Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
1.
Viruses ; 15(5)2023 05 11.
Article in English | MEDLINE | ID: covidwho-20243425

ABSTRACT

Antibody-dependent enhancement of infection (ADE) is clinically relevant to Dengue virus (DENV) infection and poses a major risk to the application of monoclonal antibody (mAb)-based therapeutics against related flaviviruses such as the Zika virus (ZIKV). Here, we tested a two-tier approach for selecting non-cross-reactive mAbs combined with modulating Fc glycosylation as a strategy to doubly secure the elimination of ADE while preserving Fc effector functions. To this end, we selected a ZIKV-specific mAb (ZV54) and generated three ZV54 variants using Chinese hamster ovary cells and wild-type (WT) and glycoengineered ΔXF Nicotiana benthamiana plants as production hosts (ZV54CHO, ZV54WT, and ZV54ΔXF). The three ZV54 variants shared an identical polypeptide backbone, but each exhibited a distinct Fc N-glycosylation profile. All three ZV54 variants showed similar neutralization potency against ZIKV but no ADE activity for DENV infection, validating the importance of selecting the virus/serotype-specific mAbs for avoiding ADE by related flaviviruses. For ZIKV infection, however, ZV54CHO and ZV54ΔXF showed significant ADE activity while ZV54WT completely forwent ADE, suggesting that Fc glycan modulation may yield mAb glycoforms that abrogate ADE even for homologous viruses. In contrast to the current strategies for Fc mutations that abrogate all effector functions along with ADE, our approach allowed the preservation of effector functions as all ZV54 glycovariants retained antibody-dependent cellular cytotoxicity (ADCC) against the ZIKV-infected cells. Furthermore, the ADE-free ZV54WT demonstrated in vivo efficacy in a ZIKV-infection mouse model. Collectively, our study provides further support for the hypothesis that antibody-viral surface antigen and Fc-mediated host cell interactions are both prerequisites for ADE, and that a dual-approach strategy, as shown herein, contributes to the development of highly safe and efficacious anti-ZIKV mAb therapeutics. Our findings may be impactful to other ADE-prone viruses, including SARS-CoV-2.


Subject(s)
COVID-19 , Dengue Virus , Dengue , Flavivirus , Zika Virus Infection , Zika Virus , Animals , Mice , Cricetinae , Zika Virus/genetics , CHO Cells , Dengue Virus/genetics , Cricetulus , SARS-CoV-2 , Antibodies, Viral , Antibodies, Monoclonal/therapeutic use , Cross Reactions , Antibodies, Neutralizing/therapeutic use
2.
Antibodies (Basel) ; 12(2)2023 May 15.
Article in English | MEDLINE | ID: covidwho-2326032

ABSTRACT

To combat infectious diseases, vaccines are considered the best prophylactic strategy for a wide range of the population, but even when vaccines are effective, the administration of therapeutic antibodies against viruses could provide further treatment options, particularly for vulnerable groups whose immunity against the viruses is compromised. Therapeutic antibodies against dengue are ideally engineered to abrogate binding to Fcγ receptors (FcγRs), which can induce antibody-dependent enhancement (ADE). However, the Fc effector functions of neutralizing antibodies against SARS-CoV-2 have recently been reported to improve post-exposure therapy, while they are dispensable when administered as prophylaxis. Hence, in this report, we investigated the influence of Fc engineering on anti-virus efficacy using the anti-dengue/Zika human antibody SIgN-3C and found it affected the viremia clearance efficacy against dengue in a mouse model. Furthermore, we demonstrated that complement activation through antibody binding to C1q could play a role in anti-dengue efficacy. We also generated a novel Fc variant, which displayed the ability for complement activation but showed very low FcγR binding and an undetectable level of the risk of ADE in a cell-based assay. This Fc engineering approach could make effective and safe anti-virus antibodies against dengue, Zika and other viruses.

3.
Immunol Lett ; 259: 21-23, 2023 07.
Article in English | MEDLINE | ID: covidwho-2323977

ABSTRACT

Inactivated vaccine is one of the platforms employed in COVID-19 vaccines. Inactivated vaccines have been associated with concerns of antibody-dependent enhancement (ADE) and original antigenic sin (OAS), which are related to non-neutralising or poorly neutralising antibodies against the pathogen. Since inactivated COVID-19 vaccines use whole-SARS-CoV-2 virus as the immunogen, they are expected to generate antibodies against non-spike structural proteins, which are highly conservative across variants of SARS-CoV-2. These antibodies against non-spike structural proteins have found to be largely non-neutralising or poorly neutralising in nature. Hence, inactivated COVID-19 vaccines could possibly be associated with ADE and OAS, especially as novel variants emerge. This article explores the potential concern of ADE and OAS in the context of inactivated COVID-19 vaccine, and outlines the future research directions.


Subject(s)
COVID-19 Vaccines , COVID-19 Vaccines/immunology , Antibodies, Viral/immunology , COVID-19/prevention & control , Humans
4.
Viral Immunol ; 35(9): 586-596, 2022 11.
Article in English | MEDLINE | ID: covidwho-2313628

ABSTRACT

Infection caused by the Zika virus (ZIKV) can lead to serious neurological complications such as microcephaly in neonates. At present, no approved ZIKV vaccine is available, but few vaccine candidates are undergoing clinical trial. One major challenge faced is antibody-dependent enhancement (ADE) reaction that may provoke severe outcome in subsequent infection by ZIKV or other flaviviruses. Thus, more efforts should be dedicated to understanding ADE in designing a safe and effective vaccine to minimize the consequence of the potentially fatal infection's complications and to tackle potential ZIKV reemergence. This review discusses different types of ZIKV vaccine candidates that are currently underway in various stages of preclinical and clinical evaluations.


Subject(s)
Viral Vaccines , Zika Virus Infection , Zika Virus , Infant, Newborn , Humans , Antibody-Dependent Enhancement , Antibodies, Viral
5.
Cureus ; 14(11): e31877, 2022 Nov.
Article in English | MEDLINE | ID: covidwho-2307222

ABSTRACT

The COVID-19 pandemic caused by SARS-CoV-2 spread across many countries between 2020 and 2022. The similarities in clinical presentation with other endemic diseases pose a challenge to physicians in effectively diagnosing and treating the infection. Approximately 129 nations have a risk of dengue infection, and more than 100 of those are endemic to dengue. During the COVID-19 pandemic, the number of dengue cases decreased in many countries owing to the isolation measures followed. However, the common clinical presentation between them has led to misdiagnosis. Both COVID-19 and dengue fever cause a surge in pro-inflammatory cytokines and chemokines, thus sharing a common pathophysiology. False positive serological test results also posed difficulty differentiating between COVID-19 and dengue fever. This review aims to compare the clinical features, pathophysiology, and immune response between dengue and COVID-19, to benefit public health management during the pandemic.

6.
Annu Rev Med ; 73: 1-16, 2022 01 27.
Article in English | MEDLINE | ID: covidwho-2268653

ABSTRACT

Prophylactic and therapeutic drugs are urgently needed to combat coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Over the past year, SARS-CoV-2 neutralizing antibodies have been developed for preventive or therapeutic uses. While neutralizing antibodies target the spike protein, their neutralization potency and breadth vary according to recognition epitopes. Several potent SARS-CoV-2 antibodies have shown degrees of success in preclinical or clinical trials, and the US Food and Drug Administration has issued emergency use authorization for two neutralizing antibody cocktails.Nevertheless, antibody therapy for SARS-CoV-2 still faces potential challenges, including emerging viral variants of concern that have antibody-escape mutations and the potential for antibody-mediated enhancement of infection or inflammation. This review summarizes representative SARS-CoV-2 neutralizing antibodies that have been reported and discusses prospects and challenges for the development of the next generation of COVID-19 preventive or therapeutic antibodies.


Subject(s)
COVID-19 , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing , Humans , SARS-CoV-2 , Spike Glycoprotein, Coronavirus
7.
Microorganisms ; 11(2)2023 Feb 13.
Article in English | MEDLINE | ID: covidwho-2243948

ABSTRACT

Severe cases of COVID-19 continue to put pressure on medical operations by prolonging hospitalization, occupying intensive care beds, and forcing medical personnel to undergo harsh labor. The eradication of SARS-CoV-2 through vaccine development has yet to be achieved, mainly due to the appearance of multiple mutant-incorporating strains. The present study explored the utility of human intravenous immunoglobulin (IVIG) preparations in suppressing the aggravation of any COVID-19 infection using a SARS-CoV-2 pseudovirus assay. Our study revealed the existence of IgG antibodies in human IVIG preparations, which recognized the spike protein of SARS-CoV-2. Remarkably, the pretreatment of ACE2/TMPRSS2-expressing host cells (HEK293T cells) with IVIG preparations (10 mg/mL) inhibited approximately 40% entry of SARS-CoV-2 pseudovirus even at extremely low concentrations of IgG (0.16-1.25 mg/mL). In contrast, the antibody-dependent enhancement of viral entry was confirmed when SARS-CoV-2 pseudovirus was treated with some products at an IgG concentration of 10 mg/mL. Our data suggest that IVIG may contribute to therapy for COVID-19, including for cases caused by SARS-CoV-2 variants, since IVIG binds not only to the spike proteins of the virus, but also to human ACE2/TMPRSS2. An even better preventive effect can be expected with blood collected after the start of the COVID-19 pandemic.

8.
Vis J Emerg Med ; 30: 101602, 2023 Jan.
Article in English | MEDLINE | ID: covidwho-2219203
9.
Aims Allergy and Immunology ; 7(1):1-23, 2023.
Article in English | Web of Science | ID: covidwho-2201198

ABSTRACT

Mast cells (MCs) are a part of the innate immune system and express receptors for microbial and viral pathogens characteristic of this system. The pathological role of MCs has been demonstrated for a number of highly virulent viral infections. The role of MCs and their Fc receptors for IgE in the immediate-type hypersensitivity reactions and in immunocomplex reactions is well-known, although the role of MCs and their Fc receptors for IgG (Fc gamma R) in immunocomplex processes is much less studied. Antibody-dependent enhancement syndrome (ADE) has been observed in a number of viral infections and is associated with greater secondary infection. ADE is enhanced by virus-specific antibodies, which are not involved in the virus penetration into the cell but are capable of forming immune complexes. The role of MCs in ADE is well-established for dengue infection, RSV infection and coronavirus (CoV) infection. The involvement of IgG-mediated mast cell responses in other human viral infections including Coronavirus disease 2019 (COVID-19) is poorly understood. Recently discovered mast cell activation disease is considered one of the causes of severe post -infectious complications in COVID-19. If the role of MCs in the pathogenesis of severe viral infections, including ADE in recurrent viral infection is clarified, these cells and the products they release may serve as promising targets for such therapeutic agents as histamine receptor blockers or membrane stabilizers to prevent possible complications.

10.
Nano Lett ; 23(2): 619-628, 2023 01 25.
Article in English | MEDLINE | ID: covidwho-2185488

ABSTRACT

Anti-spike neutralizing antibodies (S NAbs) have been developed for prevention and treatment against COVID-19. The nanoscopic characterization of the dynamic interaction between spike proteins and S NAbs remains difficult. By using high-speed atomic force microscopy (HS-AFM), we elucidate the molecular property of an S NAb and its interaction with spike proteins. The S NAb appeared as monomers with a Y conformation at low density and formed hexameric oligomers at high density. The dynamic S NAb-spike protein interaction at RBD induces neither RBD opening nor S1 subunit shedding. Furthermore, the interaction was stable at endosomal pH. These findings indicated that the S NAb could have a negligible risk of antibody-dependent enhancement. Dynamic movement of spike proteins on small extracellular vesicles (S sEV) resembled that on SARS-CoV-2. The sensitivity of variant S sEVs to S NAb could be evaluated using HS-AFM. Altogether, we demonstrate a nanoscopic assessment platform for evaluating the binding property of S NAbs.


Subject(s)
COVID-19 , Humans , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Antibodies, Viral , Antibodies, Neutralizing
11.
Open Forum Infectious Diseases ; 9(Supplement 2):S489, 2022.
Article in English | EMBASE | ID: covidwho-2189795

ABSTRACT

Background. AZD7442-a combination of 2 human, extended-half-life, SARS-CoV-2-neutralizing monoclonal antibodies (mAbs) (tixagevimab/cilgavimab)-has received US Food and Drug Administration emergency use authorization for COVID-19 prevention in immunocompromised individuals. We evaluated the effect of AZD7442 in prevention and treatment settings in Syrian hamsters challenged with SARS-CoV-2. Methods. Hamsters received intraperitoneal isotype control mAb (2 mg) or AZD7442 (0.002-2 mg) 1 day before intranasal (IN) SARS-CoV-2 challenge (USA-WA1/2020;1x105 plaque-forming units) in prevention;OR control mAb (5 mg) or AZD7442 (0.5-5 mg) 1 day after IN SARS-CoV-2 challenge in treatment. The impact of AZD7442 on lung viral RNA and pathology and AZD7442 serum levels was assessed on Days 3 and 7 post infection. Body weight was recorded daily through Day 7. Results. With AZD7442 prevention, lower lung viral loads were observed compared to controls;at Day 3 post infection, lowest infectious virus titer and viral subgenomic mRNA (sgmRNA) levels were seen with doses >=0.2 mg AZD7442. Concomitantly, increased serum levels of AZD7442 were observed. By Day 7, infectious virus titer and sgmRNA fell below the level of detection (LOD) at all doses tested. Moreover, AZD7442 at doses >=0.2 mg protected hamsters from weight loss versus controls. Lung pathology scores (scale: 0 [normal] to 25 [most severe]) were generally dose dependent, with mean scores of < 2 for AZD7442 versus 10 for controls, indicating less SARS-CoV-2-induced inflammation and alveolar damage in hamsters given AZD7442. Lower AZD7442 doses were associated with mean pathology scores similar to controls. With AZD7442 treatment, infectious virus titers were below the LOD at Day 3 post infection and at Day 7 for sgmRNA, for all doses tested. Mean lung pathology score was <2 for AZD7442 versus 12 for controls. AZD7442 doses >=0.5 mg protected against weight loss relative to controls. Conclusion. In a SARS-CoV-2 challenge model, AZD7442 administered as prevention or treatment led to significantly lower lung viral loads and improved lung pathology, without weight loss. There was also no evidence that AZD7442 mediated antibody-dependent enhancement of disease or infection.

12.
Front Immunol ; 13: 1008285, 2022.
Article in English | MEDLINE | ID: covidwho-2154728

ABSTRACT

Since immune system and internal environment in vivo are large and complex, the interpretation of the observed immune effect from the perspective of a single immune cell or antibody seems a little feeble. Many studies have shown that specific antibodies against " former" viruses have a reduced ability to neutralize "new" mutant strains. However, there is no comprehensive and clear view of whether there will be Antibody-dependent enhancement (ADE). We review the latest relevant studies, hoping to explain the ADE of SARS-CoV-2 infection sometimes observed in some patients.


Subject(s)
Antibody-Dependent Enhancement , COVID-19 , Humans , SARS-CoV-2 , Antibodies, Viral
13.
Chinese Journal of New Drugs ; 31(21):2073-2081, 2022.
Article in Chinese | EMBASE | ID: covidwho-2111995

ABSTRACT

Antibody-based biological products have gradually become a new strategy for the treatment of infectious diseases. In the past few years, especially after the outbreak of the COVID-19 in 2019, researches on therapeutic SARS-CoV-2 antibodies have greatly developed. Researchers around the world have developed a series of antibody treatment programs with extremely high efficiency to fight against COVID-19. From the early days of the pandemic, therapeutic antibodies were only used for emergency treatment of clinically severe patients, now they can be used for both pre-exposure prophylaxis and post-infection treatment. We summarize the research progress of therapeutic antibodies for SARS-CoV-2, including convalescent plasma, animal antiserum, marketed mAbs, non-targeted therapeutic mAbs, and bispecific antibodies, etc. The limitations and future application prospects of the SARS-CoV-2 therapeutic mAbs are also discussed. Copyright © 2022, Chinese Journal of New Drugs Co. Ltd. All right reserved.

14.
Front Med (Lausanne) ; 9: 952697, 2022.
Article in English | MEDLINE | ID: covidwho-2099173

ABSTRACT

Currently, neutralizing antibody and vaccine strategies have been developed by targeting the SARS-CoV-2 strain identified during the early phase of the pandemic. Early studies showed that the ability of SARS-CoV-2 RBD or NTD antibodies to elicit infection enhancement in vivo is still controversial. There are growing concerns that the plasma and neutralizing antibodies from convalescent patients or people receiving vaccines mediate ADE of SARS-CoV-2 variants infections in immune cells. Here, we constructed engineered double-mutant variants containing an RBD mutation and D614G in the spike (S) protein and natural epidemic variants to gain insights into the correlation between the mutations in S proteins and the ADE activities and tested whether convalescent plasma and TOP10 neutralizing antibodies in our laboratory mediated the ADE effects of these SARS-CoV-2 variants. We found that one out of 29 convalescent plasma samples caused the ADE effect of pandemic variant B.1.1.7 and that the ADE effect of wild-type SARS-CoV-2 was not detected for any of these plasma samples. Only one antibody, 55A8, from the same batch of convalescent patients mediated the ADE effects of multiple SARS-CoV-2 variants in vitro, including six double-mutant variants and four epidemic variants, suggesting that ADE activities may be closely related to the antibody itself and the SARS-CoV-2 variants' S proteins. Moreover, the ADE activity of 55A8 depended on FcγRII on immune cells, and the introduction of LALA mutations at the Fc end of 55A8 eliminated the ADE effects in vitro, indicating that 55A8LALA may be a clinical drug used to prevent SARS-CoV-2 variants. Altogether, ADE may occur in rare convalescent patients or vaccinees with ADE-active antibodies who are then exposed to a SARS-CoV-2 variant. These data suggested that potential neutralizing antibodies may need to undergo ADE screening tests for SARS-CoV-2 variants, which should aid in the future design of effective antibody-based therapies.

15.
Vaccine ; 40(49): 7141-7150, 2022 Nov 22.
Article in English | MEDLINE | ID: covidwho-2086812

ABSTRACT

The mass vaccination program has been actively promoted since the end of 2020. However, waning immunity, antibody-dependent enhancement (ADE), and increased transmissibility of variants make the herd immunity untenable and the implementation of dynamic zero-COVID policy challenging in China. To explore how long the vaccination program can prevent China at low resurgence risk, and how these factors affect the long-term trajectory of the COVID-19 epidemics, we developed a dynamic transmission model of COVID-19 incorporating vaccination and waning immunity, calibrated using the data of accumulative vaccine doses administered and the COVID-19 epidemic in 2020 in mainland China. The prediction suggests that the vaccination coverage with at least one dose reach 95.87%, and two doses reach 77.92% on 31 August 2021. However, despite the mass vaccination, randomly introducing infected cases in the post-vaccination period causes large outbreaks quickly with waning immunity, particularly for SARS-CoV-2 variants with higher transmissibility. The results showed that with the current vaccination program and 50% of the population wearing masks, mainland China can be protected at low resurgence risk until 8 January 2023. However, ADE and higher transmissibility for variants would significantly shorten the low-risk period by over 1 year. Furthermore, intermittent outbreaks can occur while the peak values of the subsequent outbreaks decrease, indicating that subsequent outbreaks boosted immunity in the population level, further indicating that follow-up vaccination programs can help mitigate or avoid the possible outbreaks. The findings revealed that the integrated effects of multiple factors: waning immunity, ADE, relaxed interventions, and higher variant transmissibility, make controlling COVID-19 challenging. We should prepare for a long struggle with COVID-19, and not entirely rely on the COVID-19 vaccine.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , COVID-19/epidemiology , COVID-19/prevention & control , Antibody-Dependent Enhancement , COVID-19 Vaccines , Vaccination/methods , China/epidemiology
16.
Coronavirus Drug Discovery: Volume 1: SARS-CoV-2 (COVID-19) Prevention, Diagnosis, and Treatment ; : 227-247, 2022.
Article in English | Scopus | ID: covidwho-2048789

ABSTRACT

SARS-CoV-2 is a well-known viral strain that causes COVID-19. The disease became a pandemic in early 2020 and infected millions of people and killed hundreds of thousands of people worldwide. Vaccine development against the disease was accelerated with multiple collaborations among research institutions in order to shorten the duration that vaccine development normally takes. Prior coronavirus vaccines present a basis on which vaccines against the current strain can be developed with much speed and relative ease. Among the patented coronavirus vaccines, DNA-based vaccine had the most patents registered which must have clues to guide the efforts in the current works. This work presents some progress on COVID-19 vaccine development and also possible animal venom protein sources that can potentially be used in the pipeline. The future of COVID-19 vaccine is bright with the heightened collaborative efforts and data sharing opportunities that the pandemic has brought among researchers. © 2022 Elsevier Inc. All rights reserved.

17.
Int J Mol Sci ; 23(19)2022 Sep 26.
Article in English | MEDLINE | ID: covidwho-2043780

ABSTRACT

Global vaccination against the SARS-CoV-2 virus has proved to be highly effective. However, the possibility of antibody-dependent enhancement of infection (ADE) upon vaccination remains underinvestigated. Here, we aimed to theoretically determine conditions for the occurrence of ADE in COVID-19. We developed a series of mathematical models of antibody response: model Ab-a model of antibody formation; model Cv-a model of infection spread in the body; and a complete model, which combines the two others. The models describe experimental data on SARS-CoV and SARS-CoV-2 infections in humans and cell cultures, including viral load dynamics, seroconversion times and antibody concentration kinetics. The modelling revealed that a significant proportion of macrophages can become infected only if they bind antibodies with high probability. Thus, a high probability of macrophage infection and a sufficient amount of pre-existing antibodies are necessary for the development of ADE in SARS-CoV-2 infection. However, from the point of view of the dynamics of pneumocyte infection, the two cases where the body has a high concentration of preexisting antibodies and a high probability of macrophage infection and where there is a low concentration of antibodies in the body and no macrophage infection are indistinguishable. This conclusion could explain the lack of confirmed ADE cases for COVID-19.


Subject(s)
COVID-19 , Severe acute respiratory syndrome-related coronavirus , Antibodies, Viral , Antibody-Dependent Enhancement , Humans , SARS-CoV-2
18.
Biomed Eng Adv ; 4: 100054, 2022 Dec.
Article in English | MEDLINE | ID: covidwho-2031157

ABSTRACT

With severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as an emergent human virus since December 2019, the world population is susceptible to coronavirus disease 2019 (COVID-19). SARS-CoV-2 has higher transmissibility than the previous coronaviruses, associated by the ribonucleic acid (RNA) virus nature with high mutation rate, caused SARS-CoV-2 variants to arise while circulating worldwide. Neutralizing antibodies are identified as immediate and direct-acting therapeutic against COVID-19. Single-domain antibodies (sdAbs), as small biomolecules with non-complex structure and intrinsic stability, can acquire antigen-binding capabilities comparable to conventional antibodies, which serve as an attractive neutralizing solution. SARS-CoV-2 spike protein attaches to human angiotensin-converting enzyme 2 (ACE2) receptor on lung epithelial cells to initiate viral infection, serves as potential therapeutic target. sdAbs have shown broad neutralization towards SARS-CoV-2 with various mutations, effectively stop and prevent infection while efficiently block mutational escape. In addition, sdAbs can be developed into multivalent antibodies or inhaled biotherapeutics against COVID-19.

19.
Front Immunol ; 13: 941923, 2022.
Article in English | MEDLINE | ID: covidwho-2022721

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spread globally since December 2019. Several studies reported that SARS-CoV-2 infections may produce false-positive reactions in dengue virus (DENV) serology tests and vice versa. However, it remains unclear whether SARS-CoV-2 and DENV cross-reactive antibodies provide cross-protection against each disease or promote disease severity. In this study, we confirmed that antibodies against the SARS-CoV-2 spike protein and its receptor-binding domain (S1-RBD) were significantly increased in dengue patients compared to normal controls. In addition, anti-S1-RBD IgG purified from S1-RBD hyperimmune rabbit sera could cross-react with both DENV envelope protein (E) and nonstructural protein 1 (NS1). The potential epitopes of DENV E and NS1 recognized by these antibodies were identified by a phage-displayed random peptide library. In addition, DENV infection and DENV NS1-induced endothelial hyperpermeability in vitro were inhibited in the presence of anti-S1-RBD IgG. Passive transfer anti-S1-RBD IgG into mice also reduced prolonged bleeding time and decreased NS1 seral level in DENV-infected mice. Lastly, COVID-19 patients' sera showed neutralizing ability against dengue infection in vitro. Thus, our results suggest that the antigenic cross-reactivity between the SARS-CoV-2 S1-RBD and DENV can induce the production of anti-SARS-CoV-2 S1-RBD antibodies that cross-react with DENV which may hinder dengue pathogenesis.


Subject(s)
COVID-19 , Dengue Virus , Dengue , Animals , Antibodies, Viral , Humans , Immunoglobulin G , Mice , Rabbits , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Viral Nonstructural Proteins
20.
Cell Biol Int ; 46(12): 2009-2017, 2022 Dec.
Article in English | MEDLINE | ID: covidwho-2013398

ABSTRACT

After more than 2 years of the coronavirus disease 2019 pandemic caused by severe acute respiratory syndrome coronavirus 2, several questions have remained unanswered that affected our daily lives. Although substantial vaccine development could resist this challenge, emerging new variants in different countries could be considered as potent concerns regarding the adverse effects of reinfection or postvaccination. Precisely, these concerns address some significant and probable outcomes in vaccinated or reinfected models, followed by some virus challenges, such as antibody-dependent enhancement and cytokine storm. Therefore, the importance of evaluating the effectiveness of neutralizing antibodies (nAbs) elicited by vaccination and the rise of new variants must be addressed.


Subject(s)
COVID-19 Vaccines , COVID-19 , Humans , Antibodies, Viral , COVID-19/prevention & control , COVID-19 Vaccines/adverse effects , SARS-CoV-2 , Viral Vaccines/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL