Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 555
Filter
1.
Carbohydr Polym ; 337: 122157, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38710573

ABSTRACT

Seaweed polysaccharides, particularly sulfated ones, exhibited potent antiviral activity against a wide variety of enveloped viruses, such as herpes simplex virus and respiratory viruses. Different mechanisms of action were suggested, which may range from preventing infection to intracellular antiviral activity, at different stages of the viral cycle. Herein, we generated two chemically engineered sulfated fucans (C303 and C304) from Cystoseira indica by an amalgamated extraction-sulfation procedure using chlorosulfonic acid-pyridine/N,N-dimethylformamide and sulfur trioxide-pyridine/N,N-dimethylformamide reagents, respectively. These compounds exhibited activity against HSV-1 and RSV with 50 % inhibitory concentration values in the range of 0.75-2.5 µg/mL and low cytotoxicity at concentrations up to 500 µg/mL. The antiviral activities of chemically sulfated fucans (C303 and C304) were higher than the water (C301) and CaCl2 extracted (C302) polysaccharides. Compound C303 had a (1,3)-linked fucan backbone and was branched. Sulfates were present at positions C-2, C-4, and C-2,4 of Fucp, and C-6 of Galp residues of this polymer. Compound C304 had a comparable structure but with more sulfates at C-4 of Fucp residue. Both C303 and C304 were potent antiviral candidates, acting in a dose-dependent manner on the adsorption and other intracellular stages of HSV-1 and RSV replication, in vitro.


Subject(s)
Antiviral Agents , Herpesvirus 1, Human , Polysaccharides , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Chlorocebus aethiops , Herpesvirus 1, Human/drug effects , Polysaccharides/pharmacology , Polysaccharides/chemistry , Polysaccharides/isolation & purification , Animals , Vero Cells , Humans , Sulfates/chemistry , Sulfates/pharmacology , Respiratory Syncytial Viruses/drug effects
2.
Fitoterapia ; 175: 105982, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38685512

ABSTRACT

A phytochemical investigation on the buds of edible medicinal plant, Eugenia carvophyllata, led to the discovery of seven new compounds, caryophones A-G (1-7), along with two biogenetically-related known ones, 2-methoxy-7-methyl-1,4-naphthalenedione (8) and eugenol (9). Compounds 1-3 represent the first examples of C-5-C-1' connected naphthoquinone-monoterpene adducts with a new carbon skeleton. Compounds 4-7 are a class of novel neolignans with unusual linkage patterns, in which the C-9 position of one phenylpropene unit coupled with the aromatic core of another phenylpropene unit. The chemical structures of the new compounds were determined based on extensive spectroscopic analysis, X-ray diffraction crystallography, and quantum-chemical calculation. Among the isolates, compounds (-)-2, 3, 6, and 9 showed significant in vitro inhibitory activities against respiratory syncytial virus (RSV)-induced nitric oxide (NO) production in RAW264.7 cells.


Subject(s)
Anti-Inflammatory Agents , Eugenia , Lignans , Naphthoquinones , Nitric Oxide , Phytochemicals , Mice , RAW 264.7 Cells , Animals , Nitric Oxide/metabolism , Molecular Structure , Lignans/pharmacology , Lignans/isolation & purification , Lignans/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/chemistry , Naphthoquinones/pharmacology , Naphthoquinones/isolation & purification , Naphthoquinones/chemistry , Phytochemicals/pharmacology , Phytochemicals/isolation & purification , Eugenia/chemistry , Respiratory Syncytial Viruses/drug effects , China
3.
J Cell Mol Med ; 28(9): e18338, 2024 May.
Article in English | MEDLINE | ID: mdl-38683122

ABSTRACT

Respiratory syncytial virus (RSV) infects neuronal cells in the central nervous system (CNS), resulting in neurological symptoms. In the present study, we intended to explore the mechanism of RSV infection-induced neuroinflammatory injury from the perspective of the immune response and sought to identify effective protective measures against the injury. The findings showed that toll-like receptor 4 (TLR4) was activated after RSV infection in human neuronal SY5Y cells. Furthermore, TLR4 activation induced autophagy and apoptosis in neuronal cells, promoted the formation of the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, and increased the secretion of downstream inflammatory cytokines such as interleukin-1ß (IL-1ß), interleukin-18 (IL-18) and tumour necrosis factor-α (TNF-α). Interestingly, blockade of TLR4 or treatment with exogenous melatonin significantly suppressed TLR4 activation as well as TLR4-mediated apoptosis, autophagy and immune responses. Therefore, we infer that melatonin may act on the TLR4 to ameliorate RSV-induced neuronal injury, which provides a new therapeutic target for RSV infection.


Subject(s)
Apoptosis , Autophagy , Inflammasomes , Melatonin , NLR Family, Pyrin Domain-Containing 3 Protein , Respiratory Syncytial Virus Infections , Toll-Like Receptor 4 , Humans , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Central Nervous System/virology , Central Nervous System/metabolism , Central Nervous System/drug effects , Central Nervous System/pathology , Cytokines/metabolism , Inflammasomes/drug effects , Inflammasomes/metabolism , Melatonin/pharmacology , Neurons/metabolism , Neurons/drug effects , Neurons/virology , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Viruses/drug effects , Respiratory Syncytial Viruses/physiology , Toll-Like Receptor 4/drug effects , Toll-Like Receptor 4/metabolism
4.
J Nat Prod ; 87(4): 1003-1012, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38579352

ABSTRACT

Three new (1-3) and six known rotenoids (5-10), along with three known isoflavones (11-13), were isolated from the leaves of Millettia oblata ssp. teitensis. A new glycosylated isoflavone (4), four known isoflavones (14-18), and one known chalcone (19) were isolated from the root wood extract of the same plant. The structures were elucidated by NMR and mass spectrometric analyses. The absolute configuration of the chiral compounds was established by a comparison of experimental ECD and VCD data with those calculated for the possible stereoisomers. This is the first report on the use of VCD to assign the absolute configuration of rotenoids. The crude leaves and root wood extracts displayed anti-RSV (human respiratory syncytial virus) activity with IC50 values of 0.7 and 3.4 µg/mL, respectively. Compounds 6, 8, 10, 11, and 14 showed anti-RSV activity with IC50 values of 0.4-10 µM, while compound 3 exhibited anti-HRV-2 (human rhinovirus 2) activity with an IC50 of 4.2 µM. Most of the compounds showed low cytotoxicity for laryngeal carcinoma (HEp-2) cells; however compounds 3, 11, and 14 exhibited low cytotoxicity also in primary lung fibroblasts. This is the first report on rotenoids showing antiviral activity against RSV and HRV viruses.


Subject(s)
Antiviral Agents , Isoflavones , Millettia , Isoflavones/pharmacology , Isoflavones/chemistry , Isoflavones/isolation & purification , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Millettia/chemistry , Molecular Structure , Humans , Rotenone/pharmacology , Rotenone/chemistry , Rotenone/analogs & derivatives , Plant Leaves/chemistry , Plant Roots/chemistry , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Viruses/drug effects
5.
J Hosp Infect ; 147: 83-86, 2024 May.
Article in English | MEDLINE | ID: mdl-38490488

ABSTRACT

BACKGROUND: Respiratory viruses have been reported to infect the salivary glands and the throat, which are potential reservoirs for virus replication and transmission. Therefore, strategies to reduce the amount of infective virus particles in the oral mucous membranes could lower the risk of transmission. METHODS: The viral inactivation capacity of a plant-oil-based oral rinse (Salviathymol®) was evaluated in comparison with chlorhexidine (Chlorhexamed® FORTE) using a quantitative suspension test according to EN 14476. FINDINGS: Salviathymol efficiently inactivated severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), respiratory syncytial virus (RSV) and two influenza strains to undetectable levels. CONCLUSION: Salviathymol has potential as preventive measure to lower transmission of respiratory viruses.


Subject(s)
Mouthwashes , SARS-CoV-2 , Humans , Mouthwashes/pharmacology , SARS-CoV-2/drug effects , Plant Oils/pharmacology , Antiviral Agents/pharmacology , Virus Inactivation/drug effects , Respiratory Syncytial Viruses/drug effects , COVID-19/prevention & control
6.
J Nat Prod ; 87(5): 1338-1346, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38447084

ABSTRACT

Oxabornyl polyenes represent a unique group of polyketides characterized by a central polyene core flanked by a conserved oxabornyl moiety and a structurally diverse oxygen heterocyclic ring. They are widely distributed in fungi and possess a variety of biological activities. Due to the significant spatial separation between the two stereogenic ring systems, it is difficult to establish their overall relative configurations. Here, we isolated three oxabornyl polyenes, prugosenes A1-A3 (1-3), from Talaromyces sp. JNU18266-01. Although these compounds were first reported from Penicillium rugulosum, their overall relative and absolute configurations remained unassigned. By employing ozonolysis in combination with ECD calculations, we were able to establish their absolute configurations, and additionally obtained seven new chemical derivatives (4-10). Notably, through NMR data analysis and quantum chemical calculations, we achieved the structural revision of prugosene A2. Furthermore, prugosenes A1-A3 exhibited potent antiviral activity against the respiratory syncytial virus, with compound 1 displaying an IC50 value of 6.3 µM. Our study thus provides a valuable reference for absolute configuration assignment of oxabornyl polyene compounds.


Subject(s)
Polyenes , Polyenes/chemistry , Polyenes/pharmacology , Molecular Structure , Talaromyces/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Respiratory Syncytial Viruses/drug effects , Humans
7.
ChemMedChem ; 19(9): e202400057, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38385828

ABSTRACT

A 1H-isoindol-3-amine was identified as suitable P1 group for the proprotein convertase furin using a crystallographic screening with a set of 20 fragments known to occupy the S1 pocket of trypsin-like serine proteases. Its binding mode is very similar to that observed for the P1 group of benzamidine-derived peptidic furin inhibitors suggesting an aminomethyl substitution of this fragment to obtain a couplable P1 residue for the synthesis of substrate-analogue furin inhibitors. The obtained inhibitors possess a slightly improved picomolar inhibitory potency compared to their benzamidine-derived analogues. The crystal structures of two inhibitors in complex with furin revealed that the new P1 group is perfectly suited for incorporation in peptidic furin inhibitors. Selected inhibitors were tested for antiviral activity against respiratory syncytial virus (RSV) and a furin-dependent influenza A virus (SC35M/H7N7) in A549 human lung cells and demonstrated an efficient inhibition of virus activation and replication at low micromolar or even submicromolar concentrations. First results suggest that the Mas-related G-protein coupled receptor GPCR-X2 could be a potential off-target for certain benzamidine-derived furin inhibitors.


Subject(s)
Antiviral Agents , Drug Design , Furin , Furin/antagonists & inhibitors , Furin/metabolism , Humans , Antiviral Agents/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Structure-Activity Relationship , A549 Cells , Influenza A virus/drug effects , Crystallography, X-Ray , Indoles/pharmacology , Indoles/chemistry , Indoles/chemical synthesis , Molecular Structure , Models, Molecular , Respiratory Syncytial Viruses/drug effects , Dose-Response Relationship, Drug
8.
Chem Biodivers ; 19(1): e202100668, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34812586

ABSTRACT

Forsyqinlingines C (1) and D (2), two C9 -monoterpenoid alkaloids bearing a rare skeleton, were isolated from the ripe fruits of Forsythia suspensa. Their structures, including absolute configurations, were fully elucidated by extensive spectroscopic data and ECD experiments. The plausible biogenetic pathway for compounds 1 and 2 was also proposed. In vitro, two C9 -monoterpenoid alkaloids showed anti-inflammatory activity performed by the inhibitory effect on the release of ß-glucuronidase in rat polymorphonuclear leukocytes (PMNs), as well as antiviral activity against influenza A (H1N1) virus and respiratory syncytial virus (RSV).


Subject(s)
Alkaloids/chemistry , Anti-Inflammatory Agents/chemistry , Antiviral Agents/chemistry , Forsythia/chemistry , Monoterpenes/chemistry , Alkaloids/isolation & purification , Alkaloids/pharmacology , Animals , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/pharmacology , Antiviral Agents/isolation & purification , Antiviral Agents/pharmacology , Forsythia/metabolism , Fruit/chemistry , Fruit/metabolism , Glucuronidase/metabolism , Influenza A Virus, H1N1 Subtype/drug effects , Magnetic Resonance Spectroscopy , Molecular Conformation , Neutrophils/cytology , Neutrophils/drug effects , Neutrophils/metabolism , Platelet Activating Factor/pharmacology , Rats , Respiratory Syncytial Viruses/drug effects
9.
Int J Antimicrob Agents ; 58(6): 106460, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34695564

ABSTRACT

L-sulforaphane (LSF) is an isothiocyanate derived from cruciferous vegetables that has long been known for its anticarcinogenic, antioxidant and anti-inflammatory effects. LSF also possesses antimicrobial properties, although the evidence for this is limited. Respiratory pathogens, such as Streptococcus pneumoniae, Haemophilus influenzae, Streptococcus pyogenes and respiratory syncytial virus (RSV), are leading global causes of illness and death among children aged under five years, particularly in resource-poor countries where access to vaccines are limited or, in the case of S. pyogenes and RSV, vaccines have not been licensed for use in humans. Therefore, alternative strategies to prevent and/or treat these common infectious diseases are urgently needed. This study was conducted to investigate the antimicrobial effects of LSF against common respiratory pathogens, S. pneumoniae (serotypes 1 and 6B), H. influenzae type B (HiB), non-typeable H. influenzae (NTHi), S. pyogenes and RSV in relevant human cell-based models. LSF significantly inhibited the growth of H. influenzae, but not S. pneumoniae or S. pyogenes. LSF did not improve opsonophagocytic capacity or killing by human phagocytic cell lines (HL-60s and THP-1 macrophages) for S. pneumoniae yet showed some improved killing for H. influenzae species in THP-1 macrophages. However, LSF significantly reduced RSV infection in human lung epithelial cells, associated with increased expression of cyclin D1 (CCND1) gene as well as the antioxidant genes, nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase 1 (HMOX-1). Overall, LSF represents an exciting avenue for further antimicrobial research, particularly as a novel therapy against H. influenzae species and RSV.


Subject(s)
Anti-Bacterial Agents/pharmacology , Haemophilus Infections/drug therapy , Isothiocyanates/pharmacology , Pneumococcal Infections/drug therapy , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Tract Infections/drug therapy , Sulfoxides/pharmacology , Cell Line , Cyclin D1/metabolism , HL-60 Cells , Haemophilus influenzae/drug effects , Haemophilus influenzae/growth & development , Heme Oxygenase-1/metabolism , Humans , Macrophages/drug effects , Macrophages/immunology , Microbial Sensitivity Tests , NF-E2-Related Factor 2/metabolism , Opsonization/drug effects , Respiratory Syncytial Viruses/drug effects , Respiratory Tract Infections/microbiology , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/growth & development , Streptococcus pyogenes/drug effects , Streptococcus pyogenes/growth & development , THP-1 Cells , Vegetables/chemistry
10.
Lima; IETSI; oct. 2021.
Non-conventional in Spanish | BRISA/RedTESA | ID: biblio-1358161

ABSTRACT

INTRODUCCIÓN: El presente dictamen expone la evaluación de la eficacia y seguridad de palivizumab para prevenir la enfermedad grave causada por el VSR en niños con DBP que hayan recibido tratamiento para la DBP en los últimos seis meses y tengan antecedente de haber nacido a las 29 semanas o menos de gestación. A nivel mundial, el virus sincitial respiratorio (VSR) es una de las principales causas de infección del tracto respiratorio inferior en niños. En Perú, el VSR ha sido detectado en el 86 % de niños menores de un año y en el 76 % de niños de uno a cuatro años, con infecciones respiratorias agudas. La displasia broncopulmonar (DBP) y la prematuridad son factores de riesgo para el desarrollo de enfermedad grave por VSR y están asociadas con mayores tasas de hospitalización, 388/1000 en niños con DBP y 57/1000 en niños prematuros, y muerte, tasas de casos fatales que van del 3.5 % al 23 % en niños con DBP y del 1.2 % al 6.1 % en niños prematuros. Un aspecto importante de la epidemiología del VSR es que difiere de acuerdo con las condiciones meteorológicas de las regiones. Por ejemplo, en las regiones tropicales y subtropicales, como es el caso de Perú, las infecciones por VSR se distribuyen de manera uniforme a lo largo de todo el año, con algunos aumentos variables. Actualmente, no se cuenta con un tratamiento antiviral efectivo para las infecciones por VSR, por lo que las medidas profilácticas toman gran importancia. Palivizumab, un anticuerpo monoclonal, es usado como una medida para prevenir el desarrollo de la enfermedad grave causada por el VSR, que requiera hospitalización, en niños susceptibles a desarrollar enfermedad. En el contexto de EsSalud, no se cuenta con una medida profiláctica frente a la enfermedad grave causada por el VSR. Los especialistas de EsSalud consideran que palivizumab ayudaría a prevenir la hospitalización, mortalidad y morbilidad causadas por la infección por VSR, en pacientes con alto riesgo de desarrollar enfermedad grave. En ese sentido, el presente dictamen expone la evaluación de la eficacia y seguridad de palivizumab, comparado con placebo, en la prevención de la enfermedad grave causada por el VSR en niños con DBP, que hayan recibido tratamiento para la DBP en los últimos seis meses, y con antecedente de haber nacido a las 29 semanas, o menos, de gestación. METODOLOGÍA: La búsqueda de la literatura se realizó con el objetivo de identificar evidencia sobre la eficacia y seguridad de palivizumab, en comparación con placebo, para prevenir la enfermedad grave causada por el VSR en niños con DBP que hayan recibido tratamiento para la DBP en los últimos seis meses, con antecedente de haber nacido a las 29 semanas o menos de gestación. La búsqueda de la evidencia se realizó en las bases de datos bibliográficas: PubMed, LILACS y The Cochrane Library. Adicionalmente, se amplió la búsqueda revisando la evidencia generada por grupos internacionales que realizan revisiones sistemáticas, evaluaciones de tecnologías sanitarias y guías de práctica clínica, tales como The National Institute for Health and Care Excellence (NICE), The Canadian Agency for Drugs and Technologies in Health (CADTH), Centro Nacional de Excelencia Tecnológica en Salud (CENETEC), Scottish Intercollegiate Guidelines Network (SIGN), Institute for Quality and Efficiency in Health Care (IQWiG), Scottish Medicines Consortium (SMC), Agency for Healthcare Research and Quality's (AHRQ), National Health and Medical Research Council (NHMRC), New Zealand Guidelines Group (NZGG), Canadian Medical Association (CMA), American College of Physicians Clinical Practice Guidelines y Registered Nurses Association of Ontario (RNAO). Adicionalmente, se realizó una búsqueda manual en las bases The Guidelines International Network (G-I-N), eGuidelines, y el portal de la Base Regional de Informes de Evaluación de Tecnologías en Salud de las Américas (BRISA). Finalmente, se realizó una búsqueda manual en el portal ClinicalTrials.gov del National Institutes of Health (NIH) para identificar ensayos clínicos en desarrollo o cuyos resultados aún no hayan sido publicados en una revista científica. RESULTADOS: Se describe la evidencia disponible según el tipo de publicación, siguiendo lo indicado en los criterios de elegibilidad. CONCLUSIONES: En el presente documento, se evaluó la mejor evidencia científica disponible hasta agosto del 2021, en relación con la eficacia y seguridad de palivizumab en la prevención de la enfermedad grave causada por el VSR, que requiere hospitalización, en niños con DBP que hayan recibido tratamiento para la DBP en los últimos seis meses, con antecedente de haber nacido a las 29 semanas o menos de gestación. En EsSalud no se cuenta con una medida profiláctica frente al desarrollo de enfermedad grave causada por el VSR, que requiera hospitalización, para niños con alto riesgo; enmarcándose en un contexto de vacío terapéutico. La búsqueda sistemática de la evidencia culminó con la selección de dos GPC (Castaños y Rodríguez 2019; Ralston et al. 2014), dos ETS (Ministerio de Salud de Chile 2017; DIGEMID 2016), y el ECA de fase III IMPACT (The IMpact-RSV Study Group 1998). Las guías de Castaños y Rodríguez, y de Ralston et al., coinciden en recomendar la profilaxis con palivizumab en niños con DBP y prematuridad; aunque difieren en el rango de EG del paciente prematuro y el grado de DBP. Ambas guías tomaron en consideración los resultados del ECA IMPACT para emitir sus recomendaciones. Las dos ETS, del Ministerio de Salud de Chile y la DIGEMID de Perú, brindan recomendaciones opuestas sobre el uso profiláctico de palivizumab en niños con DBP y prematuridad. El Ministerio de Salud de Chile aprobó la ampliación de uso de palivizumab en pacientes prematuros menores a 36 semanas con patologías o condiciones de riesgo asociadas. En cambio, la DIGEMID de Perú decide no incluir a palivizumab en el PNUME para su uso en neonatos con EG de 29 a 32 semanas con DBP y edad corregida menor o igual a 3 meses al inicio de la profilaxis. Ambas ETS tomaron en cuenta los resultados del ECA IMPACT. El ECA IMPACT, evaluó el uso de palivizumab versus placebo en niños prematuros con edad menor o igual a seis meses, y niños con DBP y edad menor o igual a dos años. El ECA reportó la reducción de la incidencia de hospitalizaciones por VSR en los niños que recibieron palivizumab en comparación con placebo; un menor número de ingresos a UCI a favor de palivizumab. No se observaron diferencias en la incidencia de ventilación mecánica, duración de ventilación mecánica, mortalidad y el reporte de EA, entre el uso de placebo y palivizumab. El ECA IMPACT (estudio de fase III, doble ciego y pivotal de palivizumab) es el único ECA disponible que evalúa la eficacia comparativa de palivizumab. Las limitaciones del ECA IMPACT afectan la validez y precisión de sus resultados. Tomando en cuenta estas limitaciones, el uso de palivizumab sería eficaz en reducir la incidencia de hospitalización solo en el subgrupo de pacientes de 6 meses de edad o menos y prematuridad (EG menor o igual a 35 semanas). El contexto de vacío terapéutico y la evidencia disponible sugieren que el uso de palivizumab podría tener un impacto significativo en la incidencia de hospitalizaciones por VSR en la población objetivo del presente dictamen (pacientes con DBP y antecedente de prematuridad). Por lo expuesto, el IETSI aprueba el uso de palivizumab para la prevención de la enfermedad grave causada por el VSR en niños con DBP que hayan recibido tratamiento para la DBP en los últimos seis meses, con antecedente de haber nacido a las 29 semanas o menos de gestación, según lo establecido en el Anexo N°1. La vigencia del presente dictamen preliminar es de un año a partir de la fecha de publicación. Así, la continuación de dicha aprobación estará sujeta a la evaluación de los resultados obtenidos y de mayor evidencia que pueda surgir en el tiempo.


Subject(s)
Humans , Child , Respiratory Syncytial Viruses/drug effects , Bronchopulmonary Dysplasia/physiopathology , Respiratory Syncytial Virus Infections/drug therapy , Palivizumab/therapeutic use , Efficacy , Cost-Benefit Analysis
11.
Biomed Res Int ; 2021: 6471400, 2021.
Article in English | MEDLINE | ID: mdl-34485521

ABSTRACT

OBJECTIVE: Exploration of the underlying molecular mechanism of Jinchan Oral Liquid (JOL) in treating children with the respiratory syncytial virus (RSV) pneumonia to provide new evidence for the clinical application. METHODS: The active components and target genes of JOL were screened by the TCMSP database. The targets of RSV pneumonia were obtained from the GeneCards, OMIM, DrugBank, and PharmGKB database. Then, we constructed the active component-target network and screened the core genes. The overlaps were screened for PPI network analysis, GO analysis, and KEGG analysis. Finally, result validation was performed by molecular docking. RESULTS: According to the screening criteria of the ADME, 74 active compounds of JOL were obtained; after removing redundant targets, we selected 180 potential targets. By screening the online database, 893 RSV pneumonia-related targets were obtained. A total of 82 overlapping genes were chosen by looking for the intersection. The STRING online database was used to acquire PPI relationships, and 16 core genes were obtained. GO and KEGG analyses showed that the main pathways of JOL in treating RSV pneumonia include TNF signaling pathway and IL17 signaling pathway. The molecular docking results showed that the active compounds of JOL had a good affinity with the core genes. CONCLUSION: In this study, we preliminarily discussed the main active ingredients, related targets, and pathways of JOL and predicted the pharmacodynamic basis and the potential therapeutic mechanisms of RSV pneumonia. In summary, the network pharmacology strategy may be helpful for the discovery of multitarget drugs against complex diseases.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Gene Regulatory Networks/drug effects , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Viruses/drug effects , Child , Computational Biology/methods , Databases, Genetic , Drug Development/methods , Drugs, Chinese Herbal/chemistry , Humans , Molecular Docking Simulation , Protein Interaction Maps , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/isolation & purification , Signal Transduction
12.
PLoS One ; 16(7): e0253022, 2021.
Article in English | MEDLINE | ID: mdl-34252093

ABSTRACT

Influenza and RSV are human viruses responsible for outbreaks in hospitals, long-term care facilities and nursing homes. The present study assessed an air treatment using ozone at two relative humidity conditions (RHs) in order to reduce the infectivity of airborne influenza. Bovine pulmonary surfactant (BPS) and synthetic tracheal mucus (STM) were used as aerosols protectants to better reflect the human aerosol composition. Residual ozone concentration inside the aerosol chamber was also measured. RSV's sensitivity resulted in testing its resistance to aerosolization and sampling processes instead of ozone exposure. The results showed that without supplement and with STM, a reduction in influenza A infectivity of four orders of magnitude was obtained with an exposure to 1.70 ± 0.19 ppm of ozone at 76% RH for 80 min. Consequently, ozone could be considered as a virucidal disinfectant for airborne influenza A. RSV did not withstand the aerosolization and sampling processes required for the use of the experimental setup. Therefore, ozone exposure could not be performed for this virus. Nonetheless, this study provides great insight for the efficacy of ozone as an air treatment for the control of nosocomial influenza A outbreaks.


Subject(s)
Influenza A virus/drug effects , Ozone/pharmacology , Respiratory Syncytial Viruses/drug effects , Virus Inactivation/drug effects , Aerosols , Air Microbiology , Cross Infection/prevention & control , Disinfection/methods , Humans , Influenza, Human/prevention & control , Ozone/administration & dosage , Real-Time Polymerase Chain Reaction , Respiratory Syncytial Virus Infections/prevention & control
13.
Bioorg Med Chem ; 39: 116157, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33895704

ABSTRACT

Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infection in young children, and specific treatment for RSV infections remains unavailable. We herein reported a series of substituted N-(4-amino-2-chlorophenyl)-5-chloro-2-hydroxybenzamide analogues as potent RSV inhibitors. Among them, six low cytotoxic compounds (11, 12, 15, 22, 26, and 28) have been identified and selected to study associated inhibitory mechanisms. All these compounds suppressed not only the viral replication but also RSV-induced IRF3 and NF-κB activation and associated production of cytokines/chemokines. The two most potent compounds (15 and 22) were selected for further molecular mechanism studies associated with their suppression effect on RSV-activated IRF3 and NF-κB. These two compounds decreased RSV-induced IRF3 phosphorylation at serine 396 and p65 phosphorylation at serine 536 at both early and late infection phases. In addition, compound 22 also inhibited RSV-induced p65 phosphorylation at serine 276 at the late phase of RSV infection.


Subject(s)
Antiviral Agents/pharmacology , Benzamides/chemistry , Benzamides/pharmacology , Inflammation/drug therapy , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Viruses/drug effects , Virus Replication/drug effects , A549 Cells , Benzamides/therapeutic use , Humans , Inflammation/etiology , Respiratory Syncytial Virus Infections/complications , Respiratory Syncytial Viruses/physiology
14.
Rev Med Virol ; 31(6): e2234, 2021 11.
Article in English | MEDLINE | ID: mdl-33724604

ABSTRACT

The coronavirus disease (Covid-19) pandemic is the most serious event of the year 2020, causing considerable global morbidity and mortality. The goal of this review is to provide a comprehensive summary of reported associations between inter-individual immunogenic variants and disease susceptibility or symptoms caused by the coronavirus strains severe acute respiratory syndrome-associated coronavirus, severe acute respiratory syndrome-associated coronavirus-2, and two of the main respiratory viruses, respiratory syncytial virus and influenza virus. The results suggest that the genetic background of the host could affect the levels of proinflammatory and anti-inflammatory cytokines and might modulate the progression of Covid-19 in affected patients. Notably, genetic variations in innate immune components such as toll-like receptors and mannose-binding lectin 2 play critical roles in the ability of the immune system to recognize coronavirus and initiate an early immune response to clear the virus and prevent the development of severe symptoms. This review provides promising clues related to the potential benefits of using immunotherapy and immune modulation for respiratory infectious disease treatment in a personalized manner.


Subject(s)
COVID-19/immunology , Cytokine Release Syndrome/immunology , Genetic Predisposition to Disease , Influenza, Human/immunology , Respiratory Syncytial Virus Infections/immunology , Severe Acute Respiratory Syndrome/immunology , Antiviral Agents/therapeutic use , Biological Variation, Individual , COVID-19/genetics , COVID-19/virology , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/genetics , Cytokine Release Syndrome/virology , Gene Expression , Humans , Immunity, Innate , Immunologic Factors/therapeutic use , Influenza, Human/drug therapy , Influenza, Human/genetics , Influenza, Human/virology , Mannose-Binding Lectin/genetics , Mannose-Binding Lectin/immunology , Orthomyxoviridae/drug effects , Orthomyxoviridae/immunology , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/drug effects , Respiratory Syncytial Viruses/immunology , Severe acute respiratory syndrome-related coronavirus/drug effects , Severe acute respiratory syndrome-related coronavirus/immunology , SARS-CoV-2/classification , SARS-CoV-2/drug effects , SARS-CoV-2/immunology , Severe Acute Respiratory Syndrome/drug therapy , Severe Acute Respiratory Syndrome/genetics , Severe Acute Respiratory Syndrome/virology , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , COVID-19 Drug Treatment
15.
Eur J Med Chem ; 214: 113208, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33571829

ABSTRACT

We report herein the synthesis of a series of novel quinoline derivatives, based on the lead compound 1a, identified from a rRSV-mGFP high-throughput screening assay. Our results revealed that target compounds 1b, 1g-h, 1af and 1ah (IC50 = 3.10-6.93 µM) had good in vitro activity against RSV, which were better than 1a and ribavirin. In addition, we found that compound 1g displayed the lower cytotoxicity (CC50: 2490.33 µM) and the highest selective index (SI = 673.06), suggesting its promising potential as a candidate for further development. On the other hand, compounds 1a, 1m, 1v, 1ad-1af and 1ah-1ai (IC50s: 1.87-14.28 µM) were more active against IAV than or comparable to ribavirin (IC50: 15.36 ± 0.93 µM). Particularly, the most active compound 1ae (IC50: 1.87 ± 0.58 µM) was found to be 8.2-fold more potent than the reference drug, which could inhibit the virus transcription and replication cycle at an early stage.


Subject(s)
Antiviral Agents/pharmacology , Influenza A virus/drug effects , Quinolines/pharmacology , Respiratory Syncytial Viruses/drug effects , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Cell Line , Dose-Response Relationship, Drug , Humans , Microbial Sensitivity Tests , Molecular Structure , Quinolines/chemical synthesis , Quinolines/chemistry , Structure-Activity Relationship
16.
Allergol. immunopatol ; 49(1): 153-158, ene.-feb. 2021. tab
Article in English | IBECS | ID: ibc-199237

ABSTRACT

Although there is increasing evidence showing that infants with viral bronchiolitis exhibit a high degree of heterogeneity, a core uncertainty shared by many clinicians is with regard to understanding which patients are most likely to benefit from bronchodilators such as albuterol. Based on our review, we concluded that older infants with rhinovirus (RV) bronchiolitis, espe­cially those with a nasopharyngeal microbiome dominated by Haemophilus influenzae; those affected during nonpeak months or during non-respiratory syncytial virus (RSV) predominant months; those with wheezing at presentation; those with clinical characteristics such as atopic dermatitis or a family history of asthma in a first-degree relative; and those infants infected with RSV genotypes ON1 and BA, have the greatest likelihood of benefiting from albuterol. Presently, this patient profile could serve as the basis for rational albuterol administration in patients with viral bronchiolitis, at least on a therapeutic trial basis, and it could also be the starting point for future targeted randomized clinical trials (RCTs) on the use of albuterol among a subset of infants with bronchiolitis


No disponible


Subject(s)
Humans , Male , Female , Child, Preschool , Bronchiolitis, Viral/etiology , Bronchiolitis, Viral/drug therapy , Albuterol/administration & dosage , Bronchiolitis, Viral/immunology , Practice Guidelines as Topic , Hospitalization , Respiratory Syncytial Viruses/drug effects , Respiratory Syncytial Viruses/genetics
18.
Front Immunol ; 11: 1673, 2020.
Article in English | MEDLINE | ID: mdl-32849580

ABSTRACT

Respiratory syncytial virus (RSV) remains the most common cause of lower respiratory tract infections in children worldwide. Development of a vaccine has been hindered by the risk of developing enhanced respiratory disease (ERD) upon natural exposure to the virus. Generation of higher quality neutralizing antibodies with stabilized pre-fusion F protein antigens has been proposed as a strategy to prevent ERD. We sought to test whether there was evidence of ERD in naïve BALB/c mice immunized with an unadjuvanted, stabilized pre-fusion F protein, and challenged with RSV line 19. We further sought to determine the extent to which formulation with a Th2-biased (alum) or a more Th1/Th2-balanced (Advax-SM) adjuvant influenced cellular responses and lung pathology. When exposed to RSV, mice immunized with pre-fusion F protein alone (PreF) exhibited increased airway eosinophilia and mucus accumulation. This was further exacerbated by formulation of PreF with Alum (aluminum hydroxide). Conversely, formulation of PreF with a Th1/Th2-balanced adjuvant, Advax-SM, not only suppressed RSV viral replication, but also inhibited airway eosinophilia and mucus accumulation. This was associated with lower numbers of lung innate lymphocyte cells (ILC2s) and CD4+ T cells producing IL-5+ or IL-13+ and increased IFNγ+ CD4+ and CD8+ T cells, in addition to RSV F-specific CD8+ T cells. These data suggest that in the absence of preimmunity, stabilized PreF antigens may still be associated with aberrant Th2 responses that induce lung pathology in response to RSV infection, and can be prevented by formulation with more Th1/Th2-balanced adjuvants that enhance CD4+ and CD8+ IFNγ+ T cell responses. This may support the use of stabilized PreF antigens with Th1/Th2-balanced adjuvants like, Advax-SM, as safer alternatives to alum in RSV vaccine candidates.


Subject(s)
Adjuvants, Immunologic/pharmacology , Aluminum Hydroxide/pharmacology , Lung/drug effects , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/pharmacology , Respiratory Syncytial Viruses/drug effects , Th2 Cells/drug effects , Viral Fusion Proteins/pharmacology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Immunity, Humoral/drug effects , Immunization , Immunogenicity, Vaccine/drug effects , Lung/immunology , Lung/pathology , Lung/virology , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/immunology , Respiratory Syncytial Viruses/pathogenicity , Th1-Th2 Balance/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/virology
19.
Am J Physiol Lung Cell Mol Physiol ; 319(3): L481-L496, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32640839

ABSTRACT

Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infections in children worldwide. While most develop a mild, self-limiting illness, some develop severe acute lower respiratory infection and persistent airway disease. Exposure to ambient particulate matter has been linked to asthma, bronchitis, and viral infection in multiple epidemiological studies. We hypothesized that coexposure to nanoparticles worsens RSV-induced airway epithelial barrier dysfunction. Bronchial epithelial cells were incubated with titanium dioxide nanoparticles (TiO2-NP) or a combination of TiO2-NP and RSV. Structure and function of epithelial cell barrier were analyzed. Viral titer and the role of reactive oxygen species (ROS) generation were evaluated. In vivo, mice were intranasally incubated with TiO2-NP, RSV, or a combination. Lungs and bronchoalveolar lavage (BAL) fluid were harvested for analysis of airway inflammation and apical junctional complex (AJC) disruption. RSV-induced AJC disruption was amplified by TiO2-NP. Nanoparticle exposure increased viral infection in epithelial cells. TiO2-NP induced generation of ROS, and pretreatment with antioxidant, N-acetylcysteine, reversed said barrier dysfunction. In vivo, RSV-induced injury and AJC disruption were augmented in the lungs of mice given TiO2-NP. Airway inflammation was exacerbated, as evidenced by increased white blood cell infiltration into the BAL, along with exaggeration of peribronchial inflammation and AJC disruption. These data demonstrate that TiO2-NP exposure exacerbates RSV-induced AJC dysfunction and increases inflammation by mechanisms involving generation of ROS. Further studies are required to determine whether NP exposure plays a role in the health disparities of asthma and other lung diseases, and why some children experience more severe airway disease with RSV infection.


Subject(s)
Epithelial Cells/drug effects , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Viruses/pathogenicity , Respiratory Tract Infections/drug therapy , Titanium/pharmacology , Animals , Asthma/drug therapy , Asthma/etiology , Bronchi/drug effects , Bronchi/virology , Bronchoalveolar Lavage Fluid/cytology , Epithelial Cells/virology , Inflammation/complications , Inflammation/drug therapy , Lung/drug effects , Lung/virology , Mice , Respiratory Syncytial Viruses/drug effects
20.
Nature ; 583(7817): 615-619, 2020 07.
Article in English | MEDLINE | ID: mdl-32494007

ABSTRACT

Pneumonia resulting from infection is one of the leading causes of death worldwide. Pulmonary infection by the respiratory syncytial virus (RSV) is a large burden on human health, for which there are few therapeutic options1. RSV targets ciliated epithelial cells in the airways, but how viruses such as RSV interact with receptors on these cells is not understood. Nucleolin is an entry coreceptor for RSV2 and also mediates the cellular entry of influenza, the parainfluenza virus, some enteroviruses and the bacterium that causes tularaemia3,4. Here we show a mechanism of RSV entry into cells in which outside-in signalling, involving binding of the prefusion RSV-F glycoprotein with the insulin-like growth factor-1 receptor, triggers the activation of protein kinase C zeta (PKCζ). This cellular signalling cascade recruits nucleolin from the nuclei of cells to the plasma membrane, where it also binds to RSV-F on virions. We find that inhibiting PKCζ activation prevents the trafficking of nucleolin to RSV particles on airway organoid cultures, and reduces viral replication and pathology in RSV-infected mice. These findings reveal a mechanism of virus entry in which receptor engagement and signal transduction bring the coreceptor to viral particles at the cell surface, and could form the basis of new therapeutics to treat RSV infection.


Subject(s)
Receptor, IGF Type 1/metabolism , Receptors, Virus/metabolism , Respiratory Syncytial Viruses/metabolism , Virus Internalization , Cell Line , Cell Nucleus/metabolism , Enzyme Activation , Humans , Membrane Fusion/drug effects , Phosphoproteins/metabolism , Protein Binding , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , RNA-Binding Proteins/metabolism , Receptor, IGF Type 1/antagonists & inhibitors , Respiratory Syncytial Viruses/drug effects , Respiratory Syncytial Viruses/pathogenicity , Respiratory Syncytial Viruses/physiology , Viral Load/drug effects , Virus Internalization/drug effects , Nucleolin
SELECTION OF CITATIONS
SEARCH DETAIL
...