Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Molecules ; 27(4)2022 Feb 09.
Artículo en Inglés | MEDLINE | ID: covidwho-1715566

RESUMEN

Betulinic acid (BA) and its derivatives exhibit a variety of biological activities, especially their anti-HIV-1 activity, but generally have only modest inhibitory potency against influenza virus. The entry of influenza virus into host cells can be competitively inhibited by multivalent derivatives targeting hemagglutinin. In this study, a series of hexa-, hepta- and octavalent BA derivatives based on α-, ß- and γ-cyclodextrin scaffolds, respectively, with varying lengths of flexible oligo(ethylene glycol) linkers was designed and synthesized using a microwave-assisted copper-catalyzed 1,3-dipolar cycloaddition reaction. The generated BA-cyclodextrin conjugates were tested for their in vitro activity against influenza A/WSN/33 (H1N1) virus and cytotoxicity. Among the tested compounds, 58, 80 and 82 showed slight cytotoxicity to Madin-Darby canine kidney cells with viabilities ranging from 64 to 68% at a high concentration of 100 µM. Four conjugates 51 and 69-71 showed significant inhibitory effects on influenza infection with half maximal inhibitory concentration values of 5.20, 9.82, 7.48 and 7.59 µM, respectively. The structure-activity relationships of multivalent BA-cyclodextrin conjugates were discussed, highlighting that multivalent BA derivatives may be potential antiviral agents against influenza infection.


Asunto(s)
Antivirales , Ciclodextrinas/química , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Triterpenos Pentacíclicos/química , Animales , Antivirales/síntesis química , Antivirales/química , Antivirales/farmacología , Perros , Evaluación Preclínica de Medicamentos , Células de Riñón Canino Madin Darby , Infecciones por Orthomyxoviridae/metabolismo , Relación Estructura-Actividad , Ácido Betulínico
2.
Sci Adv ; 6(48)2020 11.
Artículo en Inglés | MEDLINE | ID: covidwho-1388431

RESUMEN

Acute respiratory distress syndrome is associated with a robust inflammatory response that damages the vascular endothelium, impairing gas exchange. While restoration of microcapillaries is critical to avoid mortality, therapeutic targeting of this process requires a greater understanding of endothelial repair mechanisms. Here, we demonstrate that lung endothelium possesses substantial regenerative capacity and lineage tracing reveals that native endothelium is the source of vascular repair after influenza injury. Ablation of chicken ovalbumin upstream promoter-transcription factor 2 (COUP-TF2) (Nr2f2), a transcription factor implicated in developmental angiogenesis, reduced endothelial proliferation, exacerbating viral lung injury in vivo. In vitro, COUP-TF2 regulates proliferation and migration through activation of cyclin D1 and neuropilin 1. Upon influenza injury, nuclear factor κB suppresses COUP-TF2, but surviving endothelial cells ultimately reestablish vascular homeostasis dependent on restoration of COUP-TF2. Therefore, stabilization of COUP-TF2 may represent a therapeutic strategy to enhance recovery from pathogens, including H1N1 influenza and SARS-CoV-2.


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Subtipo H1N1 del Virus de la Influenza A , Pulmón/citología , Pulmón/fisiología , Infecciones por Orthomyxoviridae/metabolismo , Regeneración/genética , Animales , Factor de Transcripción COUP II/genética , Movimiento Celular/genética , Proliferación Celular/genética , Modelos Animales de Enfermedad , Femenino , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Masculino , Ratones , Ratones Transgénicos , Infecciones por Orthomyxoviridae/virología , Transfección
3.
J Virol ; 95(14): e0011121, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: covidwho-1358015

RESUMEN

The current fears of a future influenza pandemic have resulted in an increased emphasis on the development and testing of novel therapeutic strategies against the virus. Fundamental to this is the ferret model of influenza infection, which is critical in examining pathogenesis and treatment. Nevertheless, a precise evaluation of the efficacy of any treatment strategy in ferrets is reliant on understanding the immune response in this model. Interferon-inducible transmembrane proteins (IFITMs) are interferon-stimulated proteins shown to be critically important in the host immune response against viral infections. These proteins confer intrinsic innate immunity to pH-dependent viruses such as influenza viruses and can inhibit cytosolic entry of such viruses to limit the severity of infection following interferon upregulation. Mutations in IFITM genes in humans have been identified as key risk factors for worsened disease progression, particularly in the case of avian influenza viruses such as H7N9. While the IFITM genes of humans and mice have been well characterized, no studies have been conducted to classify the IFITM locus and interferon-driven upregulation of IFITMs in ferrets. Here, we show the architecture of the ferret IFITM locus and its synteny to the IFITM locus of other mammalian and avian species. Furthermore, we show that ferret IFITM1, -2, and -3 are functionally responsive to both interferon-α (IFN-α) and influenza virus stimulation. Thus, we show that ferret IFITMs exhibit interferon-stimulated properties similar to those shown in other species, furthering our knowledge of the innate immune response in the ferret model of human influenza virus infections. IMPORTANCE IFITM proteins can prevent the entry of several pH-dependent viruses, including high-consequence viruses such as HIV, influenza viruses, and SARS-coronaviruses. Mutations in these genes have been associated with worsened disease outcomes with mutations in their IFITM genes, highlighting these genes as potential disease risk factors. Ferrets provide a valuable tool to model infectious diseases; however, there is a critical shortage of information regarding their interferon-stimulated genes. We identified the putative ferret IFITM genes and mapped their complete gene locus. Thus, our study fills a critical gap in knowledge and supports the further use of the ferret model to explore the importance of IFITMs in these important diseases.


Asunto(s)
Hurones , Subtipo H1N1 del Virus de la Influenza A , Interferón-alfa/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Animales , Línea Celular , Secuencia Conservada , Modelos Animales de Enfermedad , Hurones/inmunología , Hurones/metabolismo , Hurones/virología , Humanos , Modelos Moleculares , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/metabolismo , Reacción en Cadena de la Polimerasa , Análisis de Secuencia de Proteína , Regulación hacia Arriba
4.
Am J Respir Cell Mol Biol ; 64(6): 677-686, 2021 06.
Artículo en Inglés | MEDLINE | ID: covidwho-1259048

RESUMEN

There is an urgent need for new drugs for patients with acute respiratory distress syndrome (ARDS), including those with coronavirus disease (COVID-19). ARDS in influenza-infected mice is associated with reduced concentrations of liponucleotides (essential precursors for de novo phospholipid synthesis) in alveolar type II (ATII) epithelial cells. Because surfactant phospholipid synthesis is a primary function of ATII cells, we hypothesized that disrupting this process could contribute significantly to the pathogenesis of influenza-induced ARDS. The goal of this study was to determine whether parenteral liponucleotide supplementation can attenuate ARDS. C57BL/6 mice inoculated intranasally with 10,000 plaque-forming units/mouse of H1N1 influenza A/WSN/33 virus were treated with CDP (cytidine 5'-diphospho)-choline (100 µg/mouse i.p.) ± CDP -diacylglycerol 16:0/16:0 (10 µg/mouse i.p.) once daily from 1 to 5 days after inoculation (to model postexposure influenza prophylaxis) or as a single dose on Day 5 (to model treatment of patients with ongoing influenza-induced ARDS). Daily postexposure prophylaxis with CDP-choline attenuated influenza-induced hypoxemia, pulmonary edema, alterations in lung mechanics, impairment of alveolar fluid clearance, and pulmonary inflammation without altering viral replication. These effects were not recapitulated by the daily administration of CTP (cytidine triphosphate) and/or choline. Daily coadministration of CDP-diacylglycerol significantly enhanced the beneficial effects of CDP-choline and also modified the ATII cell lipidome, reversing the infection-induced decrease in phosphatidylcholine and increasing concentrations of most other lipid classes in ATII cells. Single-dose treatment with both liponucleotides at 5 days after inoculation also attenuated hypoxemia, altered lung mechanics, and inflammation. Overall, our data show that liponucleotides act rapidly to reduce disease severity in mice with severe influenza-induced ARDS.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Citidina Difosfato Colina/farmacología , Citidina Difosfato Diglicéridos/farmacología , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/prevención & control , Células Epiteliales Alveolares/patología , Células Epiteliales Alveolares/virología , Animales , COVID-19/patología , Ratones , Infecciones por Orthomyxoviridae/complicaciones , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/patología , Síndrome de Dificultad Respiratoria/etiología , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/patología , SARS-CoV-2/metabolismo , Tratamiento Farmacológico de COVID-19
5.
Biochem Biophys Res Commun ; 556: 87-92, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: covidwho-1173393

RESUMEN

Virus-induced cytokine storm has been a devastating actuality in clinic. The abnormal production of type I interferon (IFN-1) and upregulation of multiple cytokines induced strong inflammation and thus lead to shock and organ failure. As an E3 ubiquitin ligase, tripartite motif-containing 37 (TRIM37) regulates the ubiquitination of multiple proteins including TRAFs. RNA sequencing was performed to investigated the alteration of transcriptional profile of H1N1-infected patients. qRT-PCR assay was performed to investigate the RNA levels of certain genes. The group of immune cells was examined by the Flow cytometry analysis. H&E staining was applied to evaluate lung inflammation of WT and TRIM37-KO mice. ELISA assay was performed to demonstrate the alteration of multiple cytokines. The protein levels in NF-kB signaling was estimated by western blotting and immunoprecipitation assays were applied to demonstrate the direct interaction between TRIM37 and TRAF-6. The RNA level of TRIM37 decreased in CD11b+ cells of Flu-infected patients. Knockout of TRIM37 inhibited the immune responses of H1N1-infected mice. TRIM37 deficiency reduced the levels of virous proinflammatory cytokines in bone marrow derived macrophages (BMDMs). Mechanically, TRIM37 promoted the K63-linked ubiquitination of TRAF6. TRIM37 negatively regulated inflammatory responses induced by virus infection via promoting TRAF6 ubiquitination at K63.


Asunto(s)
Inflamación/metabolismo , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Factor 6 Asociado a Receptor de TNF/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Animales , Femenino , Humanos , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/genética , Gripe Humana/inmunología , Gripe Humana/metabolismo , Gripe Humana/virología , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología , Factor 6 Asociado a Receptor de TNF/química , Proteínas de Motivos Tripartitos/deficiencia , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética
6.
J Ethnopharmacol ; 275: 114063, 2021 Jul 15.
Artículo en Inglés | MEDLINE | ID: covidwho-1164034

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Fufang-Yinhua-Jiedu Granules (FFYH) optimized from a Yin-Qiao-San, as traditional Chinese medicine (TCM), was used to treat influenza and upper respiratory tract infection and was recommended for the prevention and treatment of SARS in 2003 and current COVID-19 in Anhui Province in 2020. AIM OF STUDY: In the clinical studies, FFYH was very effective for the treatment of influenza, but the mechanism of action against influenza A virus remains unclear. In the present study, we investigated the antiviral effect of FFYH against influenza A virus in vitro and vivo. Moreover, the potential mechanism of FFYH against influenza A virus in vivo was investigated for the first time. MATERIALS AND METHODS: CPE inhibition assay and HA assay were used to evaluate the in vitro antiviral effects of FFYH against influenza A virus H1N1, H3N2, H5N1, H7N9 and H9N2. Mice were used to evaluate the antiviral effect of FFYH in vivo with ribavirin and lianhuaqingwen as positive controls. RT-PCR was used to quantify the mRNA transcription of TNF-α, IL-6, IFN-γ, IP10, and IL-1ß mRNA. ELISA was used to examine the expression of inflammatory factors such as TNF-α, IL-6, IFN-γ, IP10, and IL-1ß in sera. The blood parameters were analyzed with auto hematology analyzer. Moreover, the potential mechanism of FFYH against influenza A virus in vivo was also investigated. RESULTS: FFYH showed a broad-spectrum of antiviral activity against H1N1, H3N2, H5N1, H7N9, and H9N2 influenza A viruses. Furthermore, FFYH dose-dependently increased the survival rate, significantly prolonged the median survival time of mice, and markedly reduced lung injury caused by influenza A virus. Also, FFYH significantly improve the sick signs, food taken, weight loss, blood parameters, lung index, and lung pathological changes. Moreover, FFYH could markedly inhibit the inflammatory cytokine expression of TNF-α, IL-6, IFN-γ, IP10, IL-10, and IL-1ß mRNA or protein via inhibition of the TLR7/MyD88/NF-κB signaling pathway in vivo. CONCLUSION: FFYH not only showed a broad-spectrum of anti-influenza virus activity in vitro, but also exhibited a significant protective effect against lethal influenza virus infection in vivo. Furthermore, our results indicated that the in vivo antiviral effect of FFYH against influenza virus may be attributed to suppressing the expression of inflammatory cytokines via regulating the TLR7/MyD88/NF-κB signaling pathway. These findings provide evidence for the clinical treatment of influenza A virus infection with FFYH.


Asunto(s)
Antiinflamatorios/farmacología , Antivirales/farmacología , Medicamentos Herbarios Chinos/farmacología , Virus de la Influenza A/efectos de los fármacos , Pulmón/efectos de los fármacos , Glicoproteínas de Membrana/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Receptor Toll-Like 7/metabolismo , Células A549 , Animales , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Perros , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/metabolismo , Virus de la Influenza A/patogenicidad , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/virología , Células de Riñón Canino Madin Darby , Ratones Endogámicos ICR , FN-kappa B/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Transducción de Señal , Replicación Viral/efectos de los fármacos
7.
J Infect Dis ; 224(5): 821-830, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1006333

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
8.
Sci Immunol ; 5(53)2020 11 06.
Artículo en Inglés | MEDLINE | ID: covidwho-999190

RESUMEN

Lower respiratory viral infections, such as influenza virus and severe acute respiratory syndrome coronavirus 2 infections, often cause severe viral pneumonia in aged individuals. Here, we report that influenza viral pneumonia leads to chronic nonresolving lung pathology and exacerbated accumulation of CD8+ tissue-resident memory T cells (TRM) in the respiratory tract of aged hosts. TRM cell accumulation relies on elevated TGF-ß present in aged tissues. Further, we show that TRM cells isolated from aged lungs lack a subpopulation characterized by expression of molecules involved in TCR signaling and effector function. Consequently, TRM cells from aged lungs were insufficient to provide heterologous protective immunity. The depletion of CD8+ TRM cells dampens persistent chronic lung inflammation and ameliorates tissue fibrosis in aged, but not young, animals. Collectively, our data demonstrate that age-associated TRM cell malfunction supports chronic lung inflammatory and fibrotic sequelae after viral pneumonia.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , COVID-19/inmunología , Memoria Inmunológica/inmunología , Pulmón/inmunología , Neumonía Viral/inmunología , SARS-CoV-2/inmunología , Factores de Edad , Animales , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/virología , COVID-19/metabolismo , COVID-19/virología , Interacciones Huésped-Patógeno/inmunología , Humanos , Gripe Humana/inmunología , Gripe Humana/metabolismo , Gripe Humana/virología , Pulmón/metabolismo , Pulmón/virología , Ratones Endogámicos C57BL , Orthomyxoviridae/inmunología , Orthomyxoviridae/fisiología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Pandemias , Neumonía Viral/metabolismo , Neumonía Viral/virología , SARS-CoV-2/fisiología , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/metabolismo
9.
Blood Adv ; 4(13): 2967-2978, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: covidwho-625455

RESUMEN

Thrombocytopenia is a common complication of influenza virus infection, and its severity predicts the clinical outcome of critically ill patients. The underlying cause(s) remain incompletely understood. In this study, in patients with an influenza A/H1N1 virus infection, viral load and platelet count correlated inversely during the acute infection phase. We confirmed this finding in a ferret model of influenza virus infection. In these animals, platelet count decreased with the degree of virus pathogenicity varying from 0% in animals infected with the influenza A/H3N2 virus, to 22% in those with the pandemic influenza A/H1N1 virus, up to 62% in animals with a highly pathogenic A/H5N1 virus infection. This thrombocytopenia is associated with virus-containing platelets that circulate in the blood. Uptake of influenza virus particles by platelets requires binding to sialoglycans and results in the removal of sialic acids by the virus neuraminidase, a trigger for hepatic clearance of platelets. We propose the clearance of influenza virus by platelets as a paradigm. These insights clarify the pathophysiology of influenza virus infection and show how severe respiratory infections, including COVID-19, may propagate thrombocytopenia and/or thromboembolic complications.


Asunto(s)
Plaquetas/virología , Virus de la Influenza A/patogenicidad , Gripe Humana/complicaciones , Ácido N-Acetilneuramínico/metabolismo , Polisacáridos/metabolismo , Trombocitopenia/etiología , Animales , Plaquetas/metabolismo , Plaquetas/patología , Modelos Animales de Enfermedad , Hurones , Interacciones Huésped-Patógeno , Humanos , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H3N2 del Virus de la Influenza A/patogenicidad , Subtipo H3N2 del Virus de la Influenza A/fisiología , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Subtipo H5N1 del Virus de la Influenza A/fisiología , Virus de la Influenza A/fisiología , Gripe Humana/metabolismo , Gripe Humana/patología , Gripe Humana/virología , Infecciones por Orthomyxoviridae/complicaciones , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Trombocitopenia/metabolismo , Trombocitopenia/patología , Trombocitopenia/virología , Internalización del Virus
10.
Nat Commun ; 11(1): 4252, 2020 08 25.
Artículo en Inglés | MEDLINE | ID: covidwho-741685

RESUMEN

The 2019 novel respiratory virus (SARS-CoV-2) causes COVID-19 with rapid global socioeconomic disruptions and disease burden to healthcare. The COVID-19 and previous emerging virus outbreaks highlight the urgent need for broad-spectrum antivirals. Here, we show that a defensin-like peptide P9R exhibited potent antiviral activity against pH-dependent viruses that require endosomal acidification for virus infection, including the enveloped pandemic A(H1N1)pdm09 virus, avian influenza A(H7N9) virus, coronaviruses (SARS-CoV-2, MERS-CoV and SARS-CoV), and the non-enveloped rhinovirus. P9R can significantly protect mice from lethal challenge by A(H1N1)pdm09 virus and shows low possibility to cause drug-resistant virus. Mechanistic studies indicate that the antiviral activity of P9R depends on the direct binding to viruses and the inhibition of virus-host endosomal acidification, which provides a proof of concept that virus-binding alkaline peptides can broadly inhibit pH-dependent viruses. These results suggest that the dual-functional virus- and host-targeting P9R can be a promising candidate for combating pH-dependent respiratory viruses.


Asunto(s)
Antivirales/farmacología , Coronavirus/efectos de los fármacos , Virus de la Influenza A/efectos de los fármacos , Péptidos/farmacología , Secuencia de Aminoácidos , Animales , Antivirales/química , Antivirales/metabolismo , Antivirales/uso terapéutico , Línea Celular , Endosomas/química , Endosomas/efectos de los fármacos , Femenino , Humanos , Concentración de Iones de Hidrógeno , Virus de la Influenza A/metabolismo , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/metabolismo , Péptidos/química , Péptidos/metabolismo , Péptidos/uso terapéutico , Unión Proteica , Conformación Proteica , Rhinovirus/efectos de los fármacos , Rhinovirus/metabolismo , Carga Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
11.
J Breath Res ; 14(4): 041001, 2020 07 21.
Artículo en Inglés | MEDLINE | ID: covidwho-682126

RESUMEN

The COVID-19 pandemic has highlighted the importance of rapid, cost effective, accurate, and non-invasive testing for viral infections. Volatile compounds (VCs) have been suggested for several decades as fulfilling these criteria. However currently very little work has been done in trying to diagnose viral infections using VCs. Much of the work carried out to date involves the differentiation of bacterial and viral sources of infection and often the detection of bacterial and viral co-infection. However, this has usually been done in vitro and very little work has involved the use of human participants. Viruses hijack the host cell metabolism and do not produce their own metabolites so identifying virus specific VCs is at best a challenging task. However, there are proteins and lipids that are potential candidates as markers of viral infection. The current understanding is that host cell glycolysis is upregulated under viral infection to increase the available energy for viral replication. There is some evidence that viral infection leads to the increase of production of fatty acids, alkanes, and alkanes related products. For instance, 2,3-butandione, aldehydes, 2,8-dimethyl-undecane and n-propyl acetate have all been correlated with viral infection. Currently, the literature points to markers of oxidative stress (e.g. nitric oxide, aldehydes etc) being the most useful in the determination of viral infection. The issue, however, is that there are also many other conditions that can lead to oxidative stress markers being produced. In this review a range of (mainly mass spectrometric) methods are discussed for viral detection in breath, including breath condensate. Currently MALDI-ToF-MS is likely to be the preferred method for the identification of viral strains and variants of those strains, however it is limited by its need for the viral strains to have been sequenced and logged in a database.


Asunto(s)
Pruebas Respiratorias/métodos , Virosis/diagnóstico , Aldehídos/metabolismo , Animales , Betacoronavirus , Biomarcadores/metabolismo , COVID-19 , Prueba de COVID-19 , Técnicas de Laboratorio Clínico , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/metabolismo , Cromatografía de Gases y Espectrometría de Masas , Hepatitis B/diagnóstico , Hepatitis B/metabolismo , Humanos , Gripe Humana/diagnóstico , Gripe Humana/metabolismo , Espectrometría de Masas , Óxido Nítrico/metabolismo , Infecciones por Orthomyxoviridae/diagnóstico , Infecciones por Orthomyxoviridae/metabolismo , Estrés Oxidativo , Pandemias , Infecciones por Picornaviridae/diagnóstico , Infecciones por Picornaviridae/metabolismo , Neumonía Viral/diagnóstico , Neumonía Viral/metabolismo , Infecciones por Rotavirus/diagnóstico , Infecciones por Rotavirus/metabolismo , SARS-CoV-2 , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Porcinos , Virosis/metabolismo , Virus
12.
Nitric Oxide ; 102: 39-41, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: covidwho-600932

RESUMEN

COVID-19 is a severe pandemic which has caused a devastating amount of loss in lives around the world, and yet we still don't know how to appropriately treat this disease. We know very little about the pathogenesis of SARS-CoV-2, the virus which induces the COVID-19. However, COVID-19 does share many similar symptoms with SARS and influenza. Previous scientific discoveries learned from lab animal models and clinical practices shed light on possible pathogenic mechanisms in COVID-19. In the past decades, accumulated scientific findings confirmed the pathogenic role of free radicals damage in respiratory virus infection. Astonishingly very few medical professionals mention the crucial role of free radical damage in COVID-19. This hypothesis aims to summarize the crucial pathogenic role of free radical damage in respiratory virus induced pneumonia and suggest an antioxidative therapeutic strategy for COVID-19.


Asunto(s)
Betacoronavirus , Infecciones por Coronavirus/metabolismo , Radicales Libres/metabolismo , Pandemias , Neumonía Viral/metabolismo , Acetilcisteína/uso terapéutico , Animales , Antioxidantes/uso terapéutico , Ácido Ascórbico/uso terapéutico , Azitromicina/administración & dosificación , Azitromicina/farmacología , Azitromicina/uso terapéutico , COVID-19 , Ensayos Clínicos como Asunto , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/etiología , Síndrome de Liberación de Citoquinas/metabolismo , Quimioterapia Combinada , Radicales Libres/antagonistas & inhibidores , Glutatión/uso terapéutico , Humanos , Hidroxicloroquina/administración & dosificación , Hidroxicloroquina/farmacología , Hidroxicloroquina/uso terapéutico , Ratones , Insuficiencia Multiorgánica/etiología , Insuficiencia Multiorgánica/prevención & control , Factor 2 Relacionado con NF-E2/agonistas , Óxido Nítrico/metabolismo , Infecciones por Orthomyxoviridae/metabolismo , Estrés Oxidativo , Neumonía Viral/complicaciones , Neumonía Viral/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , SARS-CoV-2 , Síndrome Respiratorio Agudo Grave/tratamiento farmacológico , Síndrome Respiratorio Agudo Grave/metabolismo , Tratamiento Farmacológico de COVID-19
13.
Science ; 369(6504): 712-717, 2020 08 07.
Artículo en Inglés | MEDLINE | ID: covidwho-594812

RESUMEN

Excessive cytokine signaling frequently exacerbates lung tissue damage during respiratory viral infection. Type I (IFN-α and IFN-ß) and III (IFN-λ) interferons are host-produced antiviral cytokines. Prolonged IFN-α and IFN-ß responses can lead to harmful proinflammatory effects, whereas IFN-λ mainly signals in epithelia, thereby inducing localized antiviral immunity. In this work, we show that IFN signaling interferes with lung repair during influenza recovery in mice, with IFN-λ driving these effects most potently. IFN-induced protein p53 directly reduces epithelial proliferation and differentiation, which increases disease severity and susceptibility to bacterial superinfections. Thus, excessive or prolonged IFN production aggravates viral infection by impairing lung epithelial regeneration. Timing and duration are therefore critical parameters of endogenous IFN action and should be considered carefully for IFN therapeutic strategies against viral infections such as influenza and coronavirus disease 2019 (COVID-19).


Asunto(s)
Células Epiteliales Alveolares/patología , Citocinas/metabolismo , Interferón Tipo I/metabolismo , Interferones/metabolismo , Pulmón/patología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Células Epiteliales Alveolares/inmunología , Animales , Apoptosis , Líquido del Lavado Bronquioalveolar/inmunología , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Citocinas/administración & dosificación , Citocinas/inmunología , Femenino , Subtipo H3N2 del Virus de la Influenza A , Interferón Tipo I/administración & dosificación , Interferón Tipo I/farmacología , Interferón-alfa/administración & dosificación , Interferón-alfa/metabolismo , Interferón-alfa/farmacología , Interferón beta/administración & dosificación , Interferón beta/metabolismo , Interferón beta/farmacología , Interferones/administración & dosificación , Interferones/farmacología , Masculino , Ratones , Infecciones por Orthomyxoviridae/metabolismo , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Interferón lambda
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA