Kwok-Yung Yuen’s research while affiliated with The University of Hong Kong and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (406)


Generation and characterization of DelNS1-H5N1 vaccines
A Illustration of construction and generation of DelNS1-H5N1 vaccines. NCR, non-coding region. DCP: deletion of polybasic cleavage site. B Growth kinetics of DelNS1-cH5N1 and DelNS1-mH5N1 at 33 °C were analyzed in MDCK cells inoculated at a multiplicity of infection (MOI) of 0.01 (n = 3), and viral titers of DelNS1-cH5N1 and DelNS1-mH5N1 were measured in day 8 embryonated chicken eggs 48 h after inoculation with different initial doses (n = 3). Viral supernatants or allantoic fluids were collected and titrated by plaque assay in MDCK cells. C Virulence of DelNS1-cH5N1 and DelNS1-mH5N1 was examined in mice. Mice were inoculated intranasally with DelNS1-cH5N1 (10⁶ pfu, n = 8) or cattle H5N1 (5000 pfu, n = 8), DelNS1-mH5N1 (10⁶ pfu, n = 8) or mink H5N1 (10⁴ pfu, n = 8). Body weights were observed for 14 days. D Mice were inoculated intranasally with DelNS1-cH5N1 (10⁶ pfu, n = 3) or cattle H5N1 (5000 pfu, n = 4), with virus titers in lungs, nasal turbinates (NT) and brain tissues of mice being examined at day 2, day 4 and day 6 post infection. LOD limit of detection. Data represents mean values ± standard deviation (SD) of results. N/A not available. Statistical comparisons between means were performed by Student’s t-test (two-tailed). Cartoons were created in BioRender. Wang, P. (2025) https://BioRender.com/n81t049. Source data are provided as a Source Data file.
Immunogenicity evaluation of DelNS1-H5N1 vaccines
A Schedule of immunization, blood collection, and bronchoalveolar lavage (BAL) fluid and nasal wash collection from BALB/c mice. Mice (n = 8 for each group) were intranasally immunized with 10⁶ pfu DelNS1-cH5N1 or 10⁶ pfu DelNS1-mH5N1, or mock-immunized (PBS). B At week 3, serum were collected and tested for IgG titers against the cH5N1 (DelNS1-cH5N1) virus, mH5N1 (DelNS1-mH5N1) virus, or H5N1-HA1 protein. C Microneutralization (MN) titers were measured against live viruses. At week 4 post immunization, mice from all groups were sacrificed and BAL and nasal wash samples collected. D BAL IgA levels and E nasal wash IgA levels against cH5N1, mH5N1, or H5N1-HA1 were measured. LOD: limit of detection. Data represents the mean values ± SD of results. Statistical analysis was performed for (B) and (C) using one-way ANOVA followed by Dunn’s multiple comparisons test. Statistical analysis was performed for (D) and (E) using Student’s t-test (two-tailed). The mouse cartoon was created in BioRender. Wang, P. (2025) https://BioRender.com/n83i107. Source data are provided as a Source Data file.
DelNS1-H5N1 vaccines induce CD4+ and CD8+ T cell responses
A Schedule for immunization of BALB/c mice. At week 4 after immunization, 2 μg of PE-Cy5 conjugated CD45-specific antibody was injected intravenously via the tail vein 5 min before sacrifice (n = 8 for each group). B Lung cells and splenocytes were obtained and stained with Zombie, anti-CD8 and NP147 tetramer. Flow cytometry gating was performed to assess the frequency of live DelNS1-cH5N1-induced non-circulating (IV CD45-PE-Cy5 negative) NP-specific CD8+ T cells among lung cells and splenocytes (percentage IV-CD8 + NP147 tetramer+ out of all live IV-CD8+ T cells). C Lung or spleen cells were stimulated with NP147 or NP55 peptide overnight. Surface markers (CD69, CD103, CD4, CD8 and Zombie) were stained, followed by fixation, permeabilization, and intracellular staining for IFN-γ. DelNS1-cH5N1 induced tissue-resident memory cells in lungs and spleens were displayed (IV-IFNγ + CD69 + CD103 + CD8 + T cells and IV-IFNγ+CD69+CD4+ T cells out a percentage of all live IV- CD8+ and CD4+ T cells, respectively). D DelNS1-mH5N1 induced non-circulating T cells in lung cells and spleens (IV-CD8 + NP147 tetramer out of all live IV-CD8 T cells). E DelNS1-mH5N1 induced tissue-resident memory cells in lungs and spleen were displayed (IV-IFNγ+CD69+ CD103+CD8+ T cells and IV-IFNγ + CD69+ CD4+ T cells out a percentage of all live IV- CD8+ and CD4 + T cells, respectively). Data represents the mean values ± SD of results. Statistical comparisons between means were performed by Student’s t-test (two-tailed). The mouse cartoon was created in BioRender. Wang, P. (2025) https://BioRender.com/n83i107. Source data are provided as a Source Data file.
DelNS1-H5N1 vaccines provide protection against different 2.3.4.4b clade viruses
A Illustration of schedule of immunization, virus challenge and sacrifice for BALB/c mice. Mice were intranasally vaccinated with DelNS1-cH5N1 (10⁶ pfu, n = 32, two independent experiments), DelNS1-mH5N1 (10⁶ pfu, n = 32, two independent experiments) or mock-vaccinated (n = 16, two independent experiments), then challenged with either cattle H5N1 virus (5000 pfu) or mink H5N1 virus (10⁴ pfu) 4 weeks after immunization. B Mice immunized with DelNS1-cH5N1 and challenged with cattle H5N1 virus; body weights were monitored for 14 days and viral titers in the lungs, NT and brain were measured at 4 dpi. C Mice immunized with DelNS1-mH5N1 and challenged with mink H5N1 virus; body weights were monitored for 14 days and viral titers in the lungs, NT and brain were measured at 4 dpi. D DelNS1-cH5N1-vaccinated mice were challenged with mink H5N1 virus, and body weights monitored for 14 days and viral titers in the lungs, NT and brain measured at 4 dpi. E DelNS1-mH5N1-vaccinated mice were challenged with cattle H5N1, and body weights monitored for 14 days and viral titers in the lungs, NT and brain measured at 4 dpi. LOD: limit of detection. Data represents mean values ± SD of results. Statistical comparisons between means were performed by Student’s t-test (two-tailed). Mouse cartoon was created in BioRender. Wang, P. (2025) https://BioRender.com/n83i107. Source data are provided as a Source Data file.
Immunization reduces expression of pro-inflammatory genes in mice post-challenge
A Illustration of schedule of immunization, virus challenge and sacrifice for BALB/c mice. B Mice were intranasally vaccinated with DelNS1-cH5N1 (10⁶ pfu), or PBS (n = 4 for each group), then challenged with cattle H5N1 virus (5000 pfu) 4 weeks after immunization. Pro-inflammatory genes were measured by RT-qPCR after isolation of RNA from lungs at 4 dpi. C Mice were intranasally vaccinated with DelNS1-mH5N1 (10⁶ pfu), or PBS (n = 4 for each group), then challenged with mink H5N1 virus (10⁴ pfu) 4 weeks after immunization. Pro-inflammatory genes were measured by RT-qPCR after isolation of RNA from lungs at 4 dpi. Data represent mean values ± SD of results. Statistical analysis was performed using Student’s t-test (two-tailed). The mouse cartoon was created in BioRender. Wang, P. (2025) https://BioRender.com/n83i107. Source data are provided as a Source Data file.

+2

Intranasal influenza virus-vectored vaccine offers protection against clade 2.3.4.4b H5N1 infection in small animal models
  • Article
  • Full-text available

April 2025

·

73 Reads

Ying Liu

·

·

Shuang Ren

·

[...]

·

Pui Wang

The highly pathogenic avian influenza (HPAI) H5N1 virus has been endemic in aquatic birds since 1997, causing outbreaks in domestic poultry and occasional human infections worldwide. Recently, the cross-species transmission of a new reassortant variant from clade 2.3.4.4b of H5N1 to cattle in the US has heightened concerns regarding the expansion of host range and potential human infection. As eradicating the H5N1 virus from its reservoir is impossible, it is essential to prepare for a potential pandemic caused by an H5N1 derivative. Utilizing a deleted-NS1 live attenuated influenza viral vector vaccine system (DelNS1 LAIV), a system we have previously used in the development of a COVID-19 vaccine, we have rapidly developed an intranasal vaccine for cattle H5N1 and related clade 2.3.4.4b strains, based on publicly available sequences. Our research demonstrates that a single intranasal immunization can provide effective protection against lethal challenges from HPAI cattle or mink H5N1 variants, offering strong, sustained immunity after two months in female mouse and male hamster models. Immunogenicity analysis reveals that intranasal vaccination with DelNS1 LAIV induces robust neutralizing antibody, mucosal IgA and T cell responses in mice. It is crucial to further evaluate the DelNS1-H5N1 LAIV system to prepare for potential future H5N1 outbreaks in humans.

Download

Lineage-specific pathogenicity, immune evasion, and virological features of SARS-CoV-2 BA.2.86/JN.1 and EG.5.1/HK.3

October 2024

·

147 Reads

·

16 Citations

SARS-CoV-2 JN.1 with an additional L455S mutation on spike when compared with its parental variant BA.2.86 has outcompeted all earlier variants to become the dominant circulating variant. Recent studies investigated the immune resistance of SARS-CoV-2 JN.1 but additional factors are speculated to contribute to its global dominance, which remain elusive until today. Here, we find that SARS-CoV-2 JN.1 has a higher infectivity than BA.2.86 in differentiated primary human nasal epithelial cells (hNECs). Mechanistically, we demonstrate that the gained infectivity of SARS-CoV-2 JN.1 over BA.2.86 associates with increased entry efficiency conferred by L455S and better spike cleavage in hNECs. Structurally, S455 altered the mode of binding of JN.1 spike protein to ACE2 when compared to BA.2.86 spike at ACE2H34, and modified the internal structure of JN.1 spike protein by increasing the number of hydrogen bonds with neighboring residues. These findings indicate that a single mutation (L455S) enhances virus entry in hNECs and increases immune evasiveness, which contribute to the robust transmissibility of SARS-CoV-2 JN.1. We further evaluate the in vitro and in vivo virological characteristics between SARS-CoV-2 BA.2.86/JN.1 and EG.5.1/HK.3, and identify key lineage-specific features of the two Omicron sublineages that contribute to our understanding on Omicron antigenicity, transmissibility, and pathogenicity.


Fig. 5: Prophylactic efficacy of ZCP3B4, ZCP4C9 and ZCP4D5-1 against authentic Omicron BA.5.2, BQ.1.1 and XBB.1.5 in golden Syrian hamsters. (A) Experimental schedule and color coding for different treatment groups. Four groups of female hamsters received a single intraperitoneal injection of PBS (n = 6), 4.5 mg/kg of ZCP3B4 (n = 5), ZCP4C9 (n = 5) or ZCP4D5-1 (n = 5) one day before viral infection (−1 dpi). 24 h later (day 0), each group was challenged intranasally with a mixture of live Omicron BA.5.2, BQ.1.1 and XBB.1.5 (10 5 PFU/hamster). All animals were sacrificed on day 4 for final analysis. (B) Proportion of viral RNA copies in lungs and nasal turbinate (NT) homogenates of each group. The data is shown as mean ± SEM. (C) Daily body weight change of each group was measured after the viral infection. The data is shown as mean ± SEM. (D) Live viral plaque assay was used to quantify the number of infectious viruses in lungs and NT of each group. Log10-transformed plaque-forming unit (PFU) per mL was shown for each group. The dash line indicates the limit of detection. Each symbol represents an individual hamster with a line indicating the mean of each group. Statistics were generated using one-way ANOVA followed by Tukey's multiple comparisons test. **p < 0.01, ***p < 0.001, ****p < 0.0001; ns (not significant), p > 0.05. (E) Representative histopathology of lung tissues and NT from pre-treated hamsters after viral challenge. Tissue sections were stained with hematoxylin and eosin (H&E). For PBStreated hamsters, the infection could cause lung damage with alveolar septa thickening (black arrow), extensive inflammatory cell accumulation (blue arrow), homogeneously pink foci of edema (green arrow), and multifocal hemorrhage (red arrow). In NT, submucosal immune cell accumulation (blue arrow) as well as damage to the respiratory and olfactory epithelium (black arrow) are also indicated. The resolution is indicated by the scale bar.
Ultrapotent class I neutralizing antibodies post Omicron breakthrough infection overcome broad SARS-CoV-2 escape variants

September 2024

·

87 Reads

·

3 Citations

EBioMedicine

Background The spread of emerging SARS-CoV-2 immune escape sublineages, especially JN.1 and KP.2, has resulted in new waves of COVID-19 globally. The evolving memory B cell responses elicited by the parental Omicron variants to subvariants with substantial antigenic drift remain incompletely investigated. Methods Using the single B cell antibody cloning technology, we isolated single memory B cells, delineated the B cell receptor repertoire and conducted the pseudovirus-based assay for recovered neutralizing antibodies (NAb) screening. We analyzed the cryo-EM structures of top broadly NAbs (bnAbs) and evaluated their in vivo efficacy (golden Syrian hamster model). Findings By investigating the evolution of human B cell immunity, we discovered a new panel of bnAbs arising from vaccinees after Omicron BA.2/BA.5 breakthrough infections. Two lead bnAbs neutralized major Omicron subvariants including JN.1 and KP.2 with IC50 values less than 10 ng/mL, representing ultrapotent receptor binding domain (RBD)-specific class I bnAbs. They belonged to the IGHV3-53/3-66 clonotypes instead of evolving from the pre-existing vaccine-induced IGHV1-58/IGKV3-20 bnAb ZCB11. Despite sequence diversity, they targeted previously unrecognized, highly conserved conformational epitopes in the receptor binding motif (RBM) for ultrapotent ACE2 blockade. The lead bnAb ZCP3B4 not only protected the lungs of hamsters intranasally challenged with BA.5.2, BQ.1.1 and XBB.1.5 but also prevented their contact transmission. Interpretation Our findings demonstrated that class I bnAbs have evolved an ultrapotent mode of action protecting against highly transmissible and broad Omicron escape variants, and their epitopes are potential targets for novel bnAbs and vaccine development. Funding A full list of funding bodies that contributed to this study can be found in the Acknowledgements section.


Fig. 1: SARS-CoV-2 infection causes persistent abnormal foci of alveolar bronchiolization and fibrosis in hamster lung. a. Experimental layout: 6-8 weeks old hamsters were intranasally inoculated with 10 3 PFU SARS-CoV-2 wild-type strain HK-13 or equal volume of PBS as mock controls. Lung tissues were collected at 120dpi. Dysregulated regeneration in SARS-CoV-2 infected hamster lungs was observed in all hamsters (12/12), and illustrated by H&E-stained sections. Upper image shows lung condensation with blood vessel congestion and multiple abnormal foci, lower image shows abnormal foci of alveolar bronchiolization. b. Representative H&E images showing pulmonary consolidation, fibrosis and alveolar bronchiolization in SARS-CoV-2 infected hamster lungs at 120dpi compared to mock control (upper panel). Middle panel: SARSCoV-2 infected hamsters displayed whole lung condensation and multiple foci from proximal to distal lung regions. Bottom panel: Higher magnification images showing collapsed alveoli, pleurae thickening and alveolar bronchiolization. Black open arrows indicate pleurae thickening. Black triangles indicate alveolar epithelial cell hyperplasia. Scale bar = 500 μm, 200 μm, 100 μm, 50 μm, or 20 μm, respectively.
Fig. 9: Spatial transcriptomics of SARS-CoV-2 infected hamster lung. a. Six tissue regions representing abnormal (red circled), or normal (black circled) alveolar structures based on histological features shown in the H&E section. b. Ccdc39 gene expression in selected normal (left) and abnormal (right) tissue regions. c. Differential expression of genes expressed on ciliated cells, AT1 cells, AT2 cells, club cells and ADI cells in selected normal and abnormal tissue regions. d. Enriched pathways based on the up-regulated genes in abnormal tissue regions. e. Differential expression of genes associated with positive regulation of Notch signaling pathway in selected normal and abnormal tissue regions. f. Differential expression of genes related to cancer in selected normal and abnormal tissue regions.
Chronic lung inflammation and CK14+ basal cell proliferation induce persistent alveolar-bronchiolization in SARS-CoV-2-infected hamsters

September 2024

·

37 Reads

·

2 Citations

EBioMedicine

Background Post-acute sequalae of COVID-19 defines a wide range of ongoing symptoms and conditions long after SARS-CoV-2 infection including respiratory diseases. The histopathological changes in the lung and underlying mechanism remain elusive. Methods We investigated lung histopathological and transcriptional changes in SARS-CoV-2-infected male hamsters at 7, 14, 42, 84 and 120dpi, and compared with A (H1N1)pdm09 infection. Findings We demonstrated viral residue, inflammatory and fibrotic changes in lung after SARS-CoV-2 but not H1N1 infection. The most prominent histopathological lesion was multifocal alveolar-bronchiolization observed in every SARS-CoV-2 infected hamster (31/31), from 42dpi to 120dpi. Proliferating (Ki67+) CK14+ basal cells accumulated in alveoli adjacent to bronchioles at 7dpi, where they proliferated and differentiated into SCGB1A+ club cell or Tubulin+ ciliated cells forming alveolar-bronchiolization foci. Molecularly, Notch pathway significantly upregulated with intensive Notch3 and Hes1 protein expression in alveolar-bronchiolization foci at 42 and 120dpi, suggesting Notch signaling involving the persistence of alveolar-bronchiolization. This is further demonstrated by spatial transcriptomic analysis. Intriguingly, significant upregulation of some cell-growth promoting pathways and genes such as Tubb4b, Stxbp4, Grb14 and Mlf1 were spatially overlapping with bronchiolization lesion. Interpretation Incomplete resolution of SARS-CoV-2 infection in lung with viral residue, chronic inflammatory and fibrotic damage and alveolar-bronchiolization impaired respiratory function. Aberrant activation of CK14+ basal cells during tissue regeneration led to persistent alveolar-bronchiolization due to sustained Notch signaling. This study advances our understanding of respiratory PASC, sheds light on disease management and highlights the necessity for monitoring disease progression in people with respiratory PASC. Funding Funding is listed in the Acknowledgements section.


Figure 1. Characteristics of HEV-A4-P239 and HEV-C1-P241 peptides. (A) Genome map of hepatitis E virus (HEV) showing Open Reading Frame 2 (ORF2) region corresponding to HEV-A4-P239 and HEV-C1-P241 peptides. S: shell, M: middle, P: protruding domains of ORF2 are highlighted in pink. (B) Sodium dodecyl sulphate-polyacrylamide gel electrophoresis of HEV-A4-P239 and HEV-C1-P241 showing bands at 40-55 kDa. (C) Immunoblots based on HEV-A4-P239 and HEV-C1-P241 showing that Paslahepevirus balayani (bHEV) infected patient sera (lane 1: bHEV genotype 3, lane 2: bHEV genotype 4) reacted in HEV-A4-P239 immunoblot, but not HEV-C1-P241 immunoblot. Rocahepevirus ratti (rHEV) infected patient sera (lane 3) reacted in HEV-C1-P241 immunoblot, but not HEV-A4-P239 immunoblot. N: blank lane; P: anti-His antibody control. (D) Optical density (OD) values of bHEV-positive and rHEV-positive patient-derived IgG spiked into dog and cat serum pools using rHEV and bHEV enzymatic immunoassays. Purple lines represent rHEV-patient IgG spiked into cat serum pool. Orange lines represent rHEV-patient IgG spiked into dog serum pool. Pink lines represent bHEV-patient IgG spiked into cat serum pool. Brown lines represent rHEV-patient IgG spiked into dog serum pool. Black lines represent HEV-negative patient IgG spiked into HEV-negative dog serum pool. Green lines represent HEV-negative patient IgG spiked into HEV-negative cat serum pool. Red lines represent IgG purified from the HEV-negative dog serum pool. Gray lines represent IgG purified from the HEV-negative cat serum pool.
Figure 2. OD values of (A) dog and (B) cat serum samples tested in bHEV and rHEV EIAs. Blue dots and lines: sera testing positive only in bHEV EIA; red dots and lines: sera testing positive only in rHEV EIA; green dots and lines: sera testing positive in both EIAs. Horizontal black lines represent respective assay cutoffs (mean OD + 3 standard deviations of the group).
Figure 3. Optical Density (OD) values bHEV and rHEV EIA positive companion (A) dog and (B) cat sera in Wantai assay. Gray dots: bHEV and rHEV EIA negative control samples (n = 10); blue dots: sera testing positive only in bHEV EIA; red dots: sera testing positive only in rHEV EIA; green dots: sera testing positive in both EIAs. Horizontal lines represent cutoffs derived from mean OD + 3 standard deviations of 40 companion animal sera testing negative in both bHEV and rHEV EIAs.
Figure 4. HEV-C1-P241 immunoblot of rHEV EIA-positive companion animal sera samples. (A) Immunoblot of rHEV EIA-positive dog sera. (B) Immunoblot of rHEV EIA-positive cat sera. N: blank lane; P: anti-His antibody control. Blot assays were performed in a MiniProtean II Apparatus, which enables lane-by-lane separation of the immunoblot without cutting into strips.
Characterization of bHEV and rHEV EIA seropositive companion animals, Hong Kong.
Rat hepatitis E virus (Rocahepevirus ratti) exposure in cats and dogs, Hong Kong

March 2024

·

95 Reads

·

9 Citations

Hepatitis E virus (HEV) variants infecting humans belong to two species: Paslahepevirus balayani (bHEV) and Rocahepevirus ratti (rat hepatitis E virus; rHEV). R. ratti is a ubiquitous rodent pathogen that has recently been recognized to cause hepatitis in humans. Transmission routes of rHEV from rats to humans are currently unknown. In this study, we examined rHEV exposure in cats and dogs to determine if they are potential reservoirs of this emerging human pathogen. Virus-like particle-based IgG enzymatic immunoassays (EIAs) capable of differentiating rHEV & bHEV antibody profiles and rHEV-specific real-time RT-PCR assays were used for this purpose. The EIAs could detect bHEV and rHEV patient-derived IgG spiked in dog and cat sera. Sera from 751 companion dogs and 130 companion cats in Hong Kong were tested with these IgG enzymatic immunoassays (EIAs). Overall, 13/751 (1.7%) dogs and 5/130 (3.8%) cats were sero-reactive to HEV. 9/751 (1.2%) dogs and 2/130 (1.5%) cats tested positive for rHEV IgG, which was further confirmed by rHEV immunoblots. Most rHEV-seropositive animals were from areas in or adjacent to districts reporting human rHEV infection. Neither 881 companion animals nor 652 stray animals carried rHEV RNA in serum or rectal swabs. Therefore, we could not confirm a role for cats and dogs in transmitting rHEV to humans. Further work is required to understand the reasons for low-level seropositivity in these animals.


PMI-controlled mannose metabolism and glycosylation determines tissue tolerance and virus fitness

March 2024

·

225 Reads

·

7 Citations

Host survival depends on the elimination of virus and mitigation of tissue damage. Herein, we report the modulation of D-mannose flux rewires the virus-triggered immunometabolic response cascade and reduces tissue damage. Safe and inexpensive D-mannose can compete with glucose for the same transporter and hexokinase. Such competitions suppress glycolysis, reduce mitochondrial reactive-oxygen-species and succinate-mediated hypoxia-inducible factor-1α, and thus reduce virus-induced proinflammatory cytokine production. The combinatorial treatment by D-mannose and antiviral monotherapy exhibits in vivo synergy despite delayed antiviral treatment in mouse model of virus infections. Phosphomannose isomerase (PMI) knockout cells are viable, whereas addition of D-mannose to the PMI knockout cells blocks cell proliferation, indicating that PMI activity determines the beneficial effect of D-mannose. PMI inhibition suppress a panel of virus replication via affecting host and viral surface protein glycosylation. However, D-mannose does not suppress PMI activity or virus fitness. Taken together, PMI-centered therapeutic strategy clears virus infection while D-mannose treatment reprograms glycolysis for control of collateral damage.


An RNA-Scaffold Protein Subunit Vaccine for Nasal Immunization

September 2023

·

175 Reads

·

2 Citations

Developing recombinant proteins as nasal vaccines for inducing systemic and mucosal immunity against respiratory viruses is promising. However, additional adjuvants are required to overcome the low immunogenicity of protein antigens. Here, a self-adjuvanted protein-RNA ribonucleoprotein vaccine was developed and found to be an effective nasal vaccine in mice and the SARS-CoV-2 infection model. The vaccine consisted of spike RBD (as an antigen), nucleoprotein (as an adaptor), and ssRNA (as an adjuvant and RNA scaffold). This combination robustly induced mucosal IgA, neutralizing antibodies and activated multifunctional T-cells, while also providing sterilizing immunity against live virus challenge. In addition, high-resolution scRNA-seq analysis highlighted airway-resident immune cells profile during prime-boost immunization. The vaccine also possesses modularity (antigen/adaptor/RNA scaffold) and can be made to target other viruses. This protein-RNA ribonucleoprotein vaccine is a novel and promising approach for developing safe and potent nasal vaccines to combat respiratory virus infections.


Maximum likelihood phylogenetic tree of 11 local and 24 reference S. argenteus sequences based on total core genome SNPs. The tree was constructed by IQ-TREE 2 and was statistically supported by bootstrapping with 1000 replicates. Three MLST clusters (ST-2250, ST-1223, and ST-2854) were identified. The scale bar is in substitution per site. Bootstrap values >70 are shown at the branch nodes.
Distribution of virulence factors among the 11 local S. argenteus isolates. The coloured bars represent the presence of the factors in the genome of the isolate.
Laboratory identification results of the S. argenteus strains isolated in Hong Kong.
Phenotypic and genotypic antimicrobial resistance profiles of the 11 invasive S. argenteus isolates in Hong Kong.
Prevalence and Characteristics of Invasive Staphylococcus argenteus among Patients with Bacteremia in Hong Kong

September 2023

·

73 Reads

·

6 Citations

Staphylococcus argenteus is a novel Staphylococcus species derived from Staphylococcus aureus. Information on the prevalence and genetic characteristics of invasive S. argenteus in Asia is limited. In this study, 275 invasive S. aureus complex strains were retrieved from blood culture specimens in Hong Kong and re-analyzed using MALDI-TOF mass spectrometry and an in-house multiplex real-time PCR for S. argenteus. The prevalence of invasive S. argenteus in Hong Kong was found to be 4.0% (11/275). These strains were primarily susceptible to commonly used antibiotics, except penicillin. Whole-genome sequencing revealed the circulation of three S. argenteus genotypes (ST-2250, ST-1223, and ST-2854) in Hong Kong, with ST-2250 and ST-1223 being the predominant genotypes. The local ST-2250 and ST-1223 strains showed close phylogenetic relationships with isolates from mainland China. Antimicrobial-resistant genes (fosB, tet-38, mepA, blaI, blaZ) could be found in nearly all local S. argenteus strains. The ST-1223 and ST-2250 genotypes carried multiple staphylococcal enterotoxin genes that could cause food poisoning and toxic shock syndrome. The CRISPR/Cas locus was observed only in the ST-2250 strains. This study provides the first report on the molecular epidemiology of invasive S. argenteus in Hong Kong, and further analysis is needed to understand its transmission reservoir.


Broad-spectrum humanized monoclonal neutralizing antibody against SARS-CoV-2 variants, including the Omicron variant

August 2023

·

66 Reads

·

2 Citations

Introduction Therapeutic monoclonal antibodies (mAbs) against the SARS-CoV-2 spike protein have been shown to improve the outcome of severe COVID-19 patients in clinical trials. However, novel variants with spike protein mutations can render many currently available mAbs ineffective. Methods We produced mAbs by using hybridoma cells that generated from mice immunized with spike protein trimer and receptor binding domain (RBD). The panel of mAbs were screened for binding and neutralizing activity against different SARS-CoV-2 variants. The in vivo effectiveness of WKS13 was evaluated in a hamster model. Results Out of 960 clones, we identified 18 mAbs that could bind spike protein. Ten of the mAbs could attach to RBD, among which five had neutralizing activity against the ancestral strain and could block the binding between the spike protein and human ACE2. One of these mAbs, WKS13, had broad neutralizing activity against all Variants of Concern (VOCs), including the Omicron variant. Both murine or humanized versions of WKS13 could reduce the lung viral load in hamsters infected with the Delta variant. Conclusions Our data showed that broad-spectrum high potency mAbs can be produced from immunized mice, which can be used in humans after humanization of the Fc region. Our method represents a versatile and rapid strategy for generating therapeutic mAbs for upcoming novel variants.


The viral fitness and intrinsic pathogenicity of dominant SARS-CoV-2 Omicron sublineages BA.1, BA.2, and BA.5

August 2023

·

126 Reads

·

34 Citations

EBioMedicine

Background: Among the Omicron sublineages that have emerged, BA.1, BA.2, BA.5, and their related sublineages have resulted in the largest number of infections. While recent studies demonstrated that all Omicron sublineages robustly escape neutralizing antibody response, it remains unclear on whether these Omicron sublineages share any pattern of evolutionary trajectory on their replication efficiency and intrinsic pathogenicity along the respiratory tract. Methods: We compared the virological features, replication capacity of dominant Omicron sublineages BA.1, BA.2 and BA.5 in the human nasal epithelium, and characterized their pathogenicity in K18-hACE2, A129, young C57BL/6, and aged C57BL/6 mice. Findings: We found that BA.5 replicated most robustly, followed by BA.2 and BA.1, in the differentiated human nasal epithelium. Consistently, BA.5 infection resulted in higher viral gene copies, infectious viral titres and more abundant viral antigen expression in the nasal turbinates of the infected K18-hACE2 transgenic mice. In contrast, the Omicron sublineages are continuously attenuated in lungs of infected K18-hACE2 and C57BL/6 mice, leading to decreased pathogenicity. Nevertheless, lung manifestations remain severe in Omicron sublineages-infected A129 and aged C57BL/6 mice. Interpretation: Our results suggested that the Omicron sublineages might be gaining intrinsic replication fitness in the upper respiratory tract, therefore highlighting the importance of global surveillance of the emergence of hyper-transmissive Omicron sublineages. On the contrary, replication and intrinsic pathogenicity of Omicron is suggested to be further attenuated in the lower respiratory tract. Effective vaccination and other precautions should be in place to prevent severe infections in the immunocompromised populations at risk. Funding: A full list of funding bodies that contributed to this study can be found in the Acknowledgements section.


Citations (90)


... The dominantly circulated SARS-CoV-2 Omicron variants include descendants of the recombinant XBB lineage, such as XBB.1.16, EG.5.1, and HK.3, as well as descendants of BA.2, including JN.1 and KP.3.1.1 [13,14]. In particular, JN.1 S contains a concerning accumulation of more than 30 mutations compared to that of BA.2 and has spread rapidly worldwide [13,15]. ...

Reference:

Novel Trispecific Neutralizing Antibodies With Enhanced Potency and Breadth Against Pan‐Sarbecoviruses
Lineage-specific pathogenicity, immune evasion, and virological features of SARS-CoV-2 BA.2.86/JN.1 and EG.5.1/HK.3

... In accordance with our results Frere et al. showed that lungs from SARS-CoV-2-infected hamsters displayed localized zones of hypercellularity that stained positively for both neutrophil and macrophage populations at 31-dpi, when compared to lungs from non-infected animals, also pointing to the involvement of these cells in long term responses (Frere et al. 2022). Previous studies have also detected viral components in lung tissue of SARS-CoV-2 infected hamsters at later time points (Castellan et al. 2023, Li et al. 2024. The use of immunogold TEM for N-protein revealed the existence of this viral protein in both the nucleus and the cytoplasm of PIMs thus confirming recruited PIMs as SARS-CoV-2-containing cells. ...

Chronic lung inflammation and CK14+ basal cell proliferation induce persistent alveolar-bronchiolization in SARS-CoV-2-infected hamsters

EBioMedicine

... Notably, in the study, we determined the HEV rat IgG seroprevalence was 18.7% in men and 24.1% in women in set 1. It might be explained as a certain transmission route for rat HEV, such as house cleaning, contact with domestic animals, and subsequent exposure to potentially contaminated objects, which might be more common for women in daily housework; or potential sampling bias. Prospective cohort studies should focus on daily contact objects and foods potentially contaminated by rats, as well as frequent contact with domestic animal such as swine and dogs [72][73][74] . Interestingly, none of the patients in sets 3 and 4 with ALT abnormality tested positive for rat HEV RNA, despite the widespread circulation of rat HEV suggested by the seroprevalence results. ...

Rat hepatitis E virus (Rocahepevirus ratti) exposure in cats and dogs, Hong Kong

... The primary intracellular fate of mannose is linked to the expression of two key enzymes namely phosphomannomutase (PMM2) and phosphomannose isomerase (PMI), fulfilling respective roles in protein glycosylation or glycolytic catabolism [27,28]. Recent studies report mannose mediated protection of the intestinal barrier lessening the effects of ulcerative colitis, to immune-metabolic reprogramming limiting the infective potential of common viruses, effects linked to the glycosylation role of mannose, with little direct impact on proliferation [29,30]. Our data indicate a differential response, more likely common to tumour models. ...

PMI-controlled mannose metabolism and glycosylation determines tissue tolerance and virus fitness

... Mucosal vaccine has been advocated as a strategy to improve mucosal antibody response, especially for mucosal IgA [28][29][30]. Mice receiving mucosal vaccine have higher mucosal IgA levels than those with receiving intramuscular vaccine [31]. COVID-19 intranasal vaccines have been approved in China, Russia, India and Iran [32]. ...

An RNA-Scaffold Protein Subunit Vaccine for Nasal Immunization

... Although the nearly universal prevalence of the four haemolysin genes has been described in epidemiological and genomic studies to date [ 6 , 13 , 32 ], the absence of hlg -A has been shown in some strains despite the presence of hlg -B and -C [ 13 ]. In addition, hlb was absent in most ST2250 isolates from Indonesia [ 33 ], and in all ST2250, ST1223, and ST2854 isolates from Hong Kong [ 9 ]. In contrast, hld with a sequence almost identical to that of S. aureus was detected in all isolates in the present study, and such a wide distribution of hld has also been described previously [ 9 , 13 , 32 , 33 ]. ...

Prevalence and Characteristics of Invasive Staphylococcus argenteus among Patients with Bacteremia in Hong Kong

... Regarding clinical profiles, changes were observed in the number of symptoms, mainly in Santa Cruz Cabrália in 2022 and possibly because of the predominance of the Omicron variant in the region during this period. Omicron infection is less involved with the lower respiratory tract and has a lower probability of hospitalization 36,37,38 . Moreover, the viral loads of Omicron subvariants, which are significant factors associated with severe disease outcomes, are generally lower in the lungs than in the nasal mucosa 39,40 . ...

The viral fitness and intrinsic pathogenicity of dominant SARS-CoV-2 Omicron sublineages BA.1, BA.2, and BA.5

EBioMedicine

... According to Wen et al. [119], WKS13, a monoclonal antibody produced by the researchers using hybridoma cells derived from mice immunized with spike protein trimer and RBD, exhibited broad neutralizing activity against all VOCs, including the Omicron variant. Out of the 960 clones examined, 18 mAbs showed binding affinity to the S protein, with 5 of them demonstrating the ability to neutralize SARS-CoV-2. ...

Broad-spectrum humanized monoclonal neutralizing antibody against SARS-CoV-2 variants, including the Omicron variant

... Seven studies, five RCTs, and two non-randomised studies, have been published regarding OT for COVID-19 associated olfactory dysfunction (Table 1) [22][23][24][25][26][27][28]. Patients received OT with either the classical four scents or a modified training with eight scents. ...

A Pilot Study of Short-Course Oral Vitamin A and Aerosolised Diffuser Olfactory Training for the Treatment of Smell Loss in Long COVID

... Given the unknown accuracy of the established DARE method, the previously reported immunoblot method was applied to validate in immunized BALB/c samples 44,53 . None (0 of 6) of the samples from PBS immunized BALB/c exhibited a visible band at parallel b-4 p239 and rat p239 immunoblots (Fig. 3A), demonstrating the specificity of the DARE method. ...

An immunoassay system to investigate epidemiology of Rocahepevirus ratti (rat hepatitis E virus) infection in humans

JHEP Reports