Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
Nat Commun ; 15(1): 3449, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38664384

RESUMEN

In 2017, a novel influenza A virus (IAV) was isolated from an Egyptian fruit bat. In contrast to other bat influenza viruses, the virus was related to avian A(H9N2) viruses and was probably the result of a bird-to-bat transmission event. To determine the cross-species spill-over potential, we biologically characterize features of A/bat/Egypt/381OP/2017(H9N2). The virus has a pH inactivation profile and neuraminidase activity similar to those of human-adapted IAVs. Despite the virus having an avian virus-like preference for α2,3 sialic acid receptors, it is unable to replicate in male mallard ducks; however, it readily infects ex-vivo human respiratory cell cultures and replicates in the lungs of female mice. A/bat/Egypt/381OP/2017 replicates in the upper respiratory tract of experimentally-infected male ferrets featuring direct-contact and airborne transmission. These data suggest that the bat A(H9N2) virus has features associated with increased risk to humans without a shift to a preference for α2,6 sialic acid receptors.


Asunto(s)
Quirópteros , Patos , Hurones , Subtipo H9N2 del Virus de la Influenza A , Infecciones por Orthomyxoviridae , Receptores de Superficie Celular , Animales , Quirópteros/virología , Humanos , Hurones/virología , Femenino , Masculino , Subtipo H9N2 del Virus de la Influenza A/fisiología , Subtipo H9N2 del Virus de la Influenza A/patogenicidad , Subtipo H9N2 del Virus de la Influenza A/aislamiento & purificación , Infecciones por Orthomyxoviridae/virología , Infecciones por Orthomyxoviridae/transmisión , Ratones , Patos/virología , Replicación Viral , Gripe Humana/virología , Gripe Humana/transmisión , Pulmón/virología , Gripe Aviar/virología , Gripe Aviar/transmisión , Neuraminidasa/metabolismo
3.
J Virol ; 97(12): e0136923, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-38038429

RESUMEN

IMPORTANCE: Viral host adaptation plays an important role in inter-species transmission of coronaviruses and influenza viruses. Multiple human-adaptive mutations have been identified in influenza viruses but not so far in MERS-CoV that circulates widely in dromedary camels in the Arabian Peninsula leading to zoonotic transmission. Here, we analyzed clade B MERS-CoV sequences and identified an amino acid substitution L232F in nsp6 that repeatedly occurs in human MERS-CoV. Using a loss-of-function reverse genetics approach, we found the nsp6 L232F conferred increased viral replication competence in vitro, in cultures of the upper human respiratory tract ex vivo, and in lungs of mice infected in vivo. Our results showed that nsp6 L232F may be an adaptive mutation associated with zoonotic transmission of MERS-CoV. This study highlighted the capacity of MERS-CoV to adapt to transmission to humans and also the need for continued surveillance of MERS-CoV in camels.


Asunto(s)
Infecciones por Coronavirus , Coronavirus del Síndrome Respiratorio de Oriente Medio , Proteínas no Estructurales Virales , Animales , Humanos , Ratones , Sustitución de Aminoácidos , Camelus , Infecciones por Coronavirus/virología , Coronavirus del Síndrome Respiratorio de Oriente Medio/genética , Mutación , Proteínas no Estructurales Virales/genética
4.
Dalton Trans ; 53(1): 346-353, 2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38050668

RESUMEN

The report concerns expansion of the previously developed M-[O,N,C] [pyridine-2-phenolate-6-(σ-aryl)] catalyst system into rigid, coplanar bimetallic assemblies, which afford metal-metal distances that are predetermined yet amenable for cooperativity, as well as locked-in "syn" orientation of binding sites that offer the same direction of access for substrates. The binuclear complexes are generated in a regioselective manner to yield para hydrogen atoms (not ortho) at the central µ-aryl moiety, and have been characterised by multinuclear NMR spectroscopy. The "anti" (showing opposite directions of access) and mononuclear analogues have also been prepared for comparison purposes. Six syn-type bimetallic derivatives of Ti, Zr and Hf have been characterised by X-ray crystallography, to reveal metal-metal separations of 6.3-6.7 Å. For ethylene and ethylene/1-octene polymerisation reactions in conjunction with trityl borate, the syn-Ti2 catalysts display superior efficiencies and produced polymers with higher Mw values than for the anti and mono-Ti congeners, thus indicating the possibility of favourable enchainment interactions and cooperative reactivity.

5.
Artículo en Inglés | MEDLINE | ID: mdl-37623181

RESUMEN

BACKGROUND: The COVID-19 pandemic from 2019 to 2022 devastated many aspects of life and the economy, with the commercial aviation industry being no exception. One of the major concerns during the pandemic was the degree to which the internal aircraft environment contributed to virus transmission between humans and, in particular, the stability of SARS-CoV-2 on contact surfaces in the aircraft cabin interior. METHOD: In this study, the stability of various major strains of SARS-CoV-2 on interior aircraft surfaces was evaluated using the TCID50 assessment. RESULTS: In contrast to terrestrial materials, SARS-CoV-2 was naturally less stable on common contact points in the aircraft interior, and, over a 4 h time period, there was a 90% reduction in culturable virus. Antiviral and surface coatings were extremely effective at mitigating the persistence of the virus on surfaces; however, their benefit was diminished by regular cleaning and were ineffective after 56 days of regular use and cleaning. Finally, successive strains of SARS-CoV-2 have not evolved to be more resilient to survival on aircraft surfaces. CONCLUSIONS: We conclude that the mitigation strategies for SARS-CoV-2 on interior aircraft surfaces are more than sufficient, and epidemiological evidence over the past three years has not found that surface spread is a major route of transmission.


Asunto(s)
Aviación , COVID-19 , Humanos , SARS-CoV-2 , COVID-19/epidemiología , COVID-19/prevención & control , Pandemias , Aeronaves
6.
Emerg Infect Dis ; 29(6): 1210-1214, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37095078

RESUMEN

Human infection with avian influenza A(H3N8) virus is uncommon but can lead to acute respiratory distress syndrome. In explant cultures of the human bronchus and lung, novel H3N8 virus showed limited replication efficiency in bronchial and lung tissue but had a higher replication than avian H3N8 virus in lung tissue.


Asunto(s)
Subtipo H3N8 del Virus de la Influenza A , Gripe Humana , Infecciones por Orthomyxoviridae , Animales , Humanos , Pulmón/diagnóstico por imagen , Bronquios , Replicación Viral
7.
Inorg Chem ; 61(48): 19543-19551, 2022 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-36413484

RESUMEN

A series of conformationally rigid (Zn-salphen)2 complexes with a planar bridging component (xanthene or dibenzofuran) are described. Conformational changes for these assemblies are essentially limited to the axial rotation of the Zn-salphen moieties; however, such geometric constraints crucially permit the subtle tuning of the intermetallic separation and geometry to potentially enhance catalytic activity (and cooperative effects). The complexes have been investigated as catalysts in conjunction with nBu4NI for the coupling of CO2 with epoxides. Selected dibenzofuran derivatives are significantly more active for the production of cyclic carbonate than their mononuclear analogues under identical conditions and concentrations of Zn sites. High initial turnover frequencies (up to 29 000 h-1; 14 500 h-1 per Zn, using 10 bar of CO2 at 95 °C) and excellent efficiencies under mild conditions (1 bar of CO2 at 55 °C) have been achieved. Kinetic studies using in situ (ReactIR) spectroscopy and density functional theory calculations have been performed, which reveal the existence of an intramolecular rate component and a preference for the cooperative pathway as well as transition states that depict the Zn sites operating in tandem. Taken together, these results provide strong evidence of cooperative reactivity in these Zn2 catalysts.

8.
EBioMedicine ; 83: 104232, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35988466

RESUMEN

BACKGROUND: The Omicron BA.2 sublineage has replaced BA.1 worldwide and has comparable levels of immune evasion to BA.1. These observations suggest that the increased transmissibility of BA.2 cannot be explained by the antibody evasion. METHODS: Here, we characterized the replication competence and respiratory tissue tropism of three Omicron variants (BA.1, BA.1.1, BA.2), and compared these with the wild-type virus and Delta variant, in human nasal, bronchial and lung tissues cultured ex vivo. FINDINGS: BA.2 replicated more efficiently in nasal and bronchial tissues at 33°C than wild-type, Delta and BA.1. Both BA.2 and BA.1 had higher replication competence than wild-type and Delta viruses in bronchial tissues at 37°C. BA.1, BA.1.1 and BA.2 replicated at a lower level in lung parenchymal tissues compared to wild-type and Delta viruses. INTERPRETATION: Higher replication competence of Omicron BA.2 in the human upper airway at 33°C than BA.1 may be one of the reasons to explain the current advantage of BA.2 over BA.1. A lower replication level of the tested Omicron variants in human lung tissues is in line with the clinical manifestations of decreased disease severity of patients infected with the Omicron strains compared with other ancestral strains. FUNDING: This work was supported by US National Institute of Allergy and Infectious Diseases and the Theme-Based Research Scheme under University Grants Committee of Hong Kong Special Administrative Region, China.


Asunto(s)
COVID-19 , SARS-CoV-2 , Bronquios , Humanos , SARS-CoV-2/genética , Tropismo Viral , Replicación Viral
9.
Nature ; 603(7902): 715-720, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35104836

RESUMEN

The emergence of SARS-CoV-2 variants of concern with progressively increased transmissibility between humans is a threat to global public health. The Omicron variant of SARS-CoV-2 also evades immunity from natural infection or vaccines1, but it is unclear whether its exceptional transmissibility is due to immune evasion or intrinsic virological properties. Here we compared the replication competence and cellular tropism of the wild-type virus and the D614G, Alpha (B.1.1.7), Beta (B.1.351), Delta (B.1.617.2) and Omicron (B.1.1.529) variants in ex vivo explant cultures of human bronchi and lungs. We also evaluated the dependence on TMPRSS2 and cathepsins for infection. We show that Omicron replicates faster than all other SARS-CoV-2 variants studied in the bronchi but less efficiently in the lung parenchyma. All variants of concern have similar cellular tropism compared to the wild type. Omicron is more dependent on cathepsins than the other variants of concern tested, suggesting that the Omicron variant enters cells through a different route compared with the other variants. The lower replication competence of Omicron in the human lungs may explain the reduced severity of Omicron that is now being reported in epidemiological studies, although determinants of severity are multifactorial. These findings provide important biological correlates to previous epidemiological observations.


Asunto(s)
Bronquios/virología , Pulmón/virología , SARS-CoV-2/crecimiento & desarrollo , Tropismo Viral , Replicación Viral , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , COVID-19/epidemiología , COVID-19/transmisión , COVID-19/virología , Catepsinas/metabolismo , Chlorocebus aethiops , Endocitosis , Humanos , Técnicas In Vitro , SARS-CoV-2/inmunología , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Técnicas de Cultivo de Tejidos , Células Vero
10.
Angew Chem Int Ed Engl ; 61(9): e202115712, 2022 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-34968004

RESUMEN

The study of metallopolymers with controllable helical sense remains in its infancy. We report arabinose-functionalized (Zn-salphen)-based conjugated polymers that display mirror-image circular dichroism spectra for L- and D-sugar sidechains respectively, signifying ordered (helical) coiling of the polymer backbone with opposite screw-sense preferences. The observation of different spectroscopic behavior and Cotton effects for a variety of solvents (in a reversible manner) and temperatures, ascribed to changes in the extent of intrachain (Zn⋅⋅⋅O(salphen) and π-stacking) interactions between Zn-salphen moieties, thus indicate the flexible, responsive and dynamic nature of the folded helical conformation in these systems. An application study signifying that activity can be governed by the structure and helical sense of the polymer is described.

11.
Nat Microbiol ; 6(11): 1455-1465, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34702977

RESUMEN

Understanding the evolutionary adaptations that enable avian influenza viruses to transmit in mammalian hosts could allow better detection of zoonotic viruses with pandemic potential. We applied ancestral sequence reconstruction to gain viruses representing different adaptive stages of the European avian-like (EA) H1N1 swine influenza virus as it transitioned from avian to swine hosts since 1979. Ancestral viruses representing the avian-like precursor virus and EA swine influenza viruses from 1979-1983, 1984-1987 and 1988-1992 were reconstructed and characterized. Glycan-binding analyses showed stepwise changes in the haemagglutinin receptor-binding specificity of the EA swine influenza viruses-that is, from recognition of both α2,3- and α2,6-linked sialosides to recognition of α2,6-linked sialosides only; however, efficient transmission in piglets was enabled by adaptive changes in the viral polymerase protein and nucleoprotein, which have been fixed since 1983. PB1-Q621R and NP-R351K increased viral replication and transmission in piglets when introduced into the 1979-1983 ancestral virus that lacked efficient transmissibility. The stepwise adaptation of an avian influenza virus to a mammalian host suggests that there may be opportunities to intervene and prevent interspecies jumps through strategic coordination of surveillance and risk assessment activities.


Asunto(s)
Adaptación Fisiológica , Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Aviar/virología , Infecciones por Orthomyxoviridae/veterinaria , Enfermedades de los Porcinos/virología , Animales , Aves , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Subtipo H1N1 del Virus de la Influenza A/clasificación , Subtipo H1N1 del Virus de la Influenza A/genética , Gripe Aviar/transmisión , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/transmisión , Infecciones por Orthomyxoviridae/virología , Filogenia , Polisacáridos/química , Polisacáridos/metabolismo , Receptores Virales/química , Receptores Virales/metabolismo , Porcinos , Enfermedades de los Porcinos/metabolismo , Enfermedades de los Porcinos/transmisión , Replicación Viral
12.
Emerg Infect Dis ; 27(10): 2619-2627, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34545790

RESUMEN

The numerous global outbreaks and continuous reassortments of highly pathogenic avian influenza (HPAI) A(H5N6/H5N8) clade 2.3.4.4 viruses in birds pose a major risk to the public health. We investigated the tropism and innate host responses of 5 recent HPAI A(H5N6/H5N8) avian isolates of clades 2.3.4.4b, e, and h in human airway organoids and primary human alveolar epithelial cells. The HPAI A(H5N6/H5N8) avian isolates replicated productively but with lower competence than the influenza A(H1N1)pdm09, HPAI A(H5N1), and HPAI A(H5N6) isolates from humans in both or either models. They showed differential cellular tropism in human airway organoids; some infected all 4 major epithelial cell types: ciliated cells, club cells, goblet cells, and basal cells. Our results suggest zoonotic potential but low transmissibility of the HPAI A(H5N6/H5N8) avian isolates among humans. These viruses induced low levels of proinflammatory cytokines/chemokines, which are unlikely to contribute to the pathogenesis of severe disease.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Subtipo H5N1 del Virus de la Influenza A , Subtipo H5N8 del Virus de la Influenza A , Gripe Aviar , Gripe Humana , Animales , Aves , Humanos , Subtipo H5N1 del Virus de la Influenza A/genética , Gripe Aviar/epidemiología , Medición de Riesgo
13.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34099577

RESUMEN

Coronaviruses are pathogens of pandemic potential. Middle East respiratory syndrome coronavirus (MERS-CoV) causes a zoonotic respiratory disease of global public health concern, and dromedary camels are the only proven source of zoonotic infection. More than 70% of MERS-CoV-infected dromedaries are found in East, North, and West Africa, but zoonotic MERS disease is only reported from the Arabian Peninsula. We compared viral replication competence of clade A and B viruses from the Arabian Peninsula with genetically diverse clade C viruses found in East (Egypt, Kenya, and Ethiopia), North (Morocco), and West (Nigeria and Burkina Faso) Africa. Viruses from Africa had lower replication competence in ex vivo cultures of the human lung and in lungs of experimentally infected human-DPP4 (hDPP4) knockin mice. We used lentivirus pseudotypes expressing MERS-CoV spike from Saudi Arabian clade A prototype strain (EMC) or African clade C1.1 viruses and demonstrated that clade C1.1 spike was associated with reduced virus entry into the respiratory epithelial cell line Calu-3. Isogenic EMC viruses with spike protein from EMC or clade C1.1 generated by reverse genetics showed that the clade C1.1 spike was associated with reduced virus replication competence in Calu-3 cells in vitro, in ex vivo human bronchus, and in lungs of hDPP4 knockin mice in vivo. These findings may explain why zoonotic MERS disease has not been reported from Africa so far, despite exposure to and infection with MERS-CoV.


Asunto(s)
Coronavirus del Síndrome Respiratorio de Oriente Medio/genética , Zoonosis/virología , África , Animales , Arabia , Línea Celular , Dipeptidil Peptidasa 4/metabolismo , Técnicas de Sustitución del Gen , Humanos , Cinética , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Fenotipo , Filogenia , Glicoproteína de la Espiga del Coronavirus/metabolismo , Replicación Viral/fisiología
14.
Emerg Infect Dis ; 27(5): 1492-1495, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33900193

RESUMEN

We describe an introduction of clade GH severe acute respiratory syndrome coronavirus 2 causing a fourth wave of coronavirus disease in Hong Kong. The virus has an ORF3a-Q57H mutation, causing truncation of ORF3b. This virus evades induction of cytokine, chemokine, and interferon-stimulated gene expression in primary human respiratory cells.


Asunto(s)
COVID-19 , Epidemias , China , Hong Kong/epidemiología , Humanos , SARS-CoV-2
15.
J Infect Dis ; 224(5): 821-830, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-33395484

RESUMEN

BACKGROUND: Human spillovers of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to dogs and the emergence of a highly contagious avian-origin H3N2 canine influenza virus have raised concerns on the role of dogs in the spread of SARS-CoV-2 and their susceptibility to existing human and avian influenza viruses, which might result in further reassortment. METHODS: We systematically studied the replication kinetics of SARS-CoV-2, SARS-CoV, influenza A viruses of H1, H3, H5, H7, and H9 subtypes, and influenza B viruses of Yamagata-like and Victoria-like lineages in ex vivo canine nasal cavity, soft palate, trachea, and lung tissue explant cultures and examined ACE2 and sialic acid (SA) receptor distribution in these tissues. RESULTS: There was limited productive replication of SARS-CoV-2 in canine nasal cavity and SARS-CoV in canine nasal cavity, soft palate, and lung, with unexpectedly high ACE2 levels in canine nasal cavity and soft palate. Canine tissues were susceptible to a wide range of human and avian influenza viruses, which matched with the abundance of both human and avian SA receptors. CONCLUSIONS: Existence of suitable receptors and tropism for the same tissue foster virus adaptation and reassortment. Continuous surveillance in dog populations should be conducted given the many chances for spillover during outbreaks.


Asunto(s)
COVID-19/virología , Virus de la Influenza A/fisiología , Pulmón/virología , Cavidad Nasal/virología , SARS-CoV-2/fisiología , Tráquea/virología , Tropismo Viral/fisiología , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , COVID-19/metabolismo , Perros , Humanos , Gripe Humana/metabolismo , Gripe Humana/virología , Pulmón/metabolismo , Cavidad Nasal/metabolismo , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Tráquea/metabolismo
17.
Respir Res ; 21(1): 160, 2020 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-32576265

RESUMEN

BACKGROUND: Neutrophil is of the most abundant number in human immune system. During acute influenza virus infection, neutrophils are already active in the early phase of inflammation - a time in which clinical biopsy or autopsy material is not readily available. However, the role of neutrophil in virus infection is not well understood. Here, we studied the role of neutrophil in host defense during influenza A virus infection, specifically assessing if it contributes to the differential pathogenesis in H5N1 disease. METHODS: Neutrophils were freshly isolated from healthy volunteers and subjected to direct influenza H1N1 and H5N1 virus infection in vitro. The ability of the naïve neutrophils to infiltrate from the basolateral to the apical phase of the influenza virus infected alveolar epithelium was assessed. The viral replication, innate immune responses and Neutrophil extracellular trap (NET) formation of neutrophils upon influenza virus infection were evaluated. RESULTS: Our results demonstrated that influenza virus infected alveolar epithelium allowed neutrophil transmigration. Significantly more neutrophils migrated across the H5N1 influenza virus infected the epithelium than the counterpart infected by the seasonal influenza H1N1 virus infected. Neutrophils were equally susceptible to H5N1 and H1N1 virus infection with similar viral gene transcription. Productive replication was observed in H5N1 infected neutrophils. H5N1 induced higher cytokine and chemokine gene transcription than H1N1 infected neutrophils, including TNF-α, IFN-ß, CXCL10, MIP-1α and IL-8. This inferred a more intense inflammatory response posed by H5N1 than H1N1 virus. Strikingly, NADPH oxidase-independent NET formation was only observed in H1N1 infected neutrophils at 6 hpi while no NET formation was observed upon H5N1 infection. CONCLUSION: Our data is the first to demonstrate that NET formation is abrogated in H5N1 influenza virus infection and might contribute to the severity of H5N1 disease.


Asunto(s)
ADN/inmunología , Trampas Extracelulares/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Neutrófilos/inmunología , Adolescente , Adulto , Animales , Células Cultivadas , Niño , Preescolar , Perros , Trampas Extracelulares/virología , Femenino , Humanos , Inmunidad Celular/inmunología , Células de Riñón Canino Madin Darby , Masculino , Neutrófilos/patología , Neutrófilos/virología , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/virología
18.
Lancet Respir Med ; 8(7): 687-695, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32386571

RESUMEN

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in December 2019, causing a respiratory disease (coronavirus disease 2019, COVID-19) of varying severity in Wuhan, China, and subsequently leading to a pandemic. The transmissibility and pathogenesis of SARS-CoV-2 remain poorly understood. We evaluate its tissue and cellular tropism in human respiratory tract, conjunctiva, and innate immune responses in comparison with other coronavirus and influenza virus to provide insights into COVID-19 pathogenesis. METHODS: We isolated SARS-CoV-2 from a patient with confirmed COVID-19, and compared virus tropism and replication competence with SARS-CoV, Middle East respiratory syndrome-associated coronavirus (MERS-CoV), and 2009 pandemic influenza H1N1 (H1N1pdm) in ex-vivo cultures of human bronchus (n=5) and lung (n=4). We assessed extrapulmonary infection using ex-vivo cultures of human conjunctiva (n=3) and in-vitro cultures of human colorectal adenocarcinoma cell lines. Innate immune responses and angiotensin-converting enzyme 2 expression were investigated in human alveolar epithelial cells and macrophages. In-vitro studies included the highly pathogenic avian influenza H5N1 virus (H5N1) and mock-infected cells as controls. FINDINGS: SARS-CoV-2 infected ciliated, mucus-secreting, and club cells of bronchial epithelium, type 1 pneumocytes in the lung, and the conjunctival mucosa. In the bronchus, SARS-CoV-2 replication competence was similar to MERS-CoV, and higher than SARS-CoV, but lower than H1N1pdm. In the lung, SARS-CoV-2 replication was similar to SARS-CoV and H1N1pdm, but was lower than MERS-CoV. In conjunctiva, SARS-CoV-2 replication was greater than SARS-CoV. SARS-CoV-2 was a less potent inducer of proinflammatory cytokines than H5N1, H1N1pdm, or MERS-CoV. INTERPRETATION: The conjunctival epithelium and conducting airways appear to be potential portals of infection for SARS-CoV-2. Both SARS-CoV and SARS-CoV-2 replicated similarly in the alveolar epithelium; SARS-CoV-2 replicated more extensively in the bronchus than SARS-CoV. These findings provide important insights into the transmissibility and pathogenesis of SARS-CoV-2 infection and differences with other respiratory pathogens. FUNDING: US National Institute of Allergy and Infectious Diseases, University Grants Committee of Hong Kong Special Administrative Region, China; Health and Medical Research Fund, Food and Health Bureau, Government of Hong Kong Special Administrative Region, China.


Asunto(s)
Betacoronavirus/inmunología , Conjuntiva/virología , Infecciones por Coronavirus/inmunología , Inmunidad Innata/inmunología , Neumonía Viral/inmunología , Sistema Respiratorio/virología , Tropismo Viral/fisiología , Replicación Viral/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Betacoronavirus/fisiología , COVID-19 , Conjuntiva/inmunología , Conjuntiva/fisiopatología , Infecciones por Coronavirus/fisiopatología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pandemias , Neumonía Viral/fisiopatología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/fisiopatología , Mucosa Respiratoria/virología , Sistema Respiratorio/inmunología , Sistema Respiratorio/fisiopatología , SARS-CoV-2
19.
mBio ; 11(2)2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32127444

RESUMEN

Anti-galactose-α-1,3-galactose (anti-α-Gal) antibody is naturally expressed at a high level in humans. It constitutes about 1% of immunoglobulins found in human blood. Here, we designed a live attenuated influenza virus vaccine that can generate α-Gal epitopes in infected cells in order to facilitate opsonization of infected cells, thereby enhancing vaccine-induced immune responses. In the presence of normal human sera, cells infected with this mutant can enhance phagocytosis of human macrophages and cytotoxicity of NK cells in vitro Using a knockout mouse strain that allows expression of anti-α-Gal antibody in vivo, we showed that this strategy can increase vaccine immunogenicity and the breadth of protection. This vaccine can induce 100% protection against a lethal heterosubtypic group 1 (H5) or group 2 (mouse-adapted H3) influenza virus challenge in the mouse model. In contrast, its heterosubtypic protective effect in wild-type or knockout mice that do not have anti-α-Gal antibody expression is only partial, demonstrating that the enhanced vaccine-induced protection requires anti-α-Gal antibody upon vaccination. Anti-α-Gal-expressing knockout mice immunized with this vaccine produce robust humoral and cell-mediated responses upon a lethal virus challenge. This vaccine can stimulate CD11blo/- pulmonary dendritic cells, which are known to be crucial for clearance of influenza virus. Our approach provides a novel strategy for developing next-generation influenza virus vaccines.IMPORTANCE Influenza A viruses have multiple HA subtypes that are antigenically diverse. Classical influenza virus vaccines are subtype specific, and they cannot induce satisfactory heterosubtypic immunity against multiple influenza virus subtypes. Here, we developed a live attenuated H1N1 influenza virus vaccine that allows the expression of α-Gal epitopes by infected cells. Anti-α-Gal antibody is naturally produced by humans. In the presence of this antibody, human cells infected with this experimental vaccine virus can enhance several antibody-mediated immune responses in vitro Importantly, mice expressing anti-α-Gal antibody in vivo can be fully protected by this H1N1 vaccine against a lethal H5 or H3 virus challenge. Our work demonstrates a new strategy for using a single influenza virus strain to induce broadly cross-reactive immune responses against different influenza virus subtypes.


Asunto(s)
Reacciones Cruzadas/inmunología , Epítopos/inmunología , Galactosa/inmunología , Inmunogenicidad Vacunal , Vacunas contra la Influenza/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Animales , Humanos , Virus de la Influenza A/clasificación , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/genética , Células Asesinas Naturales/inmunología , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología
20.
Eur Respir J ; 54(2)2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31097520

RESUMEN

Despite causing regular seasonal epidemics with substantial morbidity, mortality and socioeconomic burden, there is still a lack of research into influenza B viruses (IBVs). In this study, we provide for the first time a systematic investigation on the tropism, replication kinetics and pathogenesis of IBVs in the human respiratory tract.Physiologically relevant ex vivo explant cultures of human bronchus and lung, human airway organoids, and in vitro cultures of differentiated primary human bronchial epithelial cells and type-I-like alveolar epithelial cells were used to study the cellular and tissue tropism, replication competence and induced innate immune response of 16 IBV strains isolated from 1940 to 2012 in comparison with human seasonal influenza A viruses (IAVs), H1N1 and H3N2. IBVs from the diverged Yamagata- and Victoria-like lineages and the earlier undiverged period were included.The majority of IBVs replicated productively in human bronchus and lung with similar competence to seasonal IAVs. IBVs infected a variety of cell types, including ciliated cells, club cells, goblet cells and basal cells, in human airway organoids. Like seasonal IAVs, IBVs are low inducers of pro-inflammatory cytokines and chemokines. Most results suggested a higher preference for the conducting airway than the lower lung and strain-specific rather than lineage-specific pathogenicity of IBVs.Our results highlighted the non-negligible virulence of IBVs which require more attention and further investigation to alleviate the disease burden, especially when treatment options are limited.


Asunto(s)
Virus de la Influenza B/fisiología , Organoides/patología , Organoides/virología , Sistema Respiratorio/patología , Sistema Respiratorio/virología , Tropismo Viral , Animales , Bronquios/patología , Diferenciación Celular , Perros , Células Epiteliales/virología , Eritrocitos/citología , Humanos , Inmunidad Innata , Inmunohistoquímica , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H3N2 del Virus de la Influenza A/fisiología , Concentración 50 Inhibidora , Pulmón/patología , Células de Riñón Canino Madin Darby , Técnicas de Cultivo de Órganos , Pavos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA