Your browser doesn't support javascript.
Montrer: 20 | 50 | 100
Résultats 1 - 7 de 7
Filtre
1.
Emerg Microbes Infect ; : 1-52, 2022 Nov 10.
Article Dans Anglais | MEDLINE | ID: covidwho-2242917

Résumé

Increasing spread by SARS-CoV-2 Omicron variants challenges existing vaccines and broadly reactive neutralizing antibodies (bNAbs) against COVID-19. Here we determine the diversity, potency, breadth and structural insights of bNAbs derived from memory B cells of BNT162b2-vaccinee after homogeneous Omicron BA.1 breakthrough infection. The infection activates diverse memory B cell clonotypes for generating potent class I/II or III bNAbs with new epitopes mapped to receptor-binding domain (RBD). The top eight bNAbs neutralize wildtype and BA.1 potently but display divergent IgH/IgL sequences and neuralization profiles against other variants of concern (VOCs). Two of them (P2D9 and P3E6) belonging to class III NAbs display comparable potency against BA.4/BA.5, although structural analysis reveals distinct modes of action. P3E6 neutralizes all variants tested through a unique bivalent interaction with two RBDs. Our findings provide new insights into hybrid immunity on BNT162b2-induced diverse memory B cells in response to Omicron breakthrough infection for generating diverse bNAbs with distinct structural basis.

2.
Clin Transl Med ; 12(9): e1025, 2022 09.
Article Dans Anglais | MEDLINE | ID: covidwho-2027333

Résumé

BACKGROUND: Patients with COVID-19 display a broad spectrum of manifestations from asymptomatic to life-threatening disease with dysregulated immune responses. Mechanisms underlying the detrimental immune responses and disease severity remain elusive. METHODS: We investigated a total of 137 APs infected with SARS-CoV-2. Patients were divided into mild and severe patient groups based on their requirement of oxygen supplementation. All blood samples from APs were collected within three weeks after symptom onset. Freshly isolated PBMCs were investigated for B cell subsets, their homing potential, activation state, mitochondrial functionality and proliferative response. Plasma samples were tested for cytokine concentration, and titer of Nabs, RBD-, S1-, SSA/Ro- and dsDNA-specific IgG. RESULTS: While critically ill patients displayed predominantly extrafollicular B cell activation with elevated inflammation, mild patients counteracted the disease through the timely induction of mitochondrial dysfunction in B cells within the first week post symptom onset. Rapidly increased mitochondrial dysfunction, which was caused by infection-induced excessive intracellular calcium accumulation, suppressed excessive extrafollicular responses, leading to increased neutralizing potency index and decreased inflammatory cytokine production. Patients who received prior COVID-19 vaccines before infection displayed significantly decreased extrafollicular B cell responses and mild disease. CONCLUSION: Our results reveal an immune mechanism that controls SARS-CoV-2-induced detrimental B cell responses and COVID-19 severity, which may have implications for viral pathogenesis, therapeutic interventions and vaccine development.


Sujets)
COVID-19 , Vaccins antiviraux , Lymphocytes B , Vaccins contre la COVID-19 , Cytokines , Humains , Mitochondries , SARS-CoV-2 , Indice de gravité de la maladie , Vaccins antiviraux/pharmacologie
3.
Cell Discov ; 8(1): 57, 2022 Jun 17.
Article Dans Anglais | MEDLINE | ID: covidwho-1967594

Résumé

The airways and alveoli of the human respiratory tract are lined by two distinct types of epithelium, which are the primary targets of respiratory viruses. We previously established long-term expanding human lung epithelial organoids from lung tissues and developed a 'proximal' differentiation protocol to generate mucociliary airway organoids. However, a respiratory organoid system with bipotential of the airway and alveolar differentiation remains elusive. Here we defined a 'distal' differentiation approach to generate alveolar organoids from the same source for the derivation of airway organoids. The alveolar organoids consisting of type I and type II alveolar epithelial cells (AT1 and AT2, respectively) functionally simulate the alveolar epithelium. AT2 cells maintained in lung organoids serve as progenitor cells from which alveolar organoids derive. Moreover, alveolar organoids sustain a productive SARS-CoV-2 infection, albeit a lower replicative fitness was observed compared to that in airway organoids. We further optimized 2-dimensional (2D) airway organoids. Upon differentiation under a slightly acidic pH, the 2D airway organoids exhibit enhanced viral replication, representing an optimal in vitro correlate of respiratory epithelium for modeling the high infectivity of SARS-CoV-2. Notably, the higher infectivity and replicative fitness of the Omicron variant than an ancestral strain were accurately recapitulated in these optimized airway organoids. In conclusion, we have established a bipotential organoid culture system able to reproducibly expand the entire human respiratory epithelium in vitro for modeling respiratory diseases, including COVID-19.

4.
Nat Commun ; 13(1): 3589, 2022 06 23.
Article Dans Anglais | MEDLINE | ID: covidwho-1900488

Résumé

The strikingly high transmissibility and antibody evasion of SARS-CoV-2 Omicron variants have posed great challenges to the efficacy of current vaccines and antibody immunotherapy. Here, we screen 34 BNT162b2-vaccinees and isolate a public broadly neutralizing antibody ZCB11 derived from the IGHV1-58 family. ZCB11 targets viral receptor-binding domain specifically and neutralizes all SARS-CoV-2 variants of concern, especially with great potency against authentic Omicron and Delta variants. Pseudovirus-based mapping of 57 naturally occurred spike mutations or deletions reveals that S371L results in 11-fold neutralization resistance, but it is rescued by compensating mutations in Omicron variants. Cryo-EM analysis demonstrates that ZCB11 heavy chain predominantly interacts with Omicron spike trimer with receptor-binding domain in up conformation blocking ACE2 binding. In addition, prophylactic or therapeutic ZCB11 administration protects lung infection against Omicron viral challenge in golden Syrian hamsters. These results suggest that vaccine-induced ZCB11 is a promising broadly neutralizing antibody for biomedical interventions against pandemic SARS-CoV-2.


Sujets)
Anticorps antiviraux , Anticorps neutralisants à large spectre , COVID-19 , Animaux , Anticorps antiviraux/immunologie , Vaccin BNT162 , Anticorps neutralisants à large spectre/immunologie , COVID-19/prévention et contrôle , Cricetinae , Humains , Mesocricetus , SARS-CoV-2/génétique , Glycoprotéine de spicule des coronavirus/génétique
5.
EBioMedicine ; 75: 103762, 2022 Jan.
Article Dans Anglais | MEDLINE | ID: covidwho-1587929

Résumé

BACKGROUND: Vaccines in emergency use are efficacious against COVID-19, yet vaccine-induced prevention against nasal SARS-CoV-2 infection remains suboptimal. METHODS: Since mucosal immunity is critical for nasal prevention, we investigated the efficacy of an intramuscular PD1-based receptor-binding domain (RBD) DNA vaccine (PD1-RBD-DNA) and intranasal live attenuated influenza-based vaccines (LAIV-CA4-RBD and LAIV-HK68-RBD) against SARS-CoV-2. FINDINGS: Substantially higher systemic and mucosal immune responses, including bronchoalveolar lavage IgA/IgG and lung polyfunctional memory CD8 T cells, were induced by the heterologous PD1-RBD-DNA/LAIV-HK68-RBD as compared with other regimens. When vaccinated animals were challenged at the memory phase, prevention of robust SARS-CoV-2 infection in nasal turbinate was achieved primarily by the heterologous regimen besides consistent protection in lungs. The regimen-induced antibodies cross-neutralized variants of concerns. Furthermore, LAIV-CA4-RBD could boost the BioNTech vaccine for improved mucosal immunity. INTERPRETATION: Our results demonstrated that intranasal influenza-based boost vaccination induces mucosal and systemic immunity for effective SARS-CoV-2 prevention in both upper and lower respiratory systems. FUNDING: This study was supported by the Research Grants Council Collaborative Research Fund, General Research Fund and Health and Medical Research Fund in Hong Kong; Outbreak Response to Novel Coronavirus (COVID-19) by the Coalition for Epidemic Preparedness Innovations; Shenzhen Science and Technology Program and matching fund from Shenzhen Immuno Cure BioTech Limited; the Health@InnoHK, Innovation and Technology Commission of Hong Kong; National Program on Key Research Project of China; donations from the Friends of Hope Education Fund; the Theme-Based Research Scheme.


Sujets)
Vaccins contre la COVID-19 , COVID-19/prévention et contrôle , Rappel de vaccin , Vaccins antigrippaux , SARS-CoV-2 , Vaccins à ADN , Administration par voie nasale , Animaux , COVID-19/génétique , COVID-19/immunologie , Vaccins contre la COVID-19/génétique , Vaccins contre la COVID-19/immunologie , Chlorocebus aethiops , Modèles animaux de maladie humaine , Chiens , Femelle , Cellules HEK293 , Humains , Immunité muqueuse , Vaccins antigrippaux/génétique , Vaccins antigrippaux/immunologie , Cellules rénales canines Madin-Darby , Mâle , Souris , Souris de lignée BALB C , Souris transgéniques , SARS-CoV-2/génétique , SARS-CoV-2/immunologie , Vaccins atténués/génétique , Vaccins atténués/immunologie , Vaccins à ADN/génétique , Vaccins à ADN/immunologie , Cellules Vero
6.
Cell Host Microbe ; 29(4): 551-563.e5, 2021 04 14.
Article Dans Anglais | MEDLINE | ID: covidwho-1101147

Résumé

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is characterized by a burst in the upper respiratory portal for high transmissibility. To determine human neutralizing antibodies (HuNAbs) for entry protection, we tested three potent HuNAbs (IC50 range, 0.0007-0.35 µg/mL) against live SARS-CoV-2 infection in the golden Syrian hamster model. These HuNAbs inhibit SARS-CoV-2 infection by competing with human angiotensin converting enzyme-2 for binding to the viral receptor binding domain (RBD). Prophylactic intraperitoneal or intranasal injection of individual HuNAb or DNA vaccination significantly reduces infection in the lungs but not in the nasal turbinates of hamsters intranasally challenged with SARS-CoV-2. Although postchallenge HuNAb therapy suppresses viral loads and lung damage, robust infection is observed in nasal turbinates treated within 1-3 days. Our findings demonstrate that systemic HuNAb suppresses SARS-CoV-2 replication and injury in lungs; however, robust viral infection in nasal turbinate may outcompete the antibody with significant implications to subprotection, reinfection, and vaccine.


Sujets)
Anticorps neutralisants/usage thérapeutique , Anticorps antiviraux/usage thérapeutique , COVID-19/thérapie , SARS-CoV-2/immunologie , Cornets/virologie , Angiotensin-converting enzyme 2/physiologie , Animaux , Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , COVID-19/immunologie , COVID-19/virologie , Cricetinae , Femelle , Cellules HEK293 , Humains , Mâle , Mesocricetus , Charge virale
7.
Immunity ; 53(4): 864-877.e5, 2020 10 13.
Article Dans Anglais | MEDLINE | ID: covidwho-693493

Résumé

The SARS-CoV-2 pandemic has resulted in millions of infections, yet the role of host immune responses in early COVID-19 pathogenesis remains unclear. By investigating 17 acute and 24 convalescent patients, we found that acute SARS-CoV-2 infection resulted in broad immune cell reduction including T, natural killer, monocyte, and dendritic cells (DCs). DCs were significantly reduced with functional impairment, and ratios of conventional DCs to plasmacytoid DCs were increased among acute severe patients. Besides lymphocytopenia, although neutralizing antibodies were rapidly and abundantly generated in patients, there were delayed receptor binding domain (RBD)- and nucleocapsid protein (NP)-specific T cell responses during the first 3 weeks after symptoms onset. Moreover, acute RBD- and NP-specific T cell responses included relatively more CD4 T cells than CD8 T cells. Our findings provided evidence that impaired DCs, together with timely inverted strong antibody but weak CD8 T cell responses, could contribute to acute COVID-19 pathogenesis and have implications for vaccine development.


Sujets)
Betacoronavirus/pathogénicité , Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Infections à coronavirus/immunologie , Cellules dendritiques/immunologie , Diabète/immunologie , Hypertension artérielle/immunologie , Pneumopathie virale/immunologie , Adulte , Sujet âgé , Anticorps neutralisants/biosynthèse , Anticorps antiviraux/biosynthèse , Betacoronavirus/immunologie , Lymphocytes T CD4+/anatomopathologie , Lymphocytes T CD4+/virologie , Lymphocytes T CD8+/anatomopathologie , Lymphocytes T CD8+/virologie , COVID-19 , Convalescence , Infections à coronavirus/complications , Infections à coronavirus/diagnostic , Infections à coronavirus/virologie , Cellules dendritiques/anatomopathologie , Cellules dendritiques/virologie , Complications du diabète , Diabète/diagnostic , Diabète/virologie , Évolution de la maladie , Femelle , Humains , Hypertension artérielle/complications , Hypertension artérielle/diagnostic , Hypertension artérielle/virologie , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/anatomopathologie , Cellules tueuses naturelles/virologie , Activation des lymphocytes , Numération des lymphocytes , Mâle , Adulte d'âge moyen , Monocytes/immunologie , Monocytes/anatomopathologie , Monocytes/virologie , Pandémies , Pneumopathie virale/complications , Pneumopathie virale/diagnostic , Pneumopathie virale/virologie , SARS-CoV-2 , Indice de gravité de la maladie
SÉLECTION CITATIONS
Détails de la recherche