Your browser doesn't support javascript.
Montrer: 20 | 50 | 100
Résultats 1 - 11 de 11
Filtre
1.
Elife ; 112022 08 25.
Article Dans Anglais | MEDLINE | ID: covidwho-2025329

Résumé

Large-scale populations in the world have been vaccinated with COVID-19 vaccines, however, breakthrough infections of SARS-CoV-2 are still growing rapidly due to the emergence of immune-evasive variants, especially Omicron. It is urgent to develop effective broad-spectrum vaccines to better control the pandemic of these variants. Here, we present a mosaic-type trimeric form of spike receptor-binding domain (mos-tri-RBD) as a broad-spectrum vaccine candidate, which carries the key mutations from Omicron and other circulating variants. Tests in rats showed that the designed mos-tri-RBD, whether used alone or as a booster shot, elicited potent cross-neutralizing antibodies against not only Omicron but also other immune-evasive variants. Neutralizing antibody ID50 titers induced by mos-tri-RBD were substantially higher than those elicited by homo-tri-RBD (containing homologous RBDs from prototype strain) or the BIBP inactivated COVID-19 vaccine (BBIBP-CorV). Our study indicates that mos-tri-RBD is highly immunogenic, which may serve as a broad-spectrum vaccine candidate in combating SARS-CoV-2 variants including Omicron.


The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic continues to pose a serious threat to public health and has so far resulted in over six million deaths worldwide. Mass vaccination programs have reduced the risk of serious illness and death in many people, but the virus continues to persist and circulate in communities across the globe. Furthermore, the current vaccines may be less effective against the new variants of the virus, such as Omicron and Delta, which are continually emerging and evolving. Therefore, it is urgent to develop effective vaccines that can provide broad protection against existing and future forms of SARS-CoV-2. There are several different types of SARS-CoV-2 vaccine, but they all work in a similar way. They contain molecules that induce immune responses in individuals to help the body recognize and more effectively fight SARS-CoV-2 if they happen to encounter it in the future. These immune responses may be so specific that new variants of a virus may not be recognized by them. Therefore, a commonly used strategy for producing vaccines with broad protection is to make multiple vaccines that each targets different variants and then mix them together before administering to patients. Here, Zhang et al. took a different approach by designing a new vaccine candidate against SARS-CoV2 that contained three different versions of part of a SARS-CoV2 protein ­ the so-called spike protein ­ all linked together as one molecule. The different versions of the spike protein fragment were designed to include key features of the fragments found in Omicron and several other SARS-CoV-2 variants. The experiments found that this candidate vaccine elicited a much higher immune response against Omicron and other SARS-CoV-2 variants in rats than an existing SARS-CoV-2 vaccine. It was also effective as a booster shot after a first vaccination with the existing SARS-CoV-2 vaccine. These findings demonstrate that the molecule developed by Zhang et al. induces potent and broad immune responses against different variants of SARS-CoV-2 including Omicron in rats. The next steps following on from this work are to evaluate the safety and immunogenicity of this vaccine candidate in clinical trials. In the future, it may be possible to use a similar approach to develop new broad-spectrum vaccines against other viruses.


Sujets)
Vaccins contre la COVID-19 , COVID-19 , Animaux , Anticorps neutralisants , Anticorps antiviraux , Anticorps neutralisants à large spectre , COVID-19/prévention et contrôle , Humains , Rats , SARS-CoV-2/génétique , Glycoprotéine de spicule des coronavirus/composition chimique
2.
Adv Sci (Weinh) ; 9(14): e2104333, 2022 05.
Article Dans Anglais | MEDLINE | ID: covidwho-1782562

Résumé

Coronavirus disease 2019 (COVID-19) remains a global public health threat. Hence, more effective and specific antivirals are urgently needed. Here, COVID-19 hyperimmune globulin (COVID-HIG), a passive immunotherapy, is prepared from the plasma of healthy donors vaccinated with BBIBP-CorV (Sinopharm COVID-19 vaccine). COVID-HIG shows high-affinity binding to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein, the receptor-binding domain (RBD), the N-terminal domain of the S protein, and the nucleocapsid protein; and blocks RBD binding to human angiotensin-converting enzyme 2 (hACE2). Pseudotyped and authentic virus-based assays show that COVID-HIG displays broad-spectrum neutralization effects on a wide variety of SARS-CoV-2 variants, including D614G, Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), Kappa (B.1.617.1), Delta (B.1.617.2), and Omicron (B.1.1.529) in vitro. However, a significant reduction in the neutralization titer is detected against Beta, Delta, and Omicron variants. Additionally, assessments of the prophylactic and treatment efficacy of COVID-HIG in an Adv5-hACE2-transduced IFNAR-/- mouse model of SARS-CoV-2 infection show significantly reduced weight loss, lung viral loads, and lung pathological injury. Moreover, COVID-HIG exhibits neutralization potency similar to that of anti-SARS-CoV-2 hyperimmune globulin from pooled convalescent plasma. Overall, the results demonstrate the potential of COVID-HIG against SARS-CoV-2 infection and provide reference for subsequent clinical trials.


Sujets)
Vaccins contre la COVID-19 , COVID-19 , Globulines , Animaux , COVID-19/thérapie , Globulines/usage thérapeutique , Humains , Immunisation passive , Souris , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus ,
3.
Cell Discov ; 8(1): 17, 2022 Feb 15.
Article Dans Anglais | MEDLINE | ID: covidwho-1692628

Résumé

The continuous emergence of SARS-CoV-2 variants highlights the need of developing vaccines with broad protection. Here, according to the immune-escape capability and evolutionary convergence, the representative SARS-CoV-2 strains carrying the hotspot mutations were selected. Then, guided by structural and computational analyses, we present a mutation-integrated trimeric form of spike receptor-binding domain (mutI-tri-RBD) as a broadly protective vaccine candidate, which combined heterologous RBDs from different representative strains into a hybrid immunogen and integrated immune-escape hotspots into a single antigen. When compared with a homo-tri-RBD vaccine candidate in the stage of phase II trial, of which all three RBDs are derived from the SARS-CoV-2 prototype strain, mutI-tri-RBD induced significantly higher neutralizing antibody titers against the Delta and Beta variants, and maintained a similar immune response against the prototype strain. Pseudo-virus neutralization assay demonstrated that mutI-tri-RBD also induced broadly strong neutralizing activities against all tested 23 SARS-CoV-2 variants. The in vivo protective capability of mutI-tri-RBD was further validated in hACE2-transgenic mice challenged by the live virus, and the results showed that mutI-tri-RBD provided potent protection not only against the SARS-CoV-2 prototype strain but also against the Delta and Beta variants.

4.
Arch Virol ; 167(2): 459-470, 2022 Feb.
Article Dans Anglais | MEDLINE | ID: covidwho-1653515

Résumé

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has had a major impact on global human health. During the spread of SARS-CoV-2, weakened host immunity and the use of vaccines with low efficacy may result in the development of more-virulent strains or strains with resistance to existing vaccines and antibodies. The prevalence of SARS-CoV-2 mutant strains differs between regions, and this variation may have an impact on the effectiveness of vaccines. In this study, an epidemiological investigation of SARS-CoV-2 in Portugal was performed, and the VSV-ΔG-G* pseudovirus system was used to construct 12 spike protein epidemic mutants, D614G, A222V+D614G, B.1.1.7, S477N+D614G, P1162R+D614G+A222V, D839Y+D614G, L176F+D614G, B.1.1.7+L216F, B.1.1.7+M740V, B.1.258, B.1.258+L1063F, and B.1.258+N751Y. The mutant pseudoviruses were used to infect four susceptible cell lines (Huh7, hACE2-293T-293T, Vero, and LLC-MK2) and 14 cell lines overexpressing ACE2 from different species. Mutant strains did not show increased infectivity or cross-species transmission. Neutralization activity against these pseudoviruses was evaluated using mouse serum and 11 monoclonal antibodies. The neutralizing activity of immunized mouse serum was not significantly reduced with the mutant strains, but the mutant strains from Portugal could evade nine of the 11 monoclonal antibodies tested. Neutralization resistance was mainly caused by the mutations S477N, N439K, and N501Y in the spike-receptor binding domain. These findings emphasize the importance of SARS-CoV-2 mutation tracking in different regions for epidemic prevention and control.


Sujets)
COVID-19 , SARS-CoV-2 , Animaux , Anticorps neutralisants , Humains , Souris , Mutation , Portugal/épidémiologie , Glycoprotéine de spicule des coronavirus/génétique
5.
Bioorg Chem ; 116: 105309, 2021 11.
Article Dans Anglais | MEDLINE | ID: covidwho-1372894

Résumé

Six new polyketone metabolites, compounds (1-6) and seven known polyketone compounds (7-13) were isolated from Rhodiola tibetica endophytic fungus Alternaria sp. The structural elucidation of five new polyketone metabolites were elucidated on the basis of spectroscopic including 2D NMR and HRMS and spectrometric analysis. Inhibition rate evaluation revealed that compounds 1(EC50 = 0.02 mM), 3(EC50 = 0.3 mM), 6(EC50 = 0.07 mM), 8(EC50 = 0.1 mM) and 9(EC50 = 0.04 mM) had inhibitory effect on the SARS-CoV-2 virus.


Sujets)
Alternaria/composition chimique , Antiviraux/isolement et purification , Antiviraux/pharmacologie , Cétones/isolement et purification , Cétones/pharmacologie , Polymères/isolement et purification , Polymères/pharmacologie , SARS-CoV-2/effets des médicaments et des substances chimiques , Antiviraux/composition chimique , Humains , Cétones/composition chimique , Structure moléculaire , Polymères/composition chimique
6.
Chin Med J (Engl) ; 134(11): 1289-1298, 2021 Apr 28.
Article Dans Anglais | MEDLINE | ID: covidwho-1343718

Résumé

BACKGROUND: The significant morbidity and mortality resulted from the infection of a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) call for urgent development of effective and safe vaccines. We report the immunogenicity and safety of an inactivated SARS-CoV-2 vaccine, KCONVAC, in healthy adults. METHODS: Phase 1 and phase 2 randomized, double-blind, and placebo-controlled trials of KCONVAC were conducted in healthy Chinese adults aged 18 to 59 years. The participants in the phase 1 trial were randomized to receive two doses, one each on Days 0 and 14, of either KCONVAC (5 or 10 µg/dose) or placebo. The participants in the phase 2 trial were randomized to receive either KCONVAC (at 5 or 10 µg/dose) or placebo on Days 0 and 14 (0/14 regimen) or Days 0 and 28 (0/28 regimen). In the phase 1 trial, the primary safety endpoint was the proportion of participants experiencing adverse reactions/events within 28 days following the administration of each dose. In the phase 2 trial, the primary immunogenicity endpoints were neutralization antibody seroconversion and titer and anti-receptor-binding domain immunoglobulin G seroconversion at 28 days after the second dose. RESULTS: In the phase 1 trial, 60 participants were enrolled and received at least one dose of 5-µg vaccine (n = 24), 10-µg vaccine (n = 24), or placebo (n = 12). In the phase 2 trial, 500 participants were enrolled and received at least one dose of 5-µg vaccine (n = 100 for 0/14 or 0/28 regimens), 10-µg vaccine (n = 100 for each regimen), or placebo (n = 50 for each regimen). In the phase 1 trial, 13 (54%), 11 (46%), and seven (7/12) participants reported at least one adverse event (AE) after receiving 5-, 10-µg vaccine, or placebo, respectively. In the phase 2 trial, 16 (16%), 19 (19%), and nine (18%) 0/14-regimen participants reported at least one AE after receiving 5-, 10-µg vaccine, or placebo, respectively. Similar AE incidences were observed in the three 0/28-regimen treatment groups. No AEs with an intensity of grade 3+ were reported, expect for one vaccine-unrelated serious AE (foot fracture) reported in the phase 1 trial. KCONVAC induced significant antibody responses; 0/28 regimen showed a higher immune responses than that did 0/14 regimen after receiving two vaccine doses. CONCLUSIONS: Both doses of KCONVAC are well tolerated and able to induce robust immune responses in healthy adults. These results support testing 5-µg vaccine in the 0/28 regimen in an upcoming phase 3 efficacy trial. TRIAL REGISTRATION: http://www.chictr.org.cn/index.aspx (No. ChiCTR2000038804, http://www.chictr.org.cn/showproj.aspx?proj=62350; No. ChiCTR2000039462, http://www.chictr.org.cn/showproj.aspx?proj=63353).


Sujets)
COVID-19 , SARS-CoV-2 , Adulte , Vaccins contre la COVID-19 , Méthode en double aveugle , Humains , Vaccins inactivés/effets indésirables
7.
Zhongguo Bingdubing Zazhi = Chinese Journal of Viral Diseases ; - (3):209, 2021.
Article Dans Anglais | ProQuest Central | ID: covidwho-1329332

Résumé

Most viruses require their glycoproteins to be hydrolyzed in order to enter host cells.In some cases, the severability of viral glycoproteins is a determinant of their virulence.These viral glycoproteins can be cleaved by one or more proteases in host cells with furin as the common one.Furin further promotes viral infection by identifying specific amino acid sequences in glycoprotein precursors.Morever, furin plays a critical role in the infections of many viruses including human immunodeficiency virus, flavivirus, filamentavirus and coronavirus, and is considered as a promising drug treatment and prevention target.In this review, we summarize the biological background of furin and its hydrolysis function to viral glycoproteins.

8.
Bioorg Chem ; 115: 105196, 2021 10.
Article Dans Anglais | MEDLINE | ID: covidwho-1322004

Résumé

So far, there is still no specific drug against COVID-19. Taking compound 1 with anti-EBOV activity as the lead, fifty-four 12N-substituted aloperine derivatives were synthesized and evaluated for the anti-SARS-CoV-2 activities using pseudotyped virus model. Among them, 8a exhibited the most potential effects against both pseudotyped and authentic SARS-CoV-2, as well as SARS-CoV and MERS-CoV, indicating a broad-spectrum anti-coronavirus profile. The mechanism study disclosed that 8a might block a late stage of viral entry, mainly via inhibiting host cathepsin B activity rather than directly targeting cathepsin B protein. Also, 8a could significantly reduce the release of multiple inflammatory cytokines in a time- and dose-dependent manner, such as IL-6, IL-1ß, IL-8 and MCP-1, the major contributors to cytokine storm. Therefore, 8a is a promising agent with the advantages of broad-spectrum anti-coronavirus and anti-cytokine effects, thus worthy of further investigation.


Sujets)
Antiviraux/pharmacologie , Pipéridines/pharmacologie , Quinolizidines/pharmacologie , SARS-CoV-2/effets des médicaments et des substances chimiques , Pénétration virale/effets des médicaments et des substances chimiques , Animaux , Antiviraux/synthèse chimique , Antiviraux/pharmacocinétique , Antiviraux/toxicité , Cathepsine B/antagonistes et inhibiteurs , Chlorocebus aethiops , Cytokines/métabolisme , Cellules HEK293 , Humains , Mâle , Souris , Tests de sensibilité microbienne , Structure moléculaire , Pipéridines/synthèse chimique , Pipéridines/pharmacocinétique , Pipéridines/toxicité , Quinolizidines/synthèse chimique , Quinolizidines/pharmacocinétique , Quinolizidines/toxicité , Rat Sprague-Dawley , Relation structure-activité , Cellules Vero
9.
Signal Transduct Target Ther ; 6(1): 134, 2021 03 27.
Article Dans Anglais | MEDLINE | ID: covidwho-1152831

Résumé

To discover new drugs to combat COVID-19, an understanding of the molecular basis of SARS-CoV-2 infection is urgently needed. Here, for the first time, we report the crucial role of cathepsin L (CTSL) in patients with COVID-19. The circulating level of CTSL was elevated after SARS-CoV-2 infection and was positively correlated with disease course and severity. Correspondingly, SARS-CoV-2 pseudovirus infection increased CTSL expression in human cells in vitro and human ACE2 transgenic mice in vivo, while CTSL overexpression, in turn, enhanced pseudovirus infection in human cells. CTSL functionally cleaved the SARS-CoV-2 spike protein and enhanced virus entry, as evidenced by CTSL overexpression and knockdown in vitro and application of CTSL inhibitor drugs in vivo. Furthermore, amantadine, a licensed anti-influenza drug, significantly inhibited CTSL activity after SARS-CoV-2 pseudovirus infection and prevented infection both in vitro and in vivo. Therefore, CTSL is a promising target for new anti-COVID-19 drug development.


Sujets)
Antiviraux/pharmacologie , COVID-19/métabolisme , Cathepsine L , Inhibiteurs de la cystéine protéinase/pharmacologie , Développement de médicament , SARS-CoV-2/métabolisme , Glycoprotéine de spicule des coronavirus/métabolisme , Pénétration virale/effets des médicaments et des substances chimiques , Adolescent , Adulte , Sujet âgé , Animaux , COVID-19/génétique , Cathepsine L/antagonistes et inhibiteurs , Cathepsine L/génétique , Cathepsine L/métabolisme , Femelle , Humains , Mâle , Souris , Souris transgéniques , Adulte d'âge moyen , SARS-CoV-2/génétique , Glycoprotéine de spicule des coronavirus/génétique ,
10.
Cell ; 182(5): 1271-1283.e16, 2020 09 03.
Article Dans Anglais | MEDLINE | ID: covidwho-666099

Résumé

There is an urgent need for vaccines against coronavirus disease 2019 (COVID-19) because of the ongoing SARS-CoV-2 pandemic. Among all approaches, a messenger RNA (mRNA)-based vaccine has emerged as a rapid and versatile platform to quickly respond to this challenge. Here, we developed a lipid nanoparticle-encapsulated mRNA (mRNA-LNP) encoding the receptor binding domain (RBD) of SARS-CoV-2 as a vaccine candidate (called ARCoV). Intramuscular immunization of ARCoV mRNA-LNP elicited robust neutralizing antibodies against SARS-CoV-2 as well as a Th1-biased cellular response in mice and non-human primates. Two doses of ARCoV immunization in mice conferred complete protection against the challenge of a SARS-CoV-2 mouse-adapted strain. Additionally, ARCoV is manufactured as a liquid formulation and can be stored at room temperature for at least 1 week. ARCoV is currently being evaluated in phase 1 clinical trials.


Sujets)
ARN messager/génétique , ARN viral/génétique , Vaccins synthétiques/immunologie , Vaccins antiviraux/immunologie , Animaux , Anticorps neutralisants/immunologie , Sites de fixation , Vaccins contre la COVID-19 , Chlorocebus aethiops , Infections à coronavirus/génétique , Infections à coronavirus/immunologie , Infections à coronavirus/prévention et contrôle , Femelle , Cellules HEK293 , Cellules HeLa , Humains , Immunogénicité des vaccins , Injections musculaires , Macaca fascicularis , Mâle , Souris , Souris de lignée ICR , Nanoparticules/composition chimique , ARN messager/métabolisme , ARN viral/métabolisme , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/métabolisme , Lymphocytes auxiliaires Th1/immunologie , Efficacité du vaccin , Vaccins synthétiques/administration et posologie , Vaccins synthétiques/génétique , Cellules Vero , Vaccins antiviraux/administration et posologie , Vaccins antiviraux/génétique
11.
Cell Host Microbe ; 28(1): 124-133.e4, 2020 07 08.
Article Dans Anglais | MEDLINE | ID: covidwho-378130

Résumé

Since December 2019, a novel coronavirus SARS-CoV-2 has emerged and rapidly spread throughout the world, resulting in a global public health emergency. The lack of vaccine and antivirals has brought an urgent need for an animal model. Human angiotensin-converting enzyme II (ACE2) has been identified as a functional receptor for SARS-CoV-2. In this study, we generated a mouse model expressing human ACE2 (hACE2) by using CRISPR/Cas9 knockin technology. In comparison with wild-type C57BL/6 mice, both young and aged hACE2 mice sustained high viral loads in lung, trachea, and brain upon intranasal infection. Although fatalities were not observed, interstitial pneumonia and elevated cytokines were seen in SARS-CoV-2 infected-aged hACE2 mice. Interestingly, intragastric inoculation of SARS-CoV-2 was seen to cause productive infection and lead to pulmonary pathological changes in hACE2 mice. Overall, this animal model described here provides a useful tool for studying SARS-CoV-2 transmission and pathogenesis and evaluating COVID-19 vaccines and therapeutics.


Sujets)
Betacoronavirus/physiologie , Infections à coronavirus , Modèles animaux de maladie humaine , Souris de lignée C57BL , Pandémies , Pneumopathie virale , Vieillissement , Angiotensin-converting enzyme 2 , Animaux , Encéphale/virologie , COVID-19 , Systèmes CRISPR-Cas , Infections à coronavirus/anatomopathologie , Infections à coronavirus/virologie , Cytokines/sang , Techniques de knock-in de gènes , Poumon/anatomopathologie , Poumon/virologie , Pneumopathies interstitielles/anatomopathologie , Nez/virologie , Peptidyl-Dipeptidase A/génétique , Peptidyl-Dipeptidase A/métabolisme , Pneumopathie virale/anatomopathologie , Pneumopathie virale/virologie , ARN viral/analyse , SARS-CoV-2 , Estomac/virologie , Trachée/virologie , Charge virale , Réplication virale
SÉLECTION CITATIONS
Détails de la recherche