matheson
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID)University of CambridgeCambridgeUK
- Department of MedicineUniversity of CambridgeCambridgeUK
- NHS Blood and TransplantCambridgeUK
| | | | | | - Mathew Lo
- Exosis, Inc. Palm BeachPalm BeachFloridaUSA
| | - Graham S. Taylor
- Institute of Immunology and ImmunotherapyUniversity of BirminghamBirminghamUK
| | | | - James R. Edgar
- Department of PathologyUniversity of CambridgeCambridgeUK
| |
Collapse
|
9
|
Zhang D, Kukkar D, Kim KH, Bhatt P. A comprehensive review on immunogen and immune-response proteins of SARS-CoV-2 and their applications in prevention, diagnosis, and treatment of COVID-19. Int J Biol Macromol 2024; 259:129284. [PMID: 38211928 DOI: 10.1016/j.ijbiomac.2024.129284] [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: 09/06/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/13/2024]
Abstract
Exposure to severe acute respiratory syndrome-corona virus-2 (SARS-CoV-2) prompts humoral immune responses in the human body. As the auxiliary diagnosis of a current infection, the existence of viral proteins can be checked from specific antibodies (Abs) induced by immunogenic viral proteins. For people with a weakened immune system, Ab treatment can help neutralize viral antigens to resist and treat the disease. On the other hand, highly immunogenic viral proteins can serve as effective markers for detecting prior infections. Additionally, the identification of viral particles or the presence of antibodies may help establish an immune defense against the virus. These immunogenic proteins rather than SARS-CoV-2 can be given to uninfected people as a vaccination to improve their coping ability against COVID-19 through the generation of memory plasma cells. In this work, we review immunogenic and immune-response proteins derived from SARS-CoV-2 with regard to their classification, origin, and diverse applications (e.g., prevention (vaccine development), diagnostic testing, and treatment (via neutralizing Abs)). Finally, advanced immunization strategies against COVID-19 are discussed along with the contemporary circumstances and future challenges.
Collapse
Affiliation(s)
- Daohong Zhang
- College of Food Engineering, Ludong University, Yantai 264025, Shandong, China; Bio-Nanotechnology Research Institute, Ludong University, Yantai 264025, Shandong, China
| | - Deepak Kukkar
- Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413, Punjab, India; University Center for Research and Development, Chandigarh University, Gharuan, Mohali 140413, Punjab, India
| | - Ki-Hyun Kim
- Department of Civil & Environmental Engineering, Hanyang University, 222 Wangsimni-Ro, Seoul 04763, Republic of Korea.
| | - Poornima Bhatt
- Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413, Punjab, India; University Center for Research and Development, Chandigarh University, Gharuan, Mohali 140413, Punjab, India
| |
Collapse
|
10
|
Ellis S, Way R, Nel M, Burleigh A, Doykov I, Kembou-Ringert J, Woodall M, Masonou T, Case KM, Ortez AT, McHugh TD, Casal A, McCoy LE, Murdan S, Hynds RE, Gilmour KC, Grandjean L, Cortina-Borja M, Heywood WE, Mills K, Smith CM. Salivary IgA and vimentin differentiate in vitro SARS-CoV-2 infection: A study of 290 convalescent COVID-19 patients. Mucosal Immunol 2024; 17:124-136. [PMID: 38007005 PMCID: PMC11139657 DOI: 10.1016/j.mucimm.2023.11.007] [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: 06/26/2023] [Revised: 11/07/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
SARS-CoV-2 initially infects cells in the nasopharynx and oral cavity. The immune system at these mucosal sites plays a crucial role in minimizing viral transmission and infection. To develop new strategies for preventing SARS-CoV-2 infection, this study aimed to identify proteins that protect against viral infection in saliva. We collected 551 saliva samples from 290 healthcare workers who had tested positive for COVID-19, before vaccination, between June and December 2020. The samples were categorized based on their ability to block or enhance infection using in vitro assays. Mass spectrometry and enzyme-linked immunosorbent assay experiments were used to identify and measure the abundance of proteins that specifically bind to SARS-CoV-2 antigens. Immunoglobulin (Ig)A specific to SARS-CoV-2 antigens was detectable in over 83% of the convalescent saliva samples. We found that concentrations of anti-receptor-binding domain IgA >500 pg/µg total protein in saliva correlate with reduced viral infectivity in vitro. However, there is a dissociation between the salivary IgA response to SARS-CoV-2, and systemic IgG titers in convalescent COVID-19 patients. Then, using an innovative technique known as spike-baited mass spectrometry, we identified novel spike-binding proteins in saliva, most notably vimentin, which correlated with increased viral infectivity in vitro and could serve as a therapeutic target against COVID-19.
Collapse
Affiliation(s)
- Samuel Ellis
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Rosie Way
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Miranda Nel
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Alice Burleigh
- UCL Great Ormond Street Institute of Child Health, London, UK; Centre for Adolescent Rheumatology, University College London, London, UK
| | - Ivan Doykov
- UCL Great Ormond Street Institute of Child Health, London, UK
| | | | | | - Tereza Masonou
- UCL Great Ormond Street Institute of Child Health, London, UK
| | | | | | - Timothy D McHugh
- UCL Centre for Clinical Microbiology, Royal Free Hospital, London, UK
| | - Antonio Casal
- Department of Pharmaceutics, UCL School of Pharmacy, London, UK
| | - Laura E McCoy
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | | | - Robert E Hynds
- Epithelial Cell Biology in ENT Research (EpiCENTR) Group, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Kimberly C Gilmour
- UCL Great Ormond Street Institute of Child Health, London, UK; Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Louis Grandjean
- UCL Great Ormond Street Institute of Child Health, London, UK; Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | | | - Wendy E Heywood
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Kevin Mills
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Claire M Smith
- UCL Great Ormond Street Institute of Child Health, London, UK.
| |
Collapse
|
11
|
Hoover AR, More S, Liu K, West CL, Valerio TI, Furrer CL, Adams JP, Yu N, Villalva C, Kumar A, Alleruzzo L, Lam SSK, Hode T, Papin JF, Chen WR. N-dihydrogalactochitosan serves as an effective mucosal adjuvant for intranasal vaccine in combination with recombinant viral proteins against respiratory infection. Acta Biomater 2024; 175:279-292. [PMID: 38160856 DOI: 10.1016/j.actbio.2023.12.039] [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: 04/18/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
Mucosal vaccinations for respiratory pathogens provide effective protection as they stimulate localized cellular and humoral immunities at the site of infection. Currently, the major limitation of intranasal vaccination is using effective adjuvants capable of withstanding the harsh environment imposed by the mucosa. Herein, we describe the efficacy of using a unique biopolymer, N-dihydrogalactochitosan (GC), as a nasal mucosal vaccine adjuvant against respiratory infections. Specifically, we mixed GC with recombinant SARS-CoV-2 trimeric spike (S) and nucleocapsid (NC) proteins to intranasally vaccinate K18-hACE2 transgenic mice, in comparison with Addavax (AV), an MF-59 equivalent. In contrast to AV, intranasal application of GC induces a robust, systemic antigen-specific antibody response and increases the number of T cells in the cervical lymph nodes. Moreover, GC+S+NC-vaccinated animals were largely resistant to the lethal SARS-CoV-2 challenge and experienced drastically reduced morbidity and mortality, with animal weights and behavior returning to normal 22 days post-infection. In contrast, animals intranasally vaccinated with AV+S+NC experienced severe weight loss, mortality, and respiratory distress, with none surviving beyond 6 days post-infection. Our findings demonstrate that GC can serve as a potent mucosal vaccine adjuvant against SARS-CoV-2 and potentially other respiratory viruses. STATEMENT OF SIGNIFICANCE: We demonstrated that a unique biopolymer, N-dihydrogalactochitosan (GC), was an effective nasal mucosal vaccine adjuvant against respiratory infections. Specifically, we mixed GC with recombinant SARS-CoV-2 trimeric spike (S) and nucleocapsid (NC) proteins to intranasally vaccinate K18-hACE2 transgenic mice, in comparison with Addavax (AV). In contrast to AV, GC induces a robust, systemic antigen-specific antibody response and increases the number of T cells in the cervical lymph nodes. About 90 % of the GC+S+NC-vaccinated animals survived the lethal SARS-CoV-2 challenge and remained healthy 22 days post-infection, while the AV+S+NC-vaccinated animals experienced severe weight loss and respiratory distress, and all died within 6 days post-infection. Our findings demonstrate that GC is a potent mucosal vaccine adjuvant against SARS-CoV-2 and potentially other respiratory viruses.
Collapse
Affiliation(s)
- Ashley R Hoover
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA; Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Sunil More
- Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK USA
| | - Kaili Liu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Connor L West
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Trisha I Valerio
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Coline L Furrer
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Jacob P Adams
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Ningli Yu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Crystal Villalva
- Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK USA
| | - Amit Kumar
- Biogen Inc., 225 Bnney Street, Cambridge, MA, USA
| | - Lu Alleruzzo
- Immunophotonics, Inc., 4340 Duncan Avenue, Suite 212, Saint Louis, MO, USA
| | - Samuel S K Lam
- Immunophotonics, Inc., 4340 Duncan Avenue, Suite 212, Saint Louis, MO, USA
| | - Tomas Hode
- Immunophotonics, Inc., 4340 Duncan Avenue, Suite 212, Saint Louis, MO, USA
| | - James F Papin
- Department Pathology and Division of Comparative Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Wei R Chen
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA.
| |
Collapse
|
12
|
Ren J, Wang S, Zong Z, Pan T, Liu S, Mao W, Huang H, Yan X, Yang B, He X, Zhou F, Zhang L. TRIM28-mediated nucleocapsid protein SUMOylation enhances SARS-CoV-2 virulence. Nat Commun 2024; 15:244. [PMID: 38172120 PMCID: PMC10764958 DOI: 10.1038/s41467-023-44502-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 12/15/2023] [Indexed: 01/05/2024] Open
Abstract
Viruses, as opportunistic intracellular parasites, hijack the cellular machinery of host cells to support their survival and propagation. Numerous viral proteins are subjected to host-mediated post-translational modifications. Here, we demonstrate that the SARS-CoV-2 nucleocapsid protein (SARS2-NP) is SUMOylated on the lysine 65 residue, which efficiently mediates SARS2-NP's ability in homo-oligomerization, RNA association, liquid-liquid phase separation (LLPS). Thereby the innate antiviral immune response is suppressed robustly. These roles can be achieved through intermolecular association between SUMO conjugation and a newly identified SUMO-interacting motif in SARS2-NP. Importantly, the widespread SARS2-NP R203K mutation gains a novel site of SUMOylation which further increases SARS2-NP's LLPS and immunosuppression. Notably, the SUMO E3 ligase TRIM28 is responsible for catalyzing SARS2-NP SUMOylation. An interfering peptide targeting the TRIM28 and SARS2-NP interaction was screened out to block SARS2-NP SUMOylation and LLPS, and consequently inhibit SARS-CoV-2 replication and rescue innate antiviral immunity. Collectively, these data support SARS2-NP SUMOylation is critical for SARS-CoV-2 virulence, and therefore provide a strategy to antagonize SARS-CoV-2.
Collapse
Affiliation(s)
- Jiang Ren
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Shuai Wang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, China
| | - Zhi Zong
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Ting Pan
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Sijia Liu
- International Biomed-X Research Center, Second Affiliated Hospital of Zhejiang University, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Wei Mao
- Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Huizhe Huang
- Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Xiaohua Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330031, China
| | - Bing Yang
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
- Department of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California, San Francisco, CA, 94158, USA
| | - Xin He
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Fangfang Zhou
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, China.
| | - Long Zhang
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China.
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
13
|
Chatterjee S, Zaia J. Proteomics-based mass spectrometry profiling of SARS-CoV-2 infection from human nasopharyngeal samples. MASS SPECTROMETRY REVIEWS 2024; 43:193-229. [PMID: 36177493 PMCID: PMC9538640 DOI: 10.1002/mas.21813] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 05/12/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of the on-going global pandemic of coronavirus disease 2019 (COVID-19) that continues to pose a significant threat to public health worldwide. SARS-CoV-2 encodes four structural proteins namely membrane, nucleocapsid, spike, and envelope proteins that play essential roles in viral entry, fusion, and attachment to the host cell. Extensively glycosylated spike protein efficiently binds to the host angiotensin-converting enzyme 2 initiating viral entry and pathogenesis. Reverse transcriptase polymerase chain reaction on nasopharyngeal swab is the preferred method of sample collection and viral detection because it is a rapid, specific, and high-throughput technique. Alternate strategies such as proteomics and glycoproteomics-based mass spectrometry enable a more detailed and holistic view of the viral proteins and host-pathogen interactions and help in detection of potential disease markers. In this review, we highlight the use of mass spectrometry methods to profile the SARS-CoV-2 proteome from clinical nasopharyngeal swab samples. We also highlight the necessity for a comprehensive glycoproteomics mapping of SARS-CoV-2 from biological complex matrices to identify potential COVID-19 markers.
Collapse
Affiliation(s)
- Sayantani Chatterjee
- Department of Biochemistry, Center for Biomedical Mass SpectrometryBoston University School of MedicineBostonMassachusettsUSA
| | - Joseph Zaia
- Department of Biochemistry, Center for Biomedical Mass SpectrometryBoston University School of MedicineBostonMassachusettsUSA
- Bioinformatics ProgramBoston University School of MedicineBostonMassachusettsUSA
| |
Collapse
|
14
|
Gissot L, Fontaine F, Kelemen Z, Dao O, Bouchez I, Deruyffelaere C, Winkler M, Costa AD, Pierre F, Meziadi C, Faure JD, Froissard M. E and M SARS-CoV-2 membrane protein expression and enrichment with plant lipid droplets. Biotechnol J 2024; 19:e2300512. [PMID: 37986207 DOI: 10.1002/biot.202300512] [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: 09/26/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023]
Abstract
Plants are gaining traction as a cost-effective and scalable platform for producing recombinant proteins. However, expressing integral membrane proteins in plants is challenging due to their hydrophobic nature. In our study, we used transient and stable expression systems in Nicotiana benthamiana and Camelina sativa respectively to express SARS-CoV-2 E and M integral proteins, and target them to lipid droplets (LDs). LDs offer an ideal environment for folding hydrophobic proteins and aid in their purification through flotation. We tested various protein fusions with different linkers and tags and used three dimensional structure predictions to assess their effects. E and M mostly localized in the ER in N. benthamiana leaves but E could be targeted to LDs in oil accumulating tobacco when fused with oleosin, a LD integral protein. In Camelina sativa seeds, E and M were however found associated with purified LDs. By enhancing the accumulation of E and M within LDs through oleosin, we enriched these proteins in the purified floating fraction. This strategy provides an alternative approach for efficiently producing and purifying hydrophobic pharmaceuticals and vaccines using plant systems.
Collapse
Affiliation(s)
- Lionel Gissot
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin (IJPB), Versailles, France
| | - Florent Fontaine
- SAS Core Biogenesis, 850 Bd Sébastien Brant BioParc 3, 67400, Illkirch-Graffenstaden, France
| | - Zsolt Kelemen
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin (IJPB), Versailles, France
| | - Ousmane Dao
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin (IJPB), Versailles, France
| | - Isabelle Bouchez
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin (IJPB), Versailles, France
| | - Carine Deruyffelaere
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin (IJPB), Versailles, France
| | - Michèle Winkler
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin (IJPB), Versailles, France
| | - Anais Da Costa
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin (IJPB), Versailles, France
| | - Fabienne Pierre
- SAS Core Biogenesis, 850 Bd Sébastien Brant BioParc 3, 67400, Illkirch-Graffenstaden, France
| | - Chouaib Meziadi
- SAS Core Biogenesis, 850 Bd Sébastien Brant BioParc 3, 67400, Illkirch-Graffenstaden, France
| | - Jean-Denis Faure
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin (IJPB), Versailles, France
| | - Marine Froissard
- Université Paris-Saclay, INRAE, AgroParisTech, Institut Jean-Pierre Bourgin (IJPB), Versailles, France
| |
Collapse
|
15
|
Ahmed-Hassan H, Farouk MM, Ali ME, Elsafiee EA, Hagag N, Abdelkader F. SARS-CoV-2 seroprevalence determination in pets and camels in Egypt using multispecies enzyme-linked immunosorbent assay. Vet Immunol Immunopathol 2024; 267:110683. [PMID: 38061231 DOI: 10.1016/j.vetimm.2023.110683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/29/2023] [Accepted: 11/16/2023] [Indexed: 01/03/2024]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has translated into a worldwide economic recession and public health crisis. Bats have been incriminated as the main natural host for the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is the causative agent of the COVID-19 pandemic. However, the reservoir and carrier hosts of the virus remain unknown. Therefore, a cross sectional serosurvey study was performed to estimate antibodies to SARS-CoV-2. To assess IgM antibodies to SARS-CoV-2 nucleocapsid protein (NP), a SARS-CoV-2 Double Antigen Multispecies diagnostic enzyme-linked immunosorbent assay kit was used. The seropositive samples were confirmed and validated by measuring IgG antibody titers in sera. The enrolled animals were from different locations in the Giza governorate, Egypt, and were sampled at the time of the pandemic; they comprised 92 companion animals and 92 domestic camels. The study established that 4.76% (1/21 clinical samples) of dogs, 7.69% of cats (1/13 shelter samples) and 1.08% (1/92) of camels, had measurable SARS-CoV-2 NP IgM antibodies. All IgM-seropositive samples were IgG positive with a measurable titer of 34.5, 28.6, and 25.8 UI/mL for dog, cat, and camels, respectively. According to our best knowledge, this study was the first to assess SARS-CoV-2 seroprevalence in the specific animals investigated in Egypt. These results may herald a promising epidemiological role for pet animals and camels in SARS-CoV-2 virus maintenance. Thus, our study's results ought to be confirmed with a nationwide seroprevalence study, and further studies are required to clarify whether these animals act as active or passive carriers.
Collapse
Affiliation(s)
- Hanaa Ahmed-Hassan
- Department of Zoonoses, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Manar M Farouk
- Department of Internal Medicine and Infectious Diseases, Faculty of Veterinary medicine, Cairo University, Giza 12211, Egypt
| | - M E Ali
- Department of Internal Medicine and Infectious Diseases, Faculty of Veterinary medicine, Cairo University, Giza 12211, Egypt
| | - Esraa A Elsafiee
- Department of Zoonoses, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt.
| | - Naglaa Hagag
- Genome Research Unit, Animal Health Research Institute, Dokki 12618, Egypt; Gene Analysis Unit in National Laboratory for Veterinary Quality Control on Poultry Production (NLQP), Animal Health Research Institute, Dokki 12618, Egypt
| | - Fatma Abdelkader
- Department of Zoonoses, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| |
Collapse
|
16
|
Mahdi PDBM, Almukhtar DM. Role Of Vaccines Against COVID-19 Pandemic. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2023; 28:355-357. [PMID: 37473842 DOI: 10.1016/j.slasd.2023.07.002] [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: 01/07/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/22/2023]
Abstract
Coronaviruses (CoV) are one of the largest families of viruses that infect human beings causing mild common cold or severe diseases like Middle East Respiratory Syndrome (MERS-CoV), and Severe Acute Respiratory Syndrome (SARS-CoV). A new strain emerged known as novel coronavirus (nCoV) causing fatal respiratory failure disease. This virus was characterized by rapid spread from asymptomatic and symptomatic patients to healthy people. Thus, vaccine should be considered as one of the important protective measures to control the spread of this virus. One of the challenges to this vaccine is the high mutation rate of this virus and appearance of new strains. Therefore, vaccine should stimulate the immune system in order to overcome the emergence of new strain of this virus.
Collapse
Affiliation(s)
- Professor Dr Batool Mutar Mahdi
- Consultant Clinical Immunology, Head of HLA Research Unit, Department of Microbiology, Al-Kindy College of Medicine, University of Baghdad, Baghdad, Iraq.
| | | |
Collapse
|
17
|
Mabrouk MT, Zidan AA, Aly N, Mohammed MT, Ghantous F, Seaman MS, Lovell JF, Nasr ML. Circularized Nanodiscs for Multivalent Mosaic Display of SARS-CoV-2 Spike Protein Antigens. Vaccines (Basel) 2023; 11:1655. [PMID: 38005987 PMCID: PMC10675430 DOI: 10.3390/vaccines11111655] [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: 09/29/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
The emergence of vaccine-evading SARS-CoV-2 variants urges the need for vaccines that elicit broadly neutralizing antibodies (bnAbs). Here, we assess covalently circularized nanodiscs decorated with recombinant SARS-CoV-2 spike glycoproteins from several variants for eliciting bnAbs with vaccination. Cobalt porphyrin-phospholipid (CoPoP) was incorporated into the nanodisc to allow for anchoring and functional orientation of spike trimers on the nanodisc surface through their His-tag. Monophosphoryl-lipid (MPLA) and QS-21 were incorporated as immunostimulatory adjuvants to enhance vaccine responses. Following optimization of nanodisc assembly, spike proteins were effectively displayed on the surface of the nanodiscs and maintained their conformational capacity for binding with human angiotensin-converting enzyme 2 (hACE2) as verified using electron microscopy and slot blot assay, respectively. Six different formulations were prepared where they contained mono antigens; four from the year 2020 (WT, Beta, Lambda, and Delta) and two from the year 2021 (Omicron BA.1 and BA.2). Additionally, we prepared a mosaic nanodisc displaying the four spike proteins from year 2020. Intramuscular vaccination of CD-1 female mice with the mosaic nanodisc induced antibody responses that not only neutralized matched pseudo-typed viruses, but also neutralized mismatched pseudo-typed viruses corresponding to later variants from year 2021 (Omicron BA.1 and BA.2). Interestingly, sera from mosaic-immunized mice did not effectively inhibit Omicron spike binding to human ACE-2, suggesting that some of the elicited antibodies were directed towards conserved neutralizing epitopes outside the receptor binding domain. Our results show that mosaic nanodisc vaccine displaying spike proteins from 2020 can elicit broadly neutralizing antibodies that can neutralize mismatched viruses from a following year, thus decreasing immune evasion of new emerging variants and enhancing healthcare preparedness.
Collapse
Affiliation(s)
- Moustafa T. Mabrouk
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.T.M.)
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA;
| | - Asmaa A. Zidan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Nihal Aly
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.T.M.)
- Botany and Microbiology Department, Faculty of Science, Alexandria University, Alexandria 21526, Egypt
| | - Mostafa T. Mohammed
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.T.M.)
- Clinical Pathology Department, Minia University, Minia 61519, Egypt
| | - Fadi Ghantous
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Michael S. Seaman
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Jonathan F. Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA;
| | - Mahmoud L. Nasr
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.T.M.)
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
18
|
Goswami A, Kumar M, Ullah S, Gore MM. De novo design of anti-variant COVID-19 vaccine. Biol Methods Protoc 2023; 8:bpad021. [PMID: 37854896 PMCID: PMC10580973 DOI: 10.1093/biomethods/bpad021] [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: 07/28/2023] [Revised: 09/07/2023] [Accepted: 09/21/2023] [Indexed: 10/20/2023] Open
Abstract
Recent studies highlight the effectiveness of hybrid Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) vaccines combining wild-type nucleocapsid and Spike proteins. We have further enhanced this strategy by incorporating delta and omicron variants' spike protein mutations. Both delta and omicron mark the shifts in viral transmissibility and severity in unvaccinated and vaccinated patients. So their mutations are highly crucial for future viral variants also. Omicron is particularly adept at immune evasion by mutating spike epitopes. The rapid adaptations of Omicron and sub-variants to spike-based vaccines and simultaneous transmissibility underline the urgency for new vaccines in the continuous battle against SARS-CoV-2. Therefore, we have added three persistent T-cell-stimulating nucleocapsid peptides similar to homologous sequences from seasonal Human Coronaviruses (HuCoV) and an envelope peptide that elicits a strong T-cell immune response. These peptides are clustered in the hybrid spike's cytoplasmic region with non-immunogenic linkers, enabling systematic arrangement. AlphaFold (Artificial intelligence-based model building) analysis suggests omitting the transmembrane domain enhances these cytoplasmic epitopes' folding efficiency which can ensure persistent immunity for CD4+ structural epitopes. Further molecular dynamics simulations validate the compact conformation of the modeled structures and a flexible C-terminus region. Overall, the structures show stability and less conformational fluctuation throughout the simulation. Also, the AlphaFold predicted structural epitopes maintained their folds during simulation to ensure the specificity of CD4+ T-cell response after vaccination. Our proposed approach may provide options for incorporating diverse anti-viral T-cell peptides, similar to HuCoV, into linker regions. This versatility can be promising to address outbreaks and challenges posed by various viruses for effective management in this era of innovative vaccines.
Collapse
Affiliation(s)
- Arpita Goswami
- Kshamalab, Leo’s Research Services and Suppliers, Mysuru 570016, India
| | - Madan Kumar
- Department of Chemistry-BMC Biochemistry, University of Uppsala, Uppsala 75237, Sweden
| | - Samee Ullah
- National Center for Bioinformatics (NCB), Islamabad 45320, Pakistan
| | - Milind M Gore
- 5/1B, Krutika Co-Op Housing Society, Kothrud, Pune 411039, India
| |
Collapse
|
19
|
Cao L, Kong X, Li X, Suo X, Duan Y, Yuan C, Zhang Y, Zheng H, Wang Q. A Customized Novel Blocking ELISA for Detection of Bat-Origin Swine Acute Diarrhea Syndrome Coronavirus Infection. Microbiol Spectr 2023; 11:e0393022. [PMID: 37272819 PMCID: PMC10434073 DOI: 10.1128/spectrum.03930-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 05/09/2023] [Indexed: 06/06/2023] Open
Abstract
Swine acute diarrhea syndrome coronavirus (SADS-CoV) is a newly discovered emerging alphacoronavirus. SADS-CoV shares over 90% genome sequence identity with bat alphacoronavirus HKU2. SADS-CoV was associated with severe diarrhea and high mortality rates in piglets. Accurate serological diagnosis of SADS-CoV infection is key in managing the emerging SADS-CoV. However, thus far there have been no effective antibody-based diagnostic tests for diagnose of SADS-CoV exposure. Here, monoclonal antibody (MAb) 6E8 against SADS-CoV N protein accurately recognized SADS-CoV infection. Then, MAb 6E8 was utilized as a blocking antibody to develop blocking ELISA (bELISA). We customized the rN coating antigen with concentration 0.25 μg/mL. According to receiver operator characteristic curve analysis, the cutoff value of the bELISA was determined as 38.19% when the max Youden index was 0.955, and specificity was 100%, and sensitivity was 95.5%. Specificity testing showed that there was no cross-reactivity with other serum positive swine enteric coronaviruses, such as porcine epidemic diarrhea virus (PEDV), transmissible gastroenteritis virus (TGEV), porcine deltacoronavirus (PDCoV), porcine rotavirus (PoRV), and porcine sapelovirus (PSV). In conclusion, we customized a novel and high-quality blocking ELISA for detection of SADS-CoV infection, and the current bELISA will be linked to a clinical and epidemiological assessment of SADS-CoV infection. IMPORTANCE SADS-CoV was reported to be of high potential for dissemination among various of host species. Accurate serological diagnosis of SADS-CoV infection is key in managing the emerging SADS-CoV. However, thus far there have been no effective antibody-based diagnostic tests for diagnose of SADS-CoV exposure. We customed a novel and high-quality bELISA assay for detection of SADS-CoV N protein antibodies, and the current bELISA will be linked to a clinical and epidemiological assessment of SADS-CoV infection.
Collapse
Affiliation(s)
- Liyan Cao
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, National Foot and Mouth Diseases Reference Laboratory, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Chengdu National Agricultural Science and Technology Center, Chengdu, China
| | - Xiangyu Kong
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, National Foot and Mouth Diseases Reference Laboratory, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Chengdu National Agricultural Science and Technology Center, Chengdu, China
| | - Xiangtong Li
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, National Foot and Mouth Diseases Reference Laboratory, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Chengdu National Agricultural Science and Technology Center, Chengdu, China
| | - Xuepeng Suo
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, National Foot and Mouth Diseases Reference Laboratory, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Chengdu National Agricultural Science and Technology Center, Chengdu, China
| | - Yueyue Duan
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, National Foot and Mouth Diseases Reference Laboratory, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Chengdu National Agricultural Science and Technology Center, Chengdu, China
| | - Cong Yuan
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, National Foot and Mouth Diseases Reference Laboratory, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Chengdu National Agricultural Science and Technology Center, Chengdu, China
| | - Yu Zhang
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, National Foot and Mouth Diseases Reference Laboratory, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Chengdu National Agricultural Science and Technology Center, Chengdu, China
| | - Haixue Zheng
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, National Foot and Mouth Diseases Reference Laboratory, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Qi Wang
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, National Foot and Mouth Diseases Reference Laboratory, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Chengdu National Agricultural Science and Technology Center, Chengdu, China
| |
Collapse
|
20
|
Kakavandi S, Zare I, VaezJalali M, Dadashi M, Azarian M, Akbari A, Ramezani Farani M, Zalpoor H, Hajikhani B. Structural and non-structural proteins in SARS-CoV-2: potential aspects to COVID-19 treatment or prevention of progression of related diseases. Cell Commun Signal 2023; 21:110. [PMID: 37189112 PMCID: PMC10183699 DOI: 10.1186/s12964-023-01104-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 03/15/2023] [Indexed: 05/17/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by a new member of the Coronaviridae family known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). There are structural and non-structural proteins (NSPs) in the genome of this virus. S, M, H, and E proteins are structural proteins, and NSPs include accessory and replicase proteins. The structural and NSP components of SARS-CoV-2 play an important role in its infectivity, and some of them may be important in the pathogenesis of chronic diseases, including cancer, coagulation disorders, neurodegenerative disorders, and cardiovascular diseases. The SARS-CoV-2 proteins interact with targets such as angiotensin-converting enzyme 2 (ACE2) receptor. In addition, SARS-CoV-2 can stimulate pathological intracellular signaling pathways by triggering transcription factor hypoxia-inducible factor-1 (HIF-1), neuropilin-1 (NRP-1), CD147, and Eph receptors, which play important roles in the progression of neurodegenerative diseases like Alzheimer's disease, epilepsy, and multiple sclerosis, and multiple cancers such as glioblastoma, lung malignancies, and leukemias. Several compounds such as polyphenols, doxazosin, baricitinib, and ruxolitinib could inhibit these interactions. It has been demonstrated that the SARS-CoV-2 spike protein has a stronger affinity for human ACE2 than the spike protein of SARS-CoV, leading the current study to hypothesize that the newly produced variant Omicron receptor-binding domain (RBD) binds to human ACE2 more strongly than the primary strain. SARS and Middle East respiratory syndrome (MERS) viruses against structural and NSPs have become resistant to previous vaccines. Therefore, the review of recent studies and the performance of current vaccines and their effects on COVID-19 and related diseases has become a vital need to deal with the current conditions. This review examines the potential role of these SARS-CoV-2 proteins in the initiation of chronic diseases, and it is anticipated that these proteins could serve as components of an effective vaccine or treatment for COVID-19 and related diseases. Video Abstract.
Collapse
Affiliation(s)
- Sareh Kakavandi
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Iman Zare
- Research and Development Department, Sina Medical Biochemistry Technologies Co. Ltd., Shiraz, 7178795844, Iran
| | - Maryam VaezJalali
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Dadashi
- Department of Microbiology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Maryam Azarian
- Department of Radiology, Charité - Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Abdullatif Akbari
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Marzieh Ramezani Farani
- Department of Biological Sciences and Bioengineering, Nano Bio High-Tech Materials Research Center, Inha University, Incheon, 22212, Republic of Korea
| | - Hamidreza Zalpoor
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Bahareh Hajikhani
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
21
|
Rabdano SO, Ruzanova EA, Pletyukhina IV, Saveliev NS, Kryshen KL, Katelnikova AE, Beltyukov PP, Fakhretdinova LN, Safi AS, Rudakov GO, Arakelov SA, Andreev IV, Kofiadi IA, Khaitov MR, Valenta R, Kryuchko DS, Berzin IA, Belozerova NS, Evtushenko AE, Truhin VP, Skvortsova VI. Immunogenicity and In Vivo Protective Effects of Recombinant Nucleocapsid-Based SARS-CoV-2 Vaccine Convacell ®. Vaccines (Basel) 2023; 11:vaccines11040874. [PMID: 37112786 PMCID: PMC10141225 DOI: 10.3390/vaccines11040874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
The vast majority of SARS-CoV-2 vaccines which are licensed or under development focus on the spike (S) protein and its receptor binding domain (RBD). However, the S protein shows considerable sequence variations among variants of concern. The aim of this study was to develop and characterize a SARS-CoV-2 vaccine targeting the highly conserved nucleocapsid (N) protein. Recombinant N protein was expressed in Escherichia coli, purified to homogeneity by chromatography and characterized by SDS-PAGE, immunoblotting, mass spectrometry, dynamic light scattering and differential scanning calorimetry. The vaccine, formulated as a squalane-based emulsion, was used to immunize Balb/c mice and NOD SCID gamma (NSG) mice engrafted with human PBMCs, rabbits and marmoset monkeys. Safety and immunogenicity of the vaccine was assessed via ELISA, cytokine titer assays and CFSE dilution assays. The protective effect of the vaccine was studied in SARS-CoV-2-infected Syrian hamsters. Immunization induced sustainable N-specific IgG responses and an N-specific mixed Th1/Th2 cytokine response. In marmoset monkeys, an N-specific CD4+/CD8+ T cell response was observed. Vaccinated Syrian hamsters showed reduced lung histopathology, lower virus proliferation, lower lung weight relative to the body, and faster body weight recovery. Convacell® thus is shown to be effective and may augment the existing armamentarium of vaccines against COVID-19.
Collapse
Affiliation(s)
- Sevastyan O Rabdano
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Ellina A Ruzanova
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Iuliia V Pletyukhina
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Nikita S Saveliev
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | | | | | - Petr P Beltyukov
- Scientific Research Institute of Hygiene, Occupational Pathology and Human Ecology of the Federal Medical-Biological Agency of Russia (SRIHOPHE), Kuzmolovsky 188663, Russia
| | - Liliya N Fakhretdinova
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Ariana S Safi
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - German O Rudakov
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Sergei A Arakelov
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Igor V Andreev
- National Research Center Institute of Immunology (NRCII), Federal Medical-Biological Agency of Russia, Moscow 115522, Russia
| | - Ilya A Kofiadi
- National Research Center Institute of Immunology (NRCII), Federal Medical-Biological Agency of Russia, Moscow 115522, Russia
- Department of Immunology, N.I. Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow 117997, Russia
| | - Musa R Khaitov
- National Research Center Institute of Immunology (NRCII), Federal Medical-Biological Agency of Russia, Moscow 115522, Russia
- Department of Immunology, N.I. Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow 117997, Russia
| | - Rudolf Valenta
- National Research Center Institute of Immunology (NRCII), Federal Medical-Biological Agency of Russia, Moscow 115522, Russia
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- Laboratory of Immunopathology, Department of Clinical Immunology and Allergology, I.M. Sechenov First Moscow State Medical University, Moscow 119435, Russia
- Karl Landsteiner University of Health Sciences, 3500 Krems, Austria
| | - Daria S Kryuchko
- Federal Medical-Biological Agency of Russia, Moscow 125310, Russia
| | - Igor A Berzin
- Federal Medical-Biological Agency of Russia, Moscow 125310, Russia
| | - Natalia S Belozerova
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Anatoly E Evtushenko
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Viktor P Truhin
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | | |
Collapse
|
22
|
Bonam SR, Hu H. Next-Generation Vaccines Against COVID-19 Variants: Beyond the Spike Protein. ZOONOSES (BURLINGTON, MASS.) 2023; 3:10.15212/zoonoses-2023-0003. [PMID: 38031548 PMCID: PMC10686570 DOI: 10.15212/zoonoses-2023-0003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
Vaccines are among the most effective medical countermeasures against infectious diseases. The current Coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spurred the scientific strategies to fight against the disease. Since 2020, a great number of vaccines based on different platforms have been in development in response to the pandemic, among which mRNA, adenoviral vector, and subunit vaccines have been clinically approved for use in humans. These first-generation COVID-19 vaccines largely target the viral spike (S) protein and aim for eliciting potent neutralizing antibodies. With the emergence of SARS-CoV-2 variants, especially the highly transmissible Omicron strains, the S-based vaccine strategies have been faced constant challenges due to strong immune escape by the variants. The coronavirus nucleocapsid (N) is one of the viral proteins that induces strong T-cell immunity and is more conserved across different SARS-CoV-2 variants. Inclusion of N in the development of COVID-19 vaccines has been reported. Here, we briefly reviewed and discussed COVID-19 disease, current S-based vaccine strategies, and focused on the immunobiology of N protein in SARS-CoV-2 host immunity, as well as the next-generation vaccine strategies involving N protein, to combat current and emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA 77555
| | - Haitao Hu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA 77555
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA 77555
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA 77555
| |
Collapse
|
23
|
Spicuzza L, Campagna D, Di Maria C, Sciacca E, Mancuso S, Vancheri C, Sambataro G. An update on lateral flow immunoassay for the rapid detection of SARS-CoV-2 antibodies. AIMS Microbiol 2023; 9:375-401. [PMID: 37091823 PMCID: PMC10113162 DOI: 10.3934/microbiol.2023020] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/25/2023] Open
Abstract
Over the last three years, after the outbreak of the COVID-19 pandemic, an unprecedented number of novel diagnostic tests have been developed. Assays to evaluate the immune response to SARS-CoV-2 have been widely considered as part of the control strategy. The lateral flow immunoassay (LFIA), to detect both IgM and IgG against SARS-CoV-2, has been widely studied as a point-of-care (POC) test. Compared to laboratory tests, LFIAs are faster, cheaper and user-friendly, thus available also in areas with low economic resources. Soon after the onset of the pandemic, numerous kits for rapid antibody detection were put on the market with an emergency use authorization. However, since then, scientists have tried to better define the accuracy of these tests and their usefulness in different contexts. In fact, while during the first phase of the pandemic LFIAs for antibody detection were auxiliary to molecular tests for the diagnosis of COVID-19, successively these tests became a tool of seroprevalence surveillance to address infection control policies. When in 2021 a massive vaccination campaign was implemented worldwide, the interest in LFIA reemerged due to the need to establish the extent and the longevity of immunization in the vaccinated population and to establish priorities to guide health policies in low-income countries with limited access to vaccines. Here, we summarize the accuracy, the advantages and limits of LFIAs as POC tests for antibody detection, highlighting the efforts that have been made to improve this technology over the last few years.
Collapse
Affiliation(s)
- Lucia Spicuzza
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | | | | | | | | | | | | |
Collapse
|
24
|
Benabdessalem C, Hamouda WB, Marzouki S, Faye R, Mbow AA, Diouf B, Ndiaye O, Dia N, Faye O, Sall AA, Diagne CT, Amellal H, Ezzikouri S, Mioramalala DJN, Randrianarisaona F, Trabelsi K, Boumaiza M, Hamouda SB, Ouni R, Bchiri S, Chaaban A, Gdoura M, Gorgi Y, Sfar I, Yalaoui S, Khelil JB, Hamzaoui A, Abdallah M, Cherif Y, Petres S, Mok CKP, Escriou N, Quesney S, Dellagi K, Schoenhals M, Sarih M, Vigan-Womas I, Bettaieb J, Rourou S, Barbouche MR, Ahmed MB. Development and comparative evaluation of SARS-CoV-2 S-RBD and N based ELISA tests in various African endemic settings. Diagn Microbiol Infect Dis 2023; 105:115903. [PMID: 36805620 PMCID: PMC9867841 DOI: 10.1016/j.diagmicrobio.2023.115903] [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: 11/15/2022] [Revised: 01/08/2023] [Accepted: 01/18/2023] [Indexed: 01/23/2023]
Abstract
Management of the COVID-19 pandemic relies on molecular diagnostic methods supported by serological tools. Herein, we developed S-RBD- and N- based ELISA assays useful for infection rate surveillance as well as the follow-up of acquired protective immunity against SARS-CoV-2. ELISA assays were optimized using COVID-19 Tunisian patients' sera and prepandemic controls. Assays were further validated in 3 African countries with variable endemic settings. The receiver operating curve was used to evaluate the assay performances. The N- and S-RBD-based ELISA assays performances, in Tunisia, were very high (AUC: 0.966 and 0.98, respectively, p < 0.0001). Cross-validation analysis showed similar performances in different settings. Cross-reactivity, with malaria infection, against viral antigens, was noticed. In head-to-head comparisons with different commercial assays, the developed assays showed high agreement. This study demonstrates, the added value of the developed serological assays in low-income countries, particularly in ethnically diverse populations with variable exposure to local endemic infectious diseases.
Collapse
Affiliation(s)
- Chaouki Benabdessalem
- Laboratory of Transmission, Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunisia; University Tunis El Manar, Tunis, Tunisia.
| | - Wafa Ben Hamouda
- Laboratory of Transmission, Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunisia; University Tunis El Manar, Tunis, Tunisia
| | - Soumaya Marzouki
- Laboratory of Transmission, Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunisia; University Tunis El Manar, Tunis, Tunisia
| | | | | | | | | | - Ndongo Dia
- Institut Pasteur de Dakar, Dakar, Senegal
| | | | | | | | - Houda Amellal
- Department of Parasitology and Vectorial Diseases. Institut Pasteur du Maroc. Casablanca. Morocco
| | - Sayeh Ezzikouri
- Department of Parasitology and Vectorial Diseases. Institut Pasteur du Maroc. Casablanca. Morocco
| | | | | | - Khaled Trabelsi
- University Tunis El Manar, Tunis, Tunisia; Laboratory of Molecular Microbiology, Vaccinology, and Biotechnological Development, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Mohamed Boumaiza
- University Tunis El Manar, Tunis, Tunisia; Laboratory of Molecular Microbiology, Vaccinology, and Biotechnological Development, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Sonia Ben Hamouda
- Laboratory of Transmission, Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunisia; University Tunis El Manar, Tunis, Tunisia
| | - Rym Ouni
- Laboratory of Transmission, Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunisia; University Tunis El Manar, Tunis, Tunisia
| | - Soumaya Bchiri
- Laboratory of Transmission, Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunisia; University Tunis El Manar, Tunis, Tunisia
| | - Amani Chaaban
- University Tunis El Manar, Tunis, Tunisia; Laboratory of Molecular Microbiology, Vaccinology, and Biotechnological Development, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Mariem Gdoura
- Laboratory of Clinical Virology, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Yousr Gorgi
- University Tunis El Manar, Tunis, Tunisia; Laboratory of Immunology, Charles Nicolle Hospital, Tunis, Tunisia
| | - Imen Sfar
- University Tunis El Manar, Tunis, Tunisia; Laboratory of Immunology, Charles Nicolle Hospital, Tunis, Tunisia
| | - Sadok Yalaoui
- University Tunis El Manar, Tunis, Tunisia; Laboratory of Immunology, Abderrahmen Mami University Hospital, Ariana, Tunisia
| | - Jalila Ben Khelil
- University Tunis El Manar, Tunis, Tunisia; Intensive care unit, Abderrahmen Mami University Hospital, Ariana, Tunisia
| | - Agnes Hamzaoui
- University Tunis El Manar, Tunis, Tunisia; Pneumology Department Abderrahmen Mami University Hospital, Ariana, Tunisia
| | - Meya Abdallah
- University Tunis El Manar, Tunis, Tunisia; Department of internal Medicine, Yasminette Hospital, Ben Arous, Tunisia
| | - Yosra Cherif
- University Tunis El Manar, Tunis, Tunisia; Department of internal Medicine, Yasminette Hospital, Ben Arous, Tunisia
| | | | - Chris Ka Pun Mok
- HKU-Pasteur Research Pole, School of Public Health, University of Hong Kong, Hong Kong; The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong
| | | | | | - Koussay Dellagi
- Institut Pasteur de Paris, Paris, France; Pasteur Network, Institut Pasteur, Paris, France
| | - Matthieu Schoenhals
- Immunology of Infectious Diseases, Institut Pasteur of Madagascar, Antananarivo, Madagascar
| | - M'hammed Sarih
- Department of Parasitology and Vectorial Diseases. Institut Pasteur du Maroc. Casablanca. Morocco
| | | | - Jihene Bettaieb
- Laboratory of Transmission, Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunisia; University Tunis El Manar, Tunis, Tunisia
| | - Samia Rourou
- University Tunis El Manar, Tunis, Tunisia; Laboratory of Molecular Microbiology, Vaccinology, and Biotechnological Development, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Mohamed Ridha Barbouche
- Laboratory of Transmission, Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunisia; University Tunis El Manar, Tunis, Tunisia; Department of Microbiology, Immunology, and infectious diseases. College of Medicine and Medical sciences, Arabian Gulf University, Manama, Bahrain
| | - Melika Ben Ahmed
- Laboratory of Transmission, Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunisia; University Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
25
|
Soares SR, da Silva Torres MK, Lima SS, de Sarges KML, Santos EFD, de Brito MTFM, da Silva ALS, de Meira Leite M, da Costa FP, Cantanhede MHD, da Silva R, de Oliveira Lameira Veríssimo A, Vallinoto IMVC, Feitosa RNM, Quaresma JAS, Chaves TDSS, Viana GMR, Falcão LFM, Santos EJMD, Vallinoto ACR, da Silva ANMR. Antibody Response to the SARS-CoV-2 Spike and Nucleocapsid Proteins in Patients with Different COVID-19 Clinical Profiles. Viruses 2023; 15:v15040898. [PMID: 37112878 PMCID: PMC10141342 DOI: 10.3390/v15040898] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/30/2023] [Accepted: 03/30/2023] [Indexed: 04/03/2023] Open
Abstract
The first case of coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), in Brazil was diagnosed on February 26, 2020. Due to the important epidemiological impact of COVID-19, the present study aimed to analyze the specificity of IgG antibody responses to the S1, S2 and N proteins of SARS-CoV-2 in different COVID-19 clinical profiles. This study enrolled 136 individuals who were diagnosed with or without COVID-19 based on clinical findings and laboratory results and classified as asymptomatic or as having mild, moderate or severe disease. Data collection was performed through a semistructured questionnaire to obtain demographic information and main clinical manifestations. IgG antibody responses to the S1 and S2 subunits of the spike (S) protein and the nucleocapsid (N) protein were evaluated using an enzyme-linked immunosorbent assay (ELISA) according to the manufacturer’s instructions. The results showed that among the participants, 87.5% (119/136) exhibited IgG responses to the S1 subunit and 88.25% (120/136) to N. Conversely, only 14.44% of the subjects (21/136) displayed S2 subunit responses. When analyzing the IgG antibody response while considering the different proteins of the virus, patients with severe disease had significantly higher antibody responses to N and S1 than asymptomatic individuals (p ≤ 0.0001), whereas most of the participants had low antibody titers against the S2 subunit. In addition, individuals with long COVID-19 showed a greater IgG response profile than those with symptomatology of a short duration. Based on the results of this study, it is concluded that levels of IgG antibodies may be related to the clinical evolution of COVID-19, with high levels of IgG antibodies against S1 and N in severe cases and in individuals with long COVID-19.
Collapse
Affiliation(s)
- Sinei Ramos Soares
- Laboratório de Virologia, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil
| | - Maria Karoliny da Silva Torres
- Laboratório de Virologia, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil
- Programa de Pós-Graduação em Biologia de Agentes Infecciosos e Parasitários, Universidade Federal do Pará, Belém 66075-110, Brazil
| | - Sandra Souza Lima
- Laboratório de Virologia, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil
| | - Kevin Matheus Lima de Sarges
- Laboratório de Genética de Doenças Complexas, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil
| | - Erika Ferreira dos Santos
- Laboratório de Genética de Doenças Complexas, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil
| | | | - Andréa Luciana Soares da Silva
- Laboratório de Genética de Doenças Complexas, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil
| | - Mauro de Meira Leite
- Laboratório de Genética de Doenças Complexas, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil
| | - Flávia Póvoa da Costa
- Laboratório de Genética de Doenças Complexas, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil
| | | | - Rosilene da Silva
- Laboratório de Genética de Doenças Complexas, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil
| | | | - Izaura Maria Vieira Cayres Vallinoto
- Laboratório de Virologia, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil
- Programa de Pós-Graduação em Biologia de Agentes Infecciosos e Parasitários, Universidade Federal do Pará, Belém 66075-110, Brazil
| | - Rosimar Neris Martins Feitosa
- Laboratório de Virologia, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil
- Programa de Pós-Graduação em Biologia de Agentes Infecciosos e Parasitários, Universidade Federal do Pará, Belém 66075-110, Brazil
| | - Juarez Antônio Simões Quaresma
- Programa de Pós-Graduação em Biologia de Agentes Infecciosos e Parasitários, Universidade Federal do Pará, Belém 66075-110, Brazil
- Centro de Ciências Biológicas e da Saúde, Universidade do Estado do Pará, Belém 66050-540, Brazil
| | - Tânia do Socorro Souza Chaves
- Laboratório de Pesquisas Básicas em Malária em Malária, Seção de Parasitologia, Instituto Evandro Chagas, Secretaria de Ciência, Tecnologia e Insumos Estratégicos, Ministério da Saúde do Brasil, Ananindeua 70068-900, Brazil
| | - Giselle Maria Rachid Viana
- Programa de Pós-Graduação em Biologia de Agentes Infecciosos e Parasitários, Universidade Federal do Pará, Belém 66075-110, Brazil
- Laboratório de Pesquisas Básicas em Malária em Malária, Seção de Parasitologia, Instituto Evandro Chagas, Secretaria de Ciência, Tecnologia e Insumos Estratégicos, Ministério da Saúde do Brasil, Ananindeua 70068-900, Brazil
| | - Luiz Fábio Magno Falcão
- Centro de Ciências Biológicas e da Saúde, Universidade do Estado do Pará, Belém 66050-540, Brazil
| | - Eduardo José Melo dos Santos
- Laboratório de Genética de Doenças Complexas, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil
| | | | - Andréa Nazaré Monteiro Rangel da Silva
- Laboratório de Virologia, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil
- Programa de Pós-Graduação em Biologia de Agentes Infecciosos e Parasitários, Universidade Federal do Pará, Belém 66075-110, Brazil
| |
Collapse
|
26
|
Schulte S, Huang J, Pierce NA. Hybridization Chain Reaction Lateral Flow Assays for Amplified Instrument-Free At-Home SARS-CoV-2 Testing. ACS Infect Dis 2023; 9:450-458. [PMID: 36735927 PMCID: PMC9924079 DOI: 10.1021/acsinfecdis.2c00472] [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: 09/18/2022] [Indexed: 02/05/2023]
Abstract
The lateral flow assay format enables rapid, instrument-free, at-home testing for SARS-CoV-2. Due to the absence of signal amplification, this simplicity comes at a cost in sensitivity. Here, we enhance sensitivity by developing an amplified lateral flow assay that incorporates isothermal, enzyme-free signal amplification based on the mechanism of hybridization chain reaction (HCR). The simplicity of the user experience is maintained using a disposable 3-channel lateral flow device to automatically deliver reagents to the test region in three successive stages without user interaction. To perform a test, the user loads the sample, closes the device, and reads the result by eye after 60 min. Detecting gamma-irradiated SARS-CoV-2 virions in a mixture of saliva and extraction buffer, the current amplified HCR lateral flow assay achieves a limit of detection of 200 copies/μL using available antibodies to target the SARS-CoV-2 nucleocapsid protein. By comparison, five commercial unamplified lateral flow assays that use proprietary antibodies exhibit limits of detection of 500 copies/μL, 1000 copies/μL, 2000 copies/μL, 2000 copies/μL, and 20,000 copies/μL. By swapping out antibody probes to target different pathogens, amplified HCR lateral flow assays offer a platform for simple, rapid, and sensitive at-home testing for infectious diseases. As an alternative to viral protein detection, we further introduce an HCR lateral flow assay for viral RNA detection.
Collapse
Affiliation(s)
- Samuel
J. Schulte
- Division
of Biology & Biological Engineering, California Institute of Technology, Pasadena, California 91125, United States
| | - Jining Huang
- Division
of Biology & Biological Engineering, California Institute of Technology, Pasadena, California 91125, United States
| | - Niles A. Pierce
- Division
of Biology & Biological Engineering, California Institute of Technology, Pasadena, California 91125, United States
- Division
of Engineering & Applied Science, California
Institute of Technology, Pasadena, California 91125, United States
| |
Collapse
|
27
|
Peptide microarray IgM and IgG screening of pre-SARS-CoV-2 human serum samples from Zimbabwe for reactivity with peptides from all seven human coronaviruses: a cross-sectional study. THE LANCET MICROBE 2023. [PMCID: PMC9931394 DOI: 10.1016/s2666-5247(22)00295-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
|
28
|
Bahramali G, Mashhadi Abolghasem Shirazi M, Hannan M, Aghasadeghi MR, Khosravy MS, Arjmand S, Sadat SM. Immunogenicity evaluation of a novel virus-like particle vaccine candidate against SARS-CoV-2 in BALB/c. Pathog Dis 2023; 81:ftad021. [PMID: 37667486 DOI: 10.1093/femspd/ftad021] [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: 07/19/2023] [Revised: 08/12/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023] Open
Abstract
The coronavirus disease (COVID-19) pandemic has imposed deployment of an effective vaccine as a worldwide health priority. The new variants of SARS-CoV-2 have also brought serious concerns due to virus eradiation hesitancy. In this study, we evaluated the protective immune system activity of a recombinant viral vector-based vaccine candidate encoding a fusion spike, membrane and nucleocapsid proteins, Spike (528-1273aa)-M-N, in BALB/c via two different routes of delivery, intranasal and subcutaneous. The immune responses were then assessed through specific SARS-CoV-2 antibodies, interleukin and granzyme B secretion. The outcomes showed that the IgG titer and IgA secretion was higher in intranasal route in comparison with the subcutaneous, and what is more, a higher titer of IL-4 was detected through the intranasal route, whereas IFN-γ was highly induced via the subcutaneous route. The cytotoxic cell activities were mostly achieved via subcutaneous route immunization. Vaccination with the target antigen is immunogenic and led to induction of specific antibodies. Both humoral and cellular immunity arms were well activated in immunized mice, especially through intranasal route with detectable IgA and IgG. Therefore, implication of the platform as a potential vaccine candidate has potential as a future prophylactic vaccine that guarantees further investigations for the assessment of its immunogenicity in humans.
Collapse
Affiliation(s)
- Golnaz Bahramali
- Department of Hepatitis and AIDS and Blood borne diseases, Pasteur Institute of Iran, Tehran 1316946551, Iran
| | | | - Mina Hannan
- Department of Hepatitis and AIDS and Blood borne diseases, Pasteur Institute of Iran, Tehran 1316946551, Iran
| | - Mohammad Reza Aghasadeghi
- Department of Hepatitis and AIDS and Blood borne diseases, Pasteur Institute of Iran, Tehran 1316946551, Iran
| | - Mohammad Sadeq Khosravy
- Department of Rabies Research (B), WHO Collaborating Center for Reference and Research on Rabies, Tehran 1316946551, Iran
| | - Sina Arjmand
- Department of Hepatitis and AIDS and Blood borne diseases, Pasteur Institute of Iran, Tehran 1316946551, Iran
| | - Seyed Mehdi Sadat
- Department of Hepatitis and AIDS and Blood borne diseases, Pasteur Institute of Iran, Tehran 1316946551, Iran
| |
Collapse
|
29
|
Lake CM, Breen JJ. Sequence similarity between SARS-CoV-2 nucleocapsid and multiple sclerosis-associated proteins provides insight into viral neuropathogenesis following infection. Sci Rep 2023; 13:389. [PMID: 36617594 PMCID: PMC9825799 DOI: 10.1038/s41598-022-27348-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 12/30/2022] [Indexed: 01/09/2023] Open
Abstract
The novel coronavirus SARS-CoV-2 continues to cause death and disease throughout the world, underscoring the necessity of understanding the virus and host immune response. From the start of the pandemic, a prominent pattern of central nervous system (CNS) pathologies, including demyelination, has emerged, suggesting an underlying mechanism of viral mimicry to CNS proteins. We hypothesized that immunodominant epitopes of SARS-CoV-2 share homology with proteins associated with multiple sclerosis (MS). Using PEPMatch, a newly developed bioinformatics package which predicts peptide similarity within specific amino acid mismatching parameters consistent with published MHC binding capacity, we discovered that nucleocapsid protein shares significant overlap with 22 MS-associated proteins, including myelin proteolipid protein (PLP). Further computational evaluation demonstrated that this overlap may have critical implications for T cell responses in MS patients and is likely unique to SARS-CoV-2 among the major human coronaviruses. Our findings substantiate the hypothesis of viral molecular mimicry in the pathogenesis of MS and warrant further experimental exploration.
Collapse
Affiliation(s)
- Camille M Lake
- Office of Data Science and Emerging Technologies, National Institute of Allergy and Infectious Diseases, Rockville, MD, USA.
| | - Joseph J Breen
- Division of Allergy, Immunology and Transplantation, National Institute of Allergy and Infectious Diseases, Rockville, MD, USA
| |
Collapse
|
30
|
Wey L, Masetto T, Spaeth A, Brehm J, Kochem C, Reinhart M, Müller H, Kempin U, Lorenz F, Peter C, Grimmler M. Bioinformatical Design and Performance Evaluation of a Nucleocapsid- and an RBD-Based Particle Enhanced Turbidimetric Immunoassay (PETIA) to Quantify the Wild Type and Variants of Concern-Derived Immunoreactivity of SARS-CoV-2. Biomedicines 2023; 11:160. [PMID: 36672668 PMCID: PMC9855841 DOI: 10.3390/biomedicines11010160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/29/2022] [Accepted: 12/31/2022] [Indexed: 01/11/2023] Open
Abstract
Since SARS-CoV-2 emerged in December 2019 in Wuhan, the resulting pandemic has paralyzed the economic and cultural life of the world. Variants of concern (VOC) strongly increase pressure on public health systems. Rapid, easy-to-use, and cost-effective assays are essential to manage the pandemic. Here we present a bioinformatical approach for the fast and efficient design of two innovative serological Particle Enhanced Turbidimetric Immunoassays (PETIA) to quantify the SARS-CoV-2 immunoresponse. To confirm bioinformatical assumptions, an S-RBD- and a Nucleocapsid-based PETIA were produced. Sensitivity and specificity were compared for 95 patient samples using a BioMajesty™ fully automated analyzer. The S-RBD-based PETIA showed necessary specificity (98%) over the N protein-based PETIA (21%). Further, the reactivity and cross-reactivity of the RBD-based PETIA towards variant-derived antibodies of SARS-CoV-2 were assessed by a quenching inhibition test. The inhibition kinetics of the S-RBD variants Alpha, Beta, Delta, Gamma, Kappa, and Omicron were evaluated. In summary, we showed that specific and robust PETIA immunoassays can be rapidly designed and developed. The quantification of the SARS-CoV-2-related immunoresponse of variants (Alpha to Kappa) is possible using specific RBD assays. In contrast, Omicron revealed lower cross-reactivity (approx. 50%). To ensure the quantification of the Omicron variant, modified immunoassays appear to be necessary.
Collapse
Affiliation(s)
- Leoni Wey
- DiaSys Diagnostic Systems GmbH, Alte Str. 9, 65558 Holzheim, Germany
- Hochschule Fresenius Gem. Trägergesellschaft mbH, University of Applied Sciences, Limburger Str. 2, 65510 Idstein, Germany
| | - Thomas Masetto
- DiaSys Diagnostic Systems GmbH, Alte Str. 9, 65558 Holzheim, Germany
- Institut für Molekulare Medizin I, Heinrich-Heine-Universität, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Alexander Spaeth
- MVZ Medizinische Labore Dessau Kassel GmbH, Bauhüttenstr. 6, 06847 Dessau-Roßlau, Germany
| | - Jessica Brehm
- MVZ Medizinische Labore Dessau Kassel GmbH, Bauhüttenstr. 6, 06847 Dessau-Roßlau, Germany
| | - Christian Kochem
- DiaSys Diagnostic Systems GmbH, Alte Str. 9, 65558 Holzheim, Germany
| | | | - Holger Müller
- DiaSys Diagnostic Systems GmbH, Alte Str. 9, 65558 Holzheim, Germany
| | - Uwe Kempin
- pes Medizinische Diagnosesysteme GmbH, Hauptstr. 103, 04416 Markkleeberg, Germany
| | - Franziska Lorenz
- MVZ Medizinische Labore Dessau Kassel GmbH, Bauhüttenstr. 6, 06847 Dessau-Roßlau, Germany
| | - Christoph Peter
- Institut für Molekulare Medizin I, Heinrich-Heine-Universität, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Matthias Grimmler
- DiaSys Diagnostic Systems GmbH, Alte Str. 9, 65558 Holzheim, Germany
- Hochschule Fresenius Gem. Trägergesellschaft mbH, University of Applied Sciences, Limburger Str. 2, 65510 Idstein, Germany
| |
Collapse
|
31
|
Cooper A, Sidaway A, Chandrashekar A, Latta E, Chakraborty K, Yu J, McMahan K, Giffin V, Manickam C, Kroll K, Mosher M, Reeves RK, Gam R, Arthofer E, Choudhry M, Henley T, Barouch DH. A genetically engineered, stem-cell-derived cellular vaccine. Cell Rep Med 2022; 3:100843. [PMID: 36480934 PMCID: PMC9727836 DOI: 10.1016/j.xcrm.2022.100843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 10/19/2022] [Accepted: 11/10/2022] [Indexed: 12/12/2022]
Abstract
Despite rapid clinical translation of COVID-19 vaccines in response to the global pandemic, an opportunity remains for vaccine technology innovation to address current limitations and meet challenges of inevitable future pandemics. We describe a universal vaccine cell (UVC) genetically engineered to mimic natural physiological immunity induced upon viral infection of host cells. Cells engineered to express the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike as a representative viral antigen induce robust neutralizing antibodies in immunized non-human primates. Similar titers generated in this established non-human primate (NHP) model have translated into protective human neutralizing antibody levels in SARS-CoV-2-vaccinated individuals. Animals vaccinated with ancestral spike antigens and subsequently challenged with SARS-CoV-2 Delta variant in a heterologous challenge have an approximately 3 log decrease in viral subgenomic RNA in the lungs. This cellular vaccine is designed as a scalable cell line with a modular poly-antigenic payload, allowing for rapid, large-scale clinical manufacturing and use in an evolving viral variant environment.
Collapse
Affiliation(s)
| | | | - Abishek Chandrashekar
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | | | - Jingyou Yu
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Katherine McMahan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Victoria Giffin
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Cordelia Manickam
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kyle Kroll
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Matthew Mosher
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - R Keith Reeves
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Rihab Gam
- Intima Bioscience, Inc., New York, NY, USA
| | | | - Modassir Choudhry
- Praesidium Bioscience, Inc., New York, NY, USA; Intima Bioscience, Inc., New York, NY, USA
| | - Tom Henley
- Praesidium Bioscience, Inc., New York, NY, USA; Intima Bioscience, Inc., New York, NY, USA.
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA, USA.
| |
Collapse
|
32
|
Özcengiz E, Keser D, Özcengiz G, Çelik G, Özkul A, İnçeh FN. Two formulations of coronavirus disease-19 recombinant subunit vaccine candidate made up of S1 fragment protein P1, S2 fragment protein P2, and nucleocapsid protein elicit strong immunogenicity in mice. Immun Inflamm Dis 2022; 10:e748. [PMID: 36444622 PMCID: PMC9695085 DOI: 10.1002/iid3.748] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/09/2022] [Accepted: 10/29/2022] [Indexed: 11/26/2022] Open
Abstract
INTRODUCTION Coronavirus disease (COVID-19) is ongoing as a global epidemic and there is still a need to develop much safer and more effective new vaccines that can also be easily adapted to important variants of the pathogen. In the present study in this direction, we developed a new COVID-19 vaccine, composed of two critical antigenic fragments of the S1 and S2 region of severe acute respiratory syndrome coronavirus 2 as well as the whole nucleocapsid protein (N), which was formulated with either alum or alum plus monophosphoryl lipid A (MPLA) adjuvant combinations. METHODS From within the spike protein S1 region, a fragmented protein P1 (MW:33 kDa) which includes the receptor-binding domain (RBD), another fragment protein P2 (MW:17.6) which contains important antigenic epitopes within the spike protein S2 region, and N protein (MW:46 kDa) were obtained after recombinant expression of the corresponding gene regions in Escherichia coli BL21. For use in immunization studies, three proteins were adsorbed with aluminum hydroxide gel and with the combination of aluminum hydroxide gel plus MPLA. RESULTS Each of the three protein antigens produced strong reactions in enzyme-linked immunosorbent assays and Western blot analysis studies performed with convalescent COVID-19 patient sera. In mice, these combined protein vaccine candidates elicited high titer anti-P1, anti-P2, and anti-N IgG and IgG2a responses. These also induced highly neutralizing antibodies and elicited significant cell-mediated immunity as demonstrated by enhanced antigen-specific levels of interferon-γ (INF-γ) in the splenocytes of immunized mice. CONCLUSION The results of this study showed that formulations of the three proteins with Alum or Alum + MPLA are effective in terms of humoral and cellular responses. However, since the Alum + MPLA formulation appears to be superior in Th1 response, this vaccine candidate may be recommended mainly for the elderly and immunocompromised individuals. We also believe that the alum-only formulation will provide great benefits for adults, young adolescents, and children.
Collapse
Affiliation(s)
| | - Duygu Keser
- Vaccine R&D, Pharmada Pharmaceuticals, Ankara, Turkey
| | - Gülay Özcengiz
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Gözde Çelik
- Vaccine R&D, Pharmada Pharmaceuticals, Ankara, Turkey
| | - Aykut Özkul
- Department of Virology, School of Veterinary Medicine, Ankara University, Ankara, Turkey
| | | |
Collapse
|
33
|
Franke V, Meyer S, Schulze-Tanzil GG, Braun T, Kokozidou M, Fischlein T, Silawal S. Complement Regulation in Immortalized Fibroblast-like Synoviocytes and Primary Human Endothelial Cells in Response to SARS-CoV-2 Nucleocapsid Protein and Pro-Inflammatory Cytokine TNFα. Life (Basel) 2022; 12:1527. [PMID: 36294967 PMCID: PMC9604721 DOI: 10.3390/life12101527] [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: 08/13/2022] [Revised: 09/16/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Case reports are available showing that patients develop symptoms of acute arthritis during or after recovery from SARS-CoV-2 infection. Since the interrelation is still unknown, our aim was to study the impact of the SARS-CoV-2 nucleocapsid protein (NP) on human fibroblast-like synoviocytes and human endothelial cells (hEC) in terms of complement and cytokine regulation. Methods: Non-arthritic (K4IM) synoviocyte, arthritic (HSE) synoviocyte cell lines and primary hEC were stimulated with recombinant NP and/or TNFα. Analyses of cell viability, proliferation, gene and protein expression of cytokines and complement factors were performed. Results: NP suppressed significantly the vitality of hEC and proliferation of HSE. NP alone did not induce any significant changes in the examined gene expressions. However, NP combined with TNFα induced significantly higher TNFα in HSE and K4IM as well as higher IL-6 and CD55 gene expression in HSE and suppressed C3aR1 gene expression in hEC. HSE proliferated twice as fast as K4IM, but showed significantly lesser gene expressions of CD46, CD55, CD59 and TNFα with significantly higher IL-6 gene expression. CD35 gene expression was undetectable in K4IM, HSE and hEC. Conclusions: NP might contribute in combination with other inflammatory factors to complement regulation in arthritis.
Collapse
Affiliation(s)
- Vincent Franke
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany
| | - Sophie Meyer
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany
| | - Gundula Gesine Schulze-Tanzil
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany
| | - Tobias Braun
- Department of Cardiac Surgery, Cardiovascular Center, General Hospital Nuremberg and Paracelsus Medical University, Breslauer Str. 201, 90471 Nuremberg, Germany
| | - Maria Kokozidou
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany
| | - Theodor Fischlein
- Department of Cardiac Surgery, Cardiovascular Center, General Hospital Nuremberg and Paracelsus Medical University, Breslauer Str. 201, 90471 Nuremberg, Germany
| | - Sandeep Silawal
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany
| |
Collapse
|
34
|
Dotiwala F, Upadhyay AK. A comprehensive review of BBV152 vaccine development, effectiveness, safety, challenges, and prospects. Front Immunol 2022; 13:940715. [PMID: 36177016 PMCID: PMC9513542 DOI: 10.3389/fimmu.2022.940715] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
The world has responded to the COVID-19 pandemic with unprecedented speed and vigor in the mass vaccination campaigns, targeted to reduce COVID-19 severity and mortality, reduce the pressure on the healthcare system, re-open society, and reduction in disease mortality and morbidity. Here we review the preclinical and clinical development of BBV152, a whole virus inactivated vaccine and an important tool in the fight to control this pandemic. BBV152, formulated with a TLR7/8 agonist adjuvant generates a Th1-biased immune response that induces high neutralization efficacy against different SARS-CoV-2 variants of concern and robust long-term memory B- and T-cell responses. With seroconversion rates as high as 98.3% in vaccinated individuals, BBV152 shows 77.8% and 93.4% protection from symptomatic COVID-19 disease and severe symptomatic COVID-19 disease respectively. Studies in pediatric populations show superior immunogenicity (geometric mean titer ratio of 1.76 compared to an adult) with a seroconversion rate of >95%. The reactogenicity and safety profiles were comparable across all pediatric age groups between 2-18 yrs. as in adults. Like most approved vaccines, the BBV152 booster given 6 months after full vaccination, reverses a waning immunity, restores the neutralization efficacy, and shows synergy in a heterologous prime-boost study with about 3-fold or 300% increase in neutralization titers against multiple SARS-CoV-2 variants of concern. Based on the interim Phase III data, BBV152 received full authorization for adults and emergency use authorization for children from ages 6 to 18 years in India. It is also licensed for emergency use in 14 countries globally. Over 313 million vaccine doses have already been administered in India alone by April 18th, 2022.
Collapse
|
35
|
Castrejón-Jiménez NS, García-Pérez BE, Reyes-Rodríguez NE, Vega-Sánchez V, Martínez-Juárez VM, Hernández-González JC. Challenges in the Detection of SARS-CoV-2: Evolution of the Lateral Flow Immunoassay as a Valuable Tool for Viral Diagnosis. BIOSENSORS 2022; 12:bios12090728. [PMID: 36140114 PMCID: PMC9496238 DOI: 10.3390/bios12090728] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 12/11/2022]
Abstract
SARS-CoV-2 is an emerging infectious disease of zoonotic origin that caused the coronavirus disease in late 2019 and triggered a pandemic that has severely affected human health and caused millions of deaths. Early and massive diagnosis of SARS-CoV-2 infected patients is the key to preventing the spread of the virus and controlling the outbreak. Lateral flow immunoassays (LFIA) are the simplest biosensors. These devices are clinical diagnostic tools that can detect various analytes, including viruses and antibodies, with high sensitivity and specificity. This review summarizes the advantages, limitations, and evolution of LFIA during the SARS-CoV-2 pandemic and the challenges of improving these diagnostic devices.
Collapse
Affiliation(s)
- Nayeli Shantal Castrejón-Jiménez
- Área Académica de Medicina Veterinaria y Zootecnia, Instituto de Ciencias Agropecuarias, Universidad Autónoma del Estado de Hidalgo, Av. Universidad km 1 Exhacienda de Aquetzalpa A.P. 32, Tulancingo 43600, Mexico
| | - Blanca Estela García-Pérez
- Department of Microbiology, Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas Prolongación de Carpio y Plan de Ayala S/N, Col. Santo Tomás, México City 11340, Mexico
| | - Nydia Edith Reyes-Rodríguez
- Área Académica de Medicina Veterinaria y Zootecnia, Instituto de Ciencias Agropecuarias, Universidad Autónoma del Estado de Hidalgo, Av. Universidad km 1 Exhacienda de Aquetzalpa A.P. 32, Tulancingo 43600, Mexico
| | - Vicente Vega-Sánchez
- Área Académica de Medicina Veterinaria y Zootecnia, Instituto de Ciencias Agropecuarias, Universidad Autónoma del Estado de Hidalgo, Av. Universidad km 1 Exhacienda de Aquetzalpa A.P. 32, Tulancingo 43600, Mexico
| | - Víctor Manuel Martínez-Juárez
- Área Académica de Medicina Veterinaria y Zootecnia, Instituto de Ciencias Agropecuarias, Universidad Autónoma del Estado de Hidalgo, Av. Universidad km 1 Exhacienda de Aquetzalpa A.P. 32, Tulancingo 43600, Mexico
| | - Juan Carlos Hernández-González
- Área Académica de Medicina Veterinaria y Zootecnia, Instituto de Ciencias Agropecuarias, Universidad Autónoma del Estado de Hidalgo, Av. Universidad km 1 Exhacienda de Aquetzalpa A.P. 32, Tulancingo 43600, Mexico
- Correspondence: ; Tel.: +52-775-756-0308
| |
Collapse
|
36
|
Brlić PK, Pavletić M, Lerga M, Krstanović F, Matešić MP, Miklić K, Malić S, Mikša L, Pajcur M, Peruč D, Schubert M, Bertoglio F, Arapović J, Protić A, Šustić A, Milošević M, Šain LČ, Jonjić S, Lisnić VJ, Brizić I. SARS-CoV-2 Spike and Nucleocapsid Antibody Response in Vaccinated Croatian Healthcare Workers and Infected Hospitalized Patients: A Single Center Cohort Study. Viruses 2022; 14:1966. [PMID: 36146773 PMCID: PMC9503044 DOI: 10.3390/v14091966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
Studies assessing the dynamics and duration of antibody responses following SARS-CoV-2 infection or vaccination are an invaluable tool for vaccination schedule planning, assessment of risk groups and management of pandemics. In this study, we developed and employed ELISA assays to analyze the humoral responses to Nucleocapsid and Spike proteins in vaccinated health-care workers (HCW) and critically ill COVID-19 patients. Sera of more than 1000 HCWs and critically ill patients from the Clinical Hospital Center Rijeka were tested across a one-year period, encompassing the spread of major SARS-CoV-2 variants of concern (VOCs). We observed 97% of seroconversion in HCW cohort as well as sustained anti-Spike antibody response in vaccinees for more than 6 months. In contrast, the infection-induced anti-Nucleocapsid response was waning significantly in a six-month period. Furthermore, a substantial decrease in vaccinees' anti-Spike antibodies binding to Spike protein of Omicron VOC was also observed. Critically ill COVID-19 patients had higher levels of anti-Spike and anti-Nucleocapsid antibodies compared to HCWs. No significant differences in anti-Spike and anti-Nucleocapsid antibody levels between the critically ill COVID-19 patients that were on non-invasive oxygen supplementation and those on invasive ventilation support were observed. However, stronger anti-Spike, but not anti-Nucleocapsid, antibody response correlated with a better disease outcome in the cohort of patients on invasive ventilation support. Altogether, our results contribute to the growing pool of data on humoral responses to SARS-CoV-2 infection and vaccination.
Collapse
Affiliation(s)
- Paola Kučan Brlić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Martina Pavletić
- Emergency Department, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia
| | - Mate Lerga
- Emergency Department, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia
| | - Fran Krstanović
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Marina Pribanić Matešić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Karmela Miklić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Suzana Malić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Leonarda Mikša
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Maja Pajcur
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Dolores Peruč
- Department of Microbiology and Parasitology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Maren Schubert
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Spielmannstr. 7, 38106 Braunschweig, Germany
| | - Federico Bertoglio
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Spielmannstr. 7, 38106 Braunschweig, Germany
| | - Jurica Arapović
- Faculty of Medicine, University of Mostar, Bijeli Brijeg b.b., 88000 Mostar, Bosnia and Herzegovina
| | - Alen Protić
- Department of Anesthesiology, Reanimation, Intensive Care and Emergency Medicine, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Alan Šustić
- Department of Anesthesiology, Reanimation, Intensive Care and Emergency Medicine, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
- Department of Clinical Medical Science II, Faculty of Health Studies, University of Rijeka, 51000 Rijeka, Croatia
| | - Marko Milošević
- Department of Anesthesiology, Reanimation, Intensive Care and Emergency Medicine, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Luka Čičin Šain
- Helmholtz Center for Infection Research, Department of Viral Immunology, 38124 Braunschweig, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover/Braunschweig, 38124 Braunschweig, Germany
- Centre for Individualised Infection Medicine (CiiM), Joint Venture of Helmholtz Centre for Infection Research and Hannover Medical School, 30625 Hannover, Germany
| | - Stipan Jonjić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Vanda Juranić Lisnić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Ilija Brizić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| |
Collapse
|
37
|
Farshidi H, Farshidi N, Ghaedi T, Hassaniazad M, Eftekhar E, Gouklani H, Asadi Karam MR, Shahbazi B, Kalani M, Ahmadi K. Preparation and pre-clinical evaluation of flagellin-adjuvanted NOM vaccine candidate formulated with Spike protein against SARS-CoV-2 in mouse model. Microb Pathog 2022; 171:105736. [PMID: 36030048 PMCID: PMC9400380 DOI: 10.1016/j.micpath.2022.105736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/19/2022] [Accepted: 08/19/2022] [Indexed: 10/25/2022]
Abstract
From December 2019, the outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was started as a cluster of pneumonia cases in Wuhan, Hubei Province, China. The disturbing statistics of SARS-CoV-2 promoted scientists to develop an effective vaccine against this infection. NOM protein is a multi-epitope protein that designed based on Nucleocapsid, ORF3a, and Membrane proteins of SARS-CoV-2. Flagellin is a structural protein that binds to the Toll-like receptor 5 and can enhance the immune response to a particular antigen. In this study, NOM protein as vaccine candidate was linked to the carboxyl and amino terminals of flagellin adjuvant derived from Salmonella enterica subsp. enterica serovar Dublin. Then, informatics evaluations were performed for both NOM protein and NOM protein linked to flagellin (FNOM). The interaction between the NOM and FNOM proteins with the TLR5 were assessed using docking analysis. The FNOM protein, which compared to the NOM protein, had a more suitable 3D structure and a stronger interaction with TLR5, was selected for experimental study. The FNOM and Spike (S) proteins expressed and then purified by Ni-NTA column as vaccine candidates. For analysis of immune response, anti-FNOM and anti-S proteins total IgG and IFN-γ, TNF-α, IL-6, IL-10, IL-22 and IL-17 cytokines were evaluated after vaccination of mice with vaccine candidates. The results indicated that the specific antisera (Total IgG) raised in mice that received FNOM protein formulated with S protein were higher than mice that received FNOM and S proteins alone. Also, IFN-γ and TNF-α levels after the spleen cells stimulation were significantly increased in mice that received the FNOM protein formulated with S protein compared to other groups. Immunogenic evaluations showed that, the FNOM chimeric protein could simultaneously elicit humoral and cell-mediated immune responses. Finally, it could be concluded that the FNOM protein formulated with S protein could be considered as potential vaccine candidate for protection against SARS-CoV-2 in the near future.
Collapse
Affiliation(s)
- Hossein Farshidi
- Cardiovascular Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Narges Farshidi
- Department of Immunology, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Tayebeh Ghaedi
- Infectious and Tropical Diseases Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mehdi Hassaniazad
- Infectious and Tropical Diseases Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Ebrahim Eftekhar
- Molecular Medicine Research Center، Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Hamed Gouklani
- Infectious and Tropical Diseases Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | | | - Behzad Shahbazi
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mehdi Kalani
- Professor Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Khadijeh Ahmadi
- Infectious and Tropical Diseases Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| |
Collapse
|
38
|
Zhang B, Xie Y, Lan Z, Li D, Tian J, Zhang Q, Tian H, Yang J, Zhou X, Qiu S, Lu K, Liu Y. SARS-CoV-2 Nucleocapsid Protein Has DNA-Melting and Strand-Annealing Activities With Different Properties From SARS-CoV-2 Nsp13. Front Microbiol 2022; 13:851202. [PMID: 35935242 PMCID: PMC9354549 DOI: 10.3389/fmicb.2022.851202] [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: 01/09/2022] [Accepted: 06/13/2022] [Indexed: 11/25/2022] Open
Abstract
Since December 2019, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spread throughout the world and has had a devastating impact on health and economy. The biochemical characterization of SARS-CoV-2 proteins is important for drug design and development. In this study, we discovered that the SARS-CoV-2 nucleocapsid protein can melt double-stranded DNA (dsDNA) in the 5′-3′ direction, similar to SARS-CoV-2 nonstructural protein 13. However, the unwinding activity of SARS-CoV-2 nucleocapsid protein was found to be more than 22 times weaker than that of SARS-CoV-2 nonstructural protein 13, and the melting process was independent of nucleoside triphosphates and Mg2+. Interestingly, at low concentrations, the SARS-CoV-2 nucleocapsid protein exhibited a stronger annealing activity than SARS-CoV-2 nonstructural protein 13; however, at high concentrations, it promoted the melting of dsDNA. These findings have deepened our understanding of the SARS-CoV-2 nucleocapsid protein and will help provide novel insights into antiviral drug development.
Collapse
Affiliation(s)
- Bo Zhang
- College of Basic Medicine, Zunyi Medical University, Zunyi, China
- Bo Zhang,
| | - Yan Xie
- School of Public Health, Zunyi Medical University, Zunyi, China
| | - Zhaoling Lan
- College of Basic Medicine, Zunyi Medical University, Zunyi, China
| | - Dayu Li
- College of Basic Medicine, Zunyi Medical University, Zunyi, China
| | - Junjie Tian
- College of Basic Medicine, Zunyi Medical University, Zunyi, China
| | - Qintao Zhang
- College of Basic Medicine, Zunyi Medical University, Zunyi, China
| | - Hongji Tian
- College of Basic Medicine, Zunyi Medical University, Zunyi, China
| | - Jiali Yang
- College of Basic Medicine, Zunyi Medical University, Zunyi, China
| | - Xinnan Zhou
- College of Basic Medicine, Zunyi Medical University, Zunyi, China
| | - Shuyi Qiu
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-bioengineering, Guizhou University, Guiyang, China
| | - Keyu Lu
- College of Basic Medicine, Zunyi Medical University, Zunyi, China
- Keyu Lu,
| | - Yang Liu
- School of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-bioengineering, Guizhou University, Guiyang, China
- *Correspondence: Yang Liu,
| |
Collapse
|
39
|
Gerges D, Kapps S, Hernández-Carralero E, Freire R, Aiad M, Schmidt S, Winnicki W, Reiter T, Pajenda S, Schmidt A, Sunder-Plassmann G, Wagner L. Vaccination with BNT162b2 and ChAdOx1 nCoV-19 Induces Cross-Reactive Anti-RBD IgG against SARS-CoV-2 Variants including Omicron. Viruses 2022; 14:1181. [PMID: 35746653 PMCID: PMC9231407 DOI: 10.3390/v14061181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 02/04/2023] Open
Abstract
SARS-CoV-2 variants of concern (VOCs) have caused a significant increase in infections worldwide. Despite high vaccination rates in industrialized countries, the fourth VOC, Omicron, has outpaced the Delta variant and is causing breakthrough infections in individuals with two booster vaccinations. While the magnitude of morbidity and lethality is lower in Omicron, the infection rate and global spread are rapid. Using a specific IgG multipanel-ELISA with the spike protein’s receptor-binding domain (RBD) from recombinant Alpha, Gamma, Delta, and Omicron variants, sera from health-care workers from the Medical University of Vienna were tested pre-pandemic and post-vaccination (BNT162b2; ChAdOx1 nCoV-19). The cohort was continuously monitored by SARS-CoV-2 testing and commercial nucleocapsid IgG ELISA. RBD IgG ELISA showed significantly lower reactivity against the Omicron-RBD compared to the Alpha variant in all individuals (p < 0.001). IgG levels were independent of sex, but were significantly higher in BNT162b2 recipients <45 years of age for Alpha, Gamma, and Delta (p < 0.001; p = 0.040; p = 0.004, respectively). Pre-pandemic cross-reactive anti-Omicron IgG was detected in 31 individuals and was increased 8.78-fold after vaccination, regardless of vaccine type. The low anti-RBD Omicron IgG level could explain the breakthrough infections and their presence could also contribute to a milder COVID-19 course by cross-reactivity and broadening the adaptive immunity.
Collapse
Affiliation(s)
- Daniela Gerges
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, 1090 Vienna, Austria; (D.G.); (S.K.); (M.A.); (S.S.); (W.W.); (T.R.); (S.P.); (A.S.); (G.S.-P.)
| | - Sebastian Kapps
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, 1090 Vienna, Austria; (D.G.); (S.K.); (M.A.); (S.S.); (W.W.); (T.R.); (S.P.); (A.S.); (G.S.-P.)
| | - Esperanza Hernández-Carralero
- Unidad de Investigacion, Hospital Universitario de Canarias-FIISC, 38320 La Laguna, Spain; (E.H.-C.); (R.F.)
- Instituto de Tecnologías Biomedicas, Universidad de La Laguna, 38200 La Laguna, Spain
| | - Raimundo Freire
- Unidad de Investigacion, Hospital Universitario de Canarias-FIISC, 38320 La Laguna, Spain; (E.H.-C.); (R.F.)
- Instituto de Tecnologías Biomedicas, Universidad de La Laguna, 38200 La Laguna, Spain
- Universidad Fernando Pessoa Canarias, 35450 Las Palmas de Gran Canaria, Spain
| | - Monika Aiad
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, 1090 Vienna, Austria; (D.G.); (S.K.); (M.A.); (S.S.); (W.W.); (T.R.); (S.P.); (A.S.); (G.S.-P.)
| | - Sophie Schmidt
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, 1090 Vienna, Austria; (D.G.); (S.K.); (M.A.); (S.S.); (W.W.); (T.R.); (S.P.); (A.S.); (G.S.-P.)
| | - Wolfgang Winnicki
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, 1090 Vienna, Austria; (D.G.); (S.K.); (M.A.); (S.S.); (W.W.); (T.R.); (S.P.); (A.S.); (G.S.-P.)
| | - Thomas Reiter
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, 1090 Vienna, Austria; (D.G.); (S.K.); (M.A.); (S.S.); (W.W.); (T.R.); (S.P.); (A.S.); (G.S.-P.)
| | - Sahra Pajenda
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, 1090 Vienna, Austria; (D.G.); (S.K.); (M.A.); (S.S.); (W.W.); (T.R.); (S.P.); (A.S.); (G.S.-P.)
| | - Alice Schmidt
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, 1090 Vienna, Austria; (D.G.); (S.K.); (M.A.); (S.S.); (W.W.); (T.R.); (S.P.); (A.S.); (G.S.-P.)
| | - Gere Sunder-Plassmann
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, 1090 Vienna, Austria; (D.G.); (S.K.); (M.A.); (S.S.); (W.W.); (T.R.); (S.P.); (A.S.); (G.S.-P.)
| | - Ludwig Wagner
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, 1090 Vienna, Austria; (D.G.); (S.K.); (M.A.); (S.S.); (W.W.); (T.R.); (S.P.); (A.S.); (G.S.-P.)
| |
Collapse
|
40
|
Silva EKVB, Bomfim CG, Barbosa AP, Noda P, Noronha IL, Fernandes BHV, Machado RRG, Durigon EL, Catanozi S, Rodrigues LG, Pieroni F, Lima SG, Teodoro WR, Queiroz ZAJ, Silveira LKR, Charlie-Silva I, Capelozzi VL, Guzzo CR, Fanelli C. Immunization with SARS-CoV-2 Nucleocapsid protein triggers a pulmonary immune response in rats. PLoS One 2022; 17:e0268434. [PMID: 35609032 PMCID: PMC9129034 DOI: 10.1371/journal.pone.0268434] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 04/29/2022] [Indexed: 12/23/2022] Open
Abstract
The SARS-CoV-2 pandemic have been affecting millions of people worldwide, since the beginning of 2020. COVID-19 can cause a wide range of clinical symptoms, which varies from asymptomatic presentation to severe respiratory insufficiency, exacerbation of immune response, disseminated microthrombosis and multiple organ failure, which may lead to dead. Due to the rapid spread of SARS-CoV-2, the development of vaccines to minimize COVID-19 severity in the world population is imperious. One of the employed techniques to produce vaccines against emerging viruses is the synthesis of recombinant proteins, which can be used as immunizing agents. Based on the exposed, the aim of the present study was to verify the systemic and immunological effects of IM administration of recombinant Nucleocapsid protein (NP), derived from SARS-CoV-2 and produced by this research group, in 2 different strains of rats (Rattus norvegicus); Wistar and Lewis. For this purpose, experimental animals received 4 injections of NP, once a week, and were submitted to biochemical and histological analysis. Our results showed that NP inoculations were safe for the animals, which presented no clinical symptoms of worrying side effects, nor laboratorial alterations in the main biochemical and histological parameters, suggesting the absence of toxicity induced by NP. Moreover, NP injections successfully triggered the production of specific anti-SARS-CoV-2 IgG antibodies by both Wistar and Lewis rats, showing the sensitization to have been well sufficient for the immunization of these strains of rats. Additionally, we observed the local lung activation of the Bronchus-Associated Lymphoid Tissue (BALT) of rats in the NP groups, suggesting that NP elicits specific lung immune response. Although pre-clinical and clinical studies are still required, our data support the recombinant NP produced by this research group as a potential immunizing agent for massive vaccination, and may represent advantages upon other recombinant proteins, since it seems to induce specific pulmonary protection.
Collapse
Affiliation(s)
- Everidiene K. V. B. Silva
- Renal Division, Department of Clinical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Camila G. Bomfim
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ana P. Barbosa
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Paloma Noda
- Renal Division, Department of Clinical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Irene L. Noronha
- Renal Division, Department of Clinical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Bianca H. V. Fernandes
- Laboratório de Controle Genético e Sanitário, Diretoria Técnica de Apoio ao Ensino e Pesquisa, Faculdade de Medicina da Universidade de São Paulo, Sao Paulo, Brazil
| | - Rafael R. G. Machado
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Edison L. Durigon
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Sergio Catanozi
- Laboratorio de Lipides (LIM-10), Hospital das Clinicas (HCFMUSP) da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Letícia G. Rodrigues
- Laboratorio de Lipides (LIM-10), Hospital das Clinicas (HCFMUSP) da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| | | | - Sérgio G. Lima
- Labinbraz Comercial Ltda. - Wiener lab, Sao Paulo, Brazil
| | - Walcy R. Teodoro
- Rheumatology Division of the Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo - SP, Sao Paulo, Brazil
| | - Zelita A. J. Queiroz
- Rheumatology Division of the Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo - SP, Sao Paulo, Brazil
| | - Lizandre K. R. Silveira
- Rheumatology Division of the Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo - SP, Sao Paulo, Brazil
| | - Ives Charlie-Silva
- Department of Pharmacology, Institute of Biomedical Sciences, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Vera L. Capelozzi
- Rheumatology Division of the Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo - SP, Sao Paulo, Brazil
| | - Cristiane R. Guzzo
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Camilla Fanelli
- Renal Division, Department of Clinical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
41
|
Podrazil M, Taborska P, Stakheev D, Rataj M, Lastovicka J, Vlachova A, Pohunek P, Bartunkova J, Smrz D. Effectiveness and Durability of mRNA Vaccine-Induced SARS-CoV-2-Specific Humoral and Cellular Immunity in Severe Asthma Patients on Biological Therapy. Front Immunol 2022; 13:892277. [PMID: 35669765 PMCID: PMC9163958 DOI: 10.3389/fimmu.2022.892277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/22/2022] [Indexed: 12/20/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) vaccines effectively elicit humoral and cellular immunity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in healthy populations. This immunity decreases several months after vaccination. However, the efficacy of vaccine-induced immunity and its durability in patients with severe asthma on biological therapy are unknown. In this study, we evaluated the effectiveness and durability of mRNA vaccine-induced SARS-CoV-2-specific humoral and cellular immunity in severe asthma patients on biological therapy. The study included 34 patients with severe asthma treated with anti-IgE (omalizumab, n=17), anti-IL5 (mepolizumab, n=13; reslizumab, n=3), or anti-IL5R (benralizumab, n=1) biological therapy. All patients were vaccinated with two doses of the BNT162b2 mRNA vaccine with a 6-week interval between the doses. We found that this COVID-19 vaccination regimen elicited SARS-CoV-2-specific humoral and cellular immunity, which had significantly declined 6 months after receipt of the second dose of the vaccine. The type of biological treatment did not affect vaccine-elicited immunity. However, patient age negatively impacted the vaccine-induced humoral response. On the other hand, no such age-related impact on vaccine-elicited cellular immunity was observed. Our findings show that treatment of patients with severe asthma with biological therapy does not compromise the effectiveness or durability of COVID-19 vaccine-induced immunity.
Collapse
Affiliation(s)
- Michal Podrazil
- Department of Immunology, Second Faculty of Medicine, Charles University, and Motol University Hospital, Prague, Czechia
| | - Pavla Taborska
- Department of Immunology, Second Faculty of Medicine, Charles University, and Motol University Hospital, Prague, Czechia
| | - Dmitry Stakheev
- Department of Immunology, Second Faculty of Medicine, Charles University, and Motol University Hospital, Prague, Czechia
| | - Michal Rataj
- Department of Immunology, Second Faculty of Medicine, Charles University, and Motol University Hospital, Prague, Czechia
| | - Jan Lastovicka
- Department of Immunology, Second Faculty of Medicine, Charles University, and Motol University Hospital, Prague, Czechia
| | - Alena Vlachova
- Department of Pneumology, Second Faculty of Medicine, Charles University, and Motol University Hospital, Prague, Czechia
| | - Petr Pohunek
- Department of Pediatrics, Second Faculty of Medicine, Charles University, and Motol University Hospital, Prague, Czechia
| | - Jirina Bartunkova
- Department of Immunology, Second Faculty of Medicine, Charles University, and Motol University Hospital, Prague, Czechia
| | - Daniel Smrz
- Department of Immunology, Second Faculty of Medicine, Charles University, and Motol University Hospital, Prague, Czechia
| |
Collapse
|
42
|
Bwire G, Ario AR, Eyu P, Ocom F, Wamala JF, Kusi KA, Ndeketa L, Jambo KC, Wanyenze RK, Talisuna AO. The COVID-19 pandemic in the African continent. BMC Med 2022; 20:167. [PMID: 35501853 PMCID: PMC9059455 DOI: 10.1186/s12916-022-02367-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 04/05/2022] [Indexed: 01/13/2023] Open
Abstract
In December 2019, a new coronavirus, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and associated disease, coronavirus disease 2019 (COVID-19), was identified in China. This virus spread quickly and in March, 2020, it was declared a pandemic. Scientists predicted the worst scenario to occur in Africa since it was the least developed of the continents in terms of human development index, lagged behind others in achievement of the United Nations sustainable development goals (SDGs), has inadequate resources for provision of social services, and has many fragile states. In addition, there were relatively few research reporting findings on COVID-19 in Africa. On the contrary, the more developed countries reported higher disease incidences and mortality rates. However, for Africa, the earlier predictions and modelling into COVID-19 incidence and mortality did not fit into the reality. Therefore, the main objective of this forum is to bring together infectious diseases and public health experts to give an overview of COVID-19 in Africa and share their thoughts and opinions on why Africa behaved the way it did. Furthermore, the experts highlight what needs to be done to support Africa to consolidate the status quo and overcome the negative effects of COVID-19 so as to accelerate attainment of the SDGs.
Collapse
Affiliation(s)
- Godfrey Bwire
- Department of Integrated Epidemiology Surveillance and Public Health Emergencies, Ministry of Health, P.O Box 7272, Kampala, Uganda
- School of Public Health, Makerere University, P.O. Box 7072, Kampala, Uganda
| | | | - Patricia Eyu
- Uganda National Institute of Public Health, Kampala, Uganda
| | - Felix Ocom
- Uganda National Institute of Public Health, Kampala, Uganda
| | | | - Kwadwo A. Kusi
- Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Latif Ndeketa
- Malawi-Liverpool-Wellcome Programme (MLW), Blantyre, Malawi
| | - Kondwani C. Jambo
- Malawi-Liverpool-Wellcome Programme (MLW), Blantyre, Malawi
- Liverpool School of Tropical Medicine, Liverpool, UK
| | - Rhoda K. Wanyenze
- School of Public Health, Makerere University, P.O. Box 7072, Kampala, Uganda
| | - Ambrose O. Talisuna
- Epidemic Preparedness and Response Cluster, World Health Organization, Regional Office for Africa, Brazzaville, Congo
| |
Collapse
|
43
|
Dumache R, Enache A, Macasoi I, Dehelean CA, Dumitrascu V, Mihailescu A, Popescu R, Vlad D, Vlad CS, Muresan C. SARS-CoV-2: An Overview of the Genetic Profile and Vaccine Effectiveness of the Five Variants of Concern. Pathogens 2022; 11:pathogens11050516. [PMID: 35631037 PMCID: PMC9144800 DOI: 10.3390/pathogens11050516] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 12/13/2022] Open
Abstract
With the onset of the COVID-19 pandemic, enormous efforts have been made to understand the genus SARS-CoV-2. Due to the high rate of global transmission, mutations in the viral genome were inevitable. A full understanding of the viral genome and its possible changes represents one of the crucial aspects of pandemic management. Structural protein S plays an important role in the pathogenicity of SARS-CoV-2, mutations occurring at this level leading to viral forms with increased affinity for ACE2 receptors, higher transmissibility and infectivity, resistance to neutralizing antibodies and immune escape, increasing the risk of infection and disease severity. Thus, five variants of concern are currently being discussed, Alpha, Beta, Gamma, Delta and Omicron. In the present review, a comprehensive summary of the following critical aspects regarding SARS-CoV-2 has been made: (i) the genomic characteristics of SARS-CoV-2; (ii) the pathological mechanism of transmission, penetration into the cell and action on specific receptors; (iii) mutations in the SARS-CoV-2 genome; and (iv) possible implications of mutations in diagnosis, treatment, and vaccination.
Collapse
Affiliation(s)
- Raluca Dumache
- Ethics and Human Identification Research Center, Department of Neurosciences, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (R.D.); (A.E.); (A.M.); (C.M.)
| | - Alexandra Enache
- Ethics and Human Identification Research Center, Department of Neurosciences, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (R.D.); (A.E.); (A.M.); (C.M.)
| | - Ioana Macasoi
- Departament of Toxicology and Drug Industry, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timişoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
- Correspondence: (I.M.); (C.A.D.)
| | - Cristina Adriana Dehelean
- Departament of Toxicology and Drug Industry, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timişoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
- Correspondence: (I.M.); (C.A.D.)
| | - Victor Dumitrascu
- Department of Pharmacology and Biochemistry, Discipline of Pharmacology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (V.D.); (D.V.); (C.S.V.)
| | - Alexandra Mihailescu
- Ethics and Human Identification Research Center, Department of Neurosciences, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (R.D.); (A.E.); (A.M.); (C.M.)
- Genetics, Genomic Medicine Research Center, Department of Microscopic Morphology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Roxana Popescu
- Department of Microscopic Morphology, Discipline of Molecular and Cell Biology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Daliborca Vlad
- Department of Pharmacology and Biochemistry, Discipline of Pharmacology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (V.D.); (D.V.); (C.S.V.)
| | - Cristian Sebastian Vlad
- Department of Pharmacology and Biochemistry, Discipline of Pharmacology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (V.D.); (D.V.); (C.S.V.)
| | - Camelia Muresan
- Ethics and Human Identification Research Center, Department of Neurosciences, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (R.D.); (A.E.); (A.M.); (C.M.)
| |
Collapse
|
44
|
Ramm F, Dondapati SK, Trinh HA, Wenzel D, Walter RM, Zemella A, Kubick S. The Potential of Eukaryotic Cell-Free Systems as a Rapid Response to Novel Zoonotic Pathogens: Analysis of SARS-CoV-2 Viral Proteins. Front Bioeng Biotechnol 2022; 10:896751. [PMID: 35519622 PMCID: PMC9061942 DOI: 10.3389/fbioe.2022.896751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/01/2022] [Indexed: 11/13/2022] Open
Abstract
The ongoing pandemic caused by the novel coronavirus (SARS-CoV-2) has led to more than 445 million infections and the underlying disease, COVID-19, resulted in more than 6 million deaths worldwide. The scientific world is already predicting future zoonotic diseases. Hence, rapid response systems are needed to tackle future epidemics and pandemics. Here, we present the use of eukaryotic cell-free systems for the rapid response to novel zoonotic diseases represented by SARS-CoV-2. Non-structural, structural and accessory proteins encoded by SARS-CoV-2 were synthesized by cell-free protein synthesis in a fast and efficient manner. The inhibitory effect of the non-structural protein 1 on protein synthesis could be shown in vitro. Structural proteins were quantitatively detected by commercial antibodies, therefore facilitating cell-free systems for the validation of available antibodies. The cytotoxic envelope protein was characterized in electrophysiological planar lipid bilayer measurements. Hence, our study demonstrates the potential of eukaryotic cell-free systems as a rapid response mechanism for the synthesis, functional characterization and antibody validation against a viral pathogen.
Collapse
Affiliation(s)
- Franziska Ramm
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Branch Bioanalytics and Bioprocesses (IZI-BB), Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Srujan K. Dondapati
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Branch Bioanalytics and Bioprocesses (IZI-BB), Potsdam, Germany
| | - Hoai Anh Trinh
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Branch Bioanalytics and Bioprocesses (IZI-BB), Potsdam, Germany
- Department of Applied Biochemistry, Institute of Biotechnology, Technical University Berlin, Berlin, Germany
| | - Dana Wenzel
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Branch Bioanalytics and Bioprocesses (IZI-BB), Potsdam, Germany
| | - Ruben M. Walter
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Branch Bioanalytics and Bioprocesses (IZI-BB), Potsdam, Germany
- Department of Applied Biochemistry, Institute of Biotechnology, Technical University Berlin, Berlin, Germany
| | - Anne Zemella
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Branch Bioanalytics and Bioprocesses (IZI-BB), Potsdam, Germany
| | - Stefan Kubick
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Branch Bioanalytics and Bioprocesses (IZI-BB), Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus–Senftenberg, The Brandenburg Medical School Theodor Fontane, The University of Potsdam, Potsdam, Germany
- *Correspondence: Stefan Kubick,
| |
Collapse
|
45
|
Hevesi Z, Gerges DA, Kapps S, Freire R, Schmidt S, Pollak DD, Schmetterer K, Frey T, Lang R, Winnicki W, Schmidt A, Harkany T, Wagner L. Preclinical Establishment of a Divalent Vaccine against SARS-CoV-2. Vaccines (Basel) 2022; 10:516. [PMID: 35455265 PMCID: PMC9028954 DOI: 10.3390/vaccines10040516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 12/04/2022] Open
Abstract
First-generation vaccines against SARS-CoV-2 do not provide adequate immune protection. Therefore, we engineered a divalent gene construct combining the receptor-binding domain (RBD) of the spike protein and the immunodominant region of the viral nucleocapsid. This fusion protein was produced in either E. coli or a recombinant baculovirus system. Subsequently, the fusion protein was mixed with adjuvant and administered to mice in a prime-booster mode. Mice (72%) produced an IgG response against both proteins (titer: 10-4-10-5) 14 days after the first booster injection, which was increased to 100% by a second booster. Comparable IgG responses were detected against the delta, gamma and omicron variants of the RBD region. Durability testing revealed IgGs beyond 90 days. In addition, cytolytic effector cell molecules were increased in lymphocytes isolated from peripheral blood. Ex vivo stimulation of T cells by nucleocapsid and RBD peptides showed antigen-specific upregulation of CD44 among the CD4+ and CD8+ T cells of vaccinated mice. No side effect was documented in the central nervous system. Cumulatively, these data represent a proof-of-principle approach alternative to existing mRNA vaccination strategies.
Collapse
Affiliation(s)
- Zsofia Hevesi
- Center for Brain Research, Department of Molecular Neurosciences, Medical University of Vienna, 1090 Vienna, Austria; (Z.H.); (T.H.)
| | - Daniela Anna Gerges
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, 1090 Vienna, Austria; (D.A.G.); (S.K.); (S.S.); (W.W.); (A.S.)
| | - Sebastian Kapps
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, 1090 Vienna, Austria; (D.A.G.); (S.K.); (S.S.); (W.W.); (A.S.)
| | - Raimundo Freire
- Unidad de Investigacion, Hospital Universitario de Canarias-FIISC, 38320 La Laguna, Spain;
- Instituto de Tecnologías Biomedicas, Universidad de La Laguna, 38320 La Laguna, Spain
- Universidad Fernando Pessoa Canarias, 35450 Las Palmas de Gran Canaria, Spain
| | - Sophie Schmidt
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, 1090 Vienna, Austria; (D.A.G.); (S.K.); (S.S.); (W.W.); (A.S.)
| | - Daniela D. Pollak
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Klaus Schmetterer
- Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria; (K.S.); (T.F.)
| | - Tobias Frey
- Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria; (K.S.); (T.F.)
| | - Rita Lang
- Division of Endocrinology, Department of Medicine III, Medical University of Vienna, 1090 Vienna, Austria;
| | - Wolfgang Winnicki
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, 1090 Vienna, Austria; (D.A.G.); (S.K.); (S.S.); (W.W.); (A.S.)
| | - Alice Schmidt
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, 1090 Vienna, Austria; (D.A.G.); (S.K.); (S.S.); (W.W.); (A.S.)
| | - Tibor Harkany
- Center for Brain Research, Department of Molecular Neurosciences, Medical University of Vienna, 1090 Vienna, Austria; (Z.H.); (T.H.)
- Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, 17177 Solna, Sweden
| | - Ludwig Wagner
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, 1090 Vienna, Austria; (D.A.G.); (S.K.); (S.S.); (W.W.); (A.S.)
| |
Collapse
|
46
|
Intranasal Coronavirus SARS-CoV-2 Immunization with Lipid Adjuvants Provides Systemic and Mucosal Immune Response against SARS-CoV-2 S1 Spike and Nucleocapsid Protein. Vaccines (Basel) 2022; 10:vaccines10040504. [PMID: 35455253 PMCID: PMC9029453 DOI: 10.3390/vaccines10040504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 03/16/2022] [Indexed: 01/14/2023] Open
Abstract
In this preclinical two-dose mucosal immunization study, using a combination of S1 spike and nucleocapsid proteins with cationic (N3)/or anionic (L3) lipids were investigated using an intranasal delivery route. The study showed that nasal administration of low amounts of antigens/adjuvants induced a primary and secondary immune response in systemic IgG, mIL-5, and IFN-gamma secreting T lymphocytes, as well as humoral IgA in nasal and intestinal mucosal compartments. It is believed that recipients will benefit from receiving a combination of viral antigens in promoting a border immune response against present and evolving contagious viruses. Lipid adjuvants demonstrated an enhanced response in the vaccine effect. This was seen in the significant immunogenicity effect when using the cationic lipid N3. Unlike L3, which showed a recognizable effect when administrated at a slightly higher concentration. Moreover, the findings of the study proved the efficiency of an intranasally mucosal immunization strategy, which can be less painful and more effective in enhancing the respiratory tract immunity against respiratory infectious diseases.
Collapse
|
47
|
Kim PS, Dimcheff DE, Siler A, Schildhouse RJ, Chensue SW. Effect of monoclonal antibody therapy on the endogenous SARS-CoV-2 antibody response. Clin Immunol 2022; 236:108959. [PMID: 35218964 PMCID: PMC8866167 DOI: 10.1016/j.clim.2022.108959] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/12/2022] [Accepted: 02/19/2022] [Indexed: 12/28/2022]
Abstract
Monoclonal antibody treatment of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection has been widely implemented. Effects of treatment on the endogenous primary humoral response to the virus are unknown. A retrospective cohort study performed at a Veterans Health Administration medical center compared serologic responses of treated and untreated COVID-19 patients at high risk for severe outcomes. Three anti-viral spike protein IgG monoclonal treatments were used during the study period, 1) bamlanivimab, 2) casirivimab with imdevimab, and 3) bamlanivimab with etesevimab. Data were analyzed at acute (0-9 days), seroconversion (10-19 days), and maximum antibody (20-39 days) stages. SARS-Cov-2 infection induced a dynamic primary humoral response with anti-spike IgM and anti-nucleocapsid IgG seroconversion occurring after 9 days with maximum serologic indices achieved by 20-39 days. All monoclonal antibody treatments suppressed the endogenous anti-spike IgM response by 85-90% with minor effect on the anti-nucleocapsid response. Thus, passive immunization therapy may cause immunologic interference.
Collapse
Affiliation(s)
- Paul S Kim
- Medicine Service, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA; Division of Hospital Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Derek E Dimcheff
- Medicine Service, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA; Division of Hospital Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Andrew Siler
- Pharmacy Service, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
| | - Richard J Schildhouse
- Medicine Service, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA; Division of Hospital Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Stephen W Chensue
- Pathology and Laboratory Medicine Service, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA; Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
48
|
Thakur S, Sasi S, Pillai SG, Nag A, Shukla D, Singhal R, Phalke S, Velu GSK. SARS-CoV-2 Mutations and Their Impact on Diagnostics, Therapeutics and Vaccines. Front Med (Lausanne) 2022; 9:815389. [PMID: 35273977 PMCID: PMC8902153 DOI: 10.3389/fmed.2022.815389] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/04/2022] [Indexed: 12/11/2022] Open
Abstract
With the high rate of COVID-19 infections worldwide, the emergence of SARS-CoV-2 variants was inevitable. Several mutations have been identified in the SARS-CoV-2 genome, with the spike protein as one of the mutational hot spots. Specific amino acid substitutions such as D614G and N501Y were found to alter the transmissibility and virulence of the virus. The WHO has classified the variants identified with fitness-enhancing mutations as variants of concern (VOC), variants of interest (VOI) or variants under monitoring (VUM). The VOCs pose an imminent threat as they exhibit higher transmissibility, disease severity and ability to evade vaccine-induced and natural immunity. Here we review the mutational landscape on the SARS-CoV-2 structural and non-structural proteins and their impact on diagnostics, therapeutics and vaccines. We also look at the effectiveness of approved vaccines, antibody therapy and convalescent plasma on the currently prevalent VOCs, which are B.1.17, B.1.351, P.1, B.1.617.2 and B.1.1.529. We further discuss the possible factors influencing mutation rates and future directions.
Collapse
Affiliation(s)
- Suresh Thakur
- Trivitron Healthcare Pvt., Ltd., Visakhapatnam, India
| | - Shalitha Sasi
- Blue Horizon International Therapeutic Sciences, Hackensack, NJ, United States
| | | | | | - Dhananjay Shukla
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, India
| | - Ritu Singhal
- Department of Microbiology, National Institute of Tuberculosis and Respiratory Disease, New Delhi, India
| | - Sameer Phalke
- Trivitron Healthcare Pvt., Ltd., Visakhapatnam, India
| | - G. S. K. Velu
- Trivitron Healthcare Pvt., Ltd., Visakhapatnam, India
| |
Collapse
|
49
|
Kebria MM, Milan PB, Peyravian N, Kiani J, Khatibi S, Mozafari M. Stem cell therapy for COVID-19 pneumonia. MOLECULAR BIOMEDICINE 2022; 3:6. [PMID: 35174448 PMCID: PMC8850486 DOI: 10.1186/s43556-021-00067-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 12/22/2021] [Indexed: 12/11/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) virus is a highly contagious microorganism, and despite substantial investigation, no progress has been achieved in treating post-COVID complications. However, the virus has made various mutations and has spread around the world. Researchers have tried different treatments to reduce the side effects of the COVID-19 symptoms. One of the most common and effective treatments now used is steroid therapy to reduce the complications of this disease. Long-term steroid therapy for chronic inflammation following COVID-19 is harmful and increases the risk of secondary infection, and effective treatment remains challenging owing to fibrosis and severe inflammation and infection. Sometimes our immune system can severely damage ourselves in disease. In the past, many researchers have conducted various studies on the immunomodulatory properties of stem cells. This property of stem cells led them to modulate the immune system of autoimmune diseases like diabetes, multiple sclerosis, and Parkinson's. Because of their immunomodulatory properties, stem cell-based therapy employing mesenchymal or hematopoietic stem cells may be a viable alternative treatment option in some patients. By priming the immune system and providing cytokines, chemokines, and growth factors, stem cells can be employed to build a long-term regenerative and protective response. This review addresses the latest trends and rapid progress in stem cell treatment for Acute Respiratory Distress Syndrome (ARDS) following COVID-19.
Collapse
Affiliation(s)
- Maziar Malekzadeh Kebria
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Peiman Brouki Milan
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Noshad Peyravian
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Jafar Kiani
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Present Address: Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Soheil Khatibi
- Babol University of Medical Sciences, Infection Diseases Centre, Mazandaran, Iran
| | - Masoud Mozafari
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
50
|
Bagrov DV, Glukhov GS, Moiseenko AV, Karlova MG, Litvinov DS, Zaitsev PА, Kozlovskaya LI, Shishova AA, Kovpak AA, Ivin YY, Piniaeva AN, Oksanich AS, Volok VP, Osolodkin DI, Ishmukhametov AA, Egorov AM, Shaitan KV, Kirpichnikov MP, Sokolova OS. Structural characterization of β-propiolactone inactivated severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) particles. Microsc Res Tech 2022; 85:562-569. [PMID: 34498784 PMCID: PMC8646525 DOI: 10.1002/jemt.23931] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/22/2021] [Indexed: 12/23/2022]
Abstract
The severe COVID-19 pandemic drives the research toward the SARS-CoV-2 virion structure and the possible therapies against it. Here, we characterized the β-propiolactone inactivated SARS-CoV-2 virions using transmission electron microscopy (TEM) and atomic force microscopy (AFM). We compared the SARS-CoV-2 samples purified by two consecutive chromatographic procedures (size exclusion chromatography [SEC], followed by ion-exchange chromatography [IEC]) with samples purified by ultracentrifugation. The samples prepared using SEC and IEC retained more spikes on the surface than the ones prepared using ultracentrifugation, as confirmed by TEM and AFM. TEM showed that the spike (S) proteins were in the pre-fusion conformation. Notably, the S proteins could be recognized by specific monoclonal antibodies. Analytical TEM showed that the inactivated virions retained nucleic acid. Altogether, we demonstrated that the inactivated SARS-CoV-2 virions retain the structural features of native viruses and provide a prospective vaccine candidate.
Collapse
Affiliation(s)
- Dmitry V. Bagrov
- Faculty of BiologyLomonosov Moscow State UniversityMoscowRussia
- Faculty of ChemistryLomonosov Moscow State UniversityMoscowRussia
| | | | - Andrey V. Moiseenko
- Faculty of BiologyLomonosov Moscow State UniversityMoscowRussia
- N. N. Semenov Federal Research Center for Chemical PhysicsRussian Academy of SciencesMoscowRussia
| | | | | | - Petr А. Zaitsev
- Faculty of BiologyLomonosov Moscow State UniversityMoscowRussia
| | - Liubov I. Kozlovskaya
- Chumakov Federal Scientific Center for Research and Development of Immune‐and‐Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis)MoscowRussia
- Institute of Translational Medicine and BiotechnologySechenov First Moscow State Medical UniversityMoscowRussia
| | - Anna A. Shishova
- Chumakov Federal Scientific Center for Research and Development of Immune‐and‐Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis)MoscowRussia
- Institute of Translational Medicine and BiotechnologySechenov First Moscow State Medical UniversityMoscowRussia
| | - Anastasia A. Kovpak
- Chumakov Federal Scientific Center for Research and Development of Immune‐and‐Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis)MoscowRussia
| | - Yury Y. Ivin
- Chumakov Federal Scientific Center for Research and Development of Immune‐and‐Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis)MoscowRussia
| | - Anastasia N. Piniaeva
- Chumakov Federal Scientific Center for Research and Development of Immune‐and‐Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis)MoscowRussia
| | | | - Viktor P. Volok
- Faculty of BiologyLomonosov Moscow State UniversityMoscowRussia
- Chumakov Federal Scientific Center for Research and Development of Immune‐and‐Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis)MoscowRussia
| | - Dmitry I. Osolodkin
- Faculty of ChemistryLomonosov Moscow State UniversityMoscowRussia
- Chumakov Federal Scientific Center for Research and Development of Immune‐and‐Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis)MoscowRussia
- Institute of Translational Medicine and BiotechnologySechenov First Moscow State Medical UniversityMoscowRussia
| | - Aydar A. Ishmukhametov
- Chumakov Federal Scientific Center for Research and Development of Immune‐and‐Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis)MoscowRussia
- Institute of Translational Medicine and BiotechnologySechenov First Moscow State Medical UniversityMoscowRussia
| | - Alexey M. Egorov
- Faculty of BiologyLomonosov Moscow State UniversityMoscowRussia
- N. N. Semenov Federal Research Center for Chemical PhysicsRussian Academy of SciencesMoscowRussia
- Mechnikov Research Institute of Vaccines and SeraMoscowRussia
| | - Konstantin V. Shaitan
- Faculty of BiologyLomonosov Moscow State UniversityMoscowRussia
- N. N. Semenov Federal Research Center for Chemical PhysicsRussian Academy of SciencesMoscowRussia
| | | | - Olga S. Sokolova
- Faculty of BiologyLomonosov Moscow State UniversityMoscowRussia
- Biology DepartmentMSU‐BIT UniversityShenzhenChina
| |
Collapse
|