Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 178
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
ACS Infect Dis ; 10(5): 1552-1560, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38623820

RESUMEN

Tyrosine cross-linking has recently been used to produce nanoclusters (NCs) from peptides to enhance their immunogenicity. In this study, NCs were generated using the ectodomain of the ion channel Matrix 2 (M2e) protein, a conserved influenza surface antigen. The NCs were administered via intranasal (IN) or intramuscular (IM) routes in a mouse model in a prime-boost regimen in the presence of the adjuvant CpG. After boost, a significant increase in anti-M2e IgG and its subtypes was observed in the serum and lungs of mice vaccinated through the IM and IN routes; however, significant enhancement in anti-M2e IgA in lungs was observed only in the IN group. Analysis of cytokine concentrations in stimulated splenocyte cultures indicated a Th1/Th17-biased response. Mice were challenged with a lethal dose of A/California/07/2009 (H1N1pdm), A/Puerto Rico/08/1934 (H1N1), or A/Hong Kong/08/1968 (H3N2) strains. Mice that received M2e NCs + CpG were significantly protected against these strains and showed decreased lung viral titers compared with the naive mice and M2e NC-alone groups. The IN-vaccinated group showed superior protection against the H3N2 strain as compared to the IM group. This research extends our earlier efforts involving the tyrosine-based cross-linking method and highlights the potential of this technology in enhancing the immunogenicity of short peptide immunogens.


Asunto(s)
Anticuerpos Antivirales , Subtipo H1N1 del Virus de la Influenza A , Vacunas contra la Influenza , Infecciones por Orthomyxoviridae , Tirosina , Animales , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Ratones , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/inmunología , Tirosina/química , Tirosina/farmacología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Femenino , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Proteínas de la Matriz Viral/inmunología , Proteínas de la Matriz Viral/genética , Ratones Endogámicos BALB C , Subtipo H3N2 del Virus de la Influenza A/inmunología , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/administración & dosificación , Pulmón/virología , Pulmón/inmunología , Administración Intranasal , Inyecciones Intramusculares , Citocinas , Protección Cruzada , Proteínas Viroporinas
2.
Phytomedicine ; 128: 155446, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38518643

RESUMEN

BACKGROUND: Influenza viral pneumonia is a common complication after influenza virus infection. Xijiao Dihuang Decoction combined with Yinqiao Powder (XDY) is effective on improving influenza viral pneumonia. PURPOSE: This study further explores the anti-inflammatory mechanism of XDY in the treatment of influenza viral pneumonia. STUDY DESIGN: The effects of XDY on inflammation, autophagy, NACHT-LRR-PYD-containing protein 3 (NLRP3) inflammasome and pyroptosis were assessed in the mice with influenza viral pneumonia. In addition, the mouse macrophage cell line (J774A.1) infected with influenza virus was adopted to decode the in vitro effects of XDY on autophagy, reactive oxygen species (ROS), NLRP3 inflammasome and pyroptosis. We analyzed the XDY-induced autophagy, especially the mitophagy-related ROS clearance, and the subsequent inhibition of ROS/NLRP3 inflammasome/pyroptosis signaling in the infected macrophages by different assays based on quantitative polymerase chain reaction, western blot, flow cytometry, immunofluorescence and enzyme-linked immunosorbent assay. RESULTS: In vivo, XDY could effectively improve the lung inflammatory response in the mice with influenza virus pneumonia, due to an intact autophagy flux-promoting effect and the inhibiting roles on NLRP3 inflammasome and pyroptosis. Notably, in vitro, compared with the infected macrophages treated by the NLRP3 inflammasome agonist (Monosodium urate) or the mitochondrial-targeted antioxidant agent, the XDY-dependent treating could inhibit pyroptosis by negatively regulating the signaling axis of ROS/NLRP3 inflammasome/pyroptosis in the influenza virus-infected macrophages. More interestingly, XDY could promote an intact autophagy flux, inducing mitophagy eliminating the damaged mitochondria to reduce the intracellular ROS accumulation, and thus decrease the oxidative stress in the infected macrophages. Especially, the inhibitor of autophagy inition, 3-Methyladenine, could reverse the inhibitory effect of XDY on ROS-NLRP3 inflammasome-mediated pyroptosis, indicating an XDY-promoted mitophagy-dependent ROS scavenging. CONCLUSION: XDY can promote an intact autophagy flux to eliminate damaged mitochondria, namely mitophagy, which reduces the intracellular ROS accumulation contributing to NLRP3 inflammasome activation, restricting pyroptosis and eventually alleviating the influenza virus-induced inflammatory lesions. The obtained results provide new insights into the mechanism of action of XDY in alleviating influenza virus pneumonia, especially the roles of XDY in anti-oxidation, anti-inflammation and anti-pyroptosis, with potential therapeutic targets for future application in integrative medicine.


Asunto(s)
Autofagia , Medicamentos Herbarios Chinos , Proteína con Dominio Pirina 3 de la Familia NLR , Piroptosis , Especies Reactivas de Oxígeno , Animales , Medicamentos Herbarios Chinos/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Ratones , Autofagia/efectos de los fármacos , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Línea Celular , Ratones Endogámicos C57BL , Masculino , Pulmón/efectos de los fármacos , Pulmón/virología
3.
BMC Complement Med Ther ; 22(1): 164, 2022 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-35733131

RESUMEN

BACKGROUND: Influenza A virus infection due to drug resistance and side effects of the conventional antiviral drugs yet remains a serious public health threat for humans and animals. Forsythiaside A is an effective ingredient isolated from the Chinese herbal medicine forsythia. It has various pharmacological effects and has a good therapeutic effect against a variety of infectious diseases. This study aimed to further explore the immunological mechanism of Forsythiaside A in the treatment of influenza virus-infected mice and its effect on the Toll-like receptor 7 (TLR7) signaling pathway in the lungs of these mice. METHODS: C57/BL6J mice and TLR7-/- mice were infected with the FM1 strains (H1N1 and A/FM/1/4) of the Influenza A virus. Each group of experimental mice were divided into the mock, virus, oseltamivir, and Forsythiaside A groups. Weight change, lung index change, and the mRNA and protein expression levels of key factors in the TLR7 signaling pathway were detected. Flow cytometry was used to detect the changes in the Th1/Th2 and Th17/Treg ratios. RESULTS: After infection with the Influenza A virus, the weight loss of C57/BL6J mice treated with forsythoside A and oseltamivir decreased, and the pathological tissue sections showed that the inflammatory damage was reduced. The expression levels of the key factors, TLR7, myeloid differentiation factor 88(Myd88), and nuclear factor-kappa B (NF-κB) in the TLR7 signaling pathway were significantly reduced. Flow cytometry showed that Th1/Th2 and Th17/Treg ratios decreased after Forsythiaside A treatment. In the TLR7-/- mice, there was no significant change after Forsythiaside A treatment in the virus group. CONCLUSIONS: Forsythiaside A affects the TLR7 signaling pathway in mouse lung immune cells and reduces the inflammatory response caused by the Influenza A virus FM1 strain in mouse lungs.


Asunto(s)
Glicósidos , Subtipo H1N1 del Virus de la Influenza A , Infecciones por Orthomyxoviridae , Receptor Toll-Like 7 , Animales , Glicósidos/farmacología , Pulmón/virología , Ratones , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Oseltamivir/farmacología , Transducción de Señal , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo
4.
PLoS Pathog ; 18(1): e1010171, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35025963

RESUMEN

The development of physiological models that reproduce SARS-CoV-2 infection in primary human cells will be instrumental to identify host-pathogen interactions and potential therapeutics. Here, using cell suspensions directly from primary human lung tissues (HLT), we have developed a rapid platform for the identification of viral targets and the expression of viral entry factors, as well as for the screening of viral entry inhibitors and anti-inflammatory compounds. The direct use of HLT cells, without long-term cell culture and in vitro differentiation approaches, preserves main immune and structural cell populations, including the most susceptible cell targets for SARS-CoV-2; alveolar type II (AT-II) cells, while maintaining the expression of proteins involved in viral infection, such as ACE2, TMPRSS2, CD147 and AXL. Further, antiviral testing of 39 drug candidates reveals a highly reproducible method, suitable for different SARS-CoV-2 variants, and provides the identification of new compounds missed by conventional systems, such as VeroE6. Using this method, we also show that interferons do not modulate ACE2 expression, and that stimulation of local inflammatory responses can be modulated by different compounds with antiviral activity. Overall, we present a relevant and rapid method for the study of SARS-CoV-2.


Asunto(s)
Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Pulmón/virología , SARS-CoV-2/fisiología , Internalización del Virus , Adulto , Animales , Antivirales/farmacología , COVID-19/inmunología , COVID-19/patología , Células Cultivadas , Chlorocebus aethiops , Evaluación Preclínica de Medicamentos , Drogas en Investigación/farmacología , Drogas en Investigación/uso terapéutico , Células HEK293 , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Inflamación/patología , Inflamación/terapia , Inflamación/virología , Pulmón/patología , SARS-CoV-2/efectos de los fármacos , Células Vero , Internalización del Virus/efectos de los fármacos
5.
Mol Cell Biochem ; 477(1): 225-240, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34655418

RESUMEN

Severe acute respiratory syndrome-coronavirus-2 (COVID-19) virus uses Angiotensin-Converting Enzyme 2 (ACE2) as a gateway for their entry into the human body. The ACE2 with cleaved products have emerged as major contributing factors to multiple physiological functions and pathogenic complications leading to the clinical consequences of the COVID-19 infection Decreased ACE2 expression restricts the viral entry into the human cells and reduces the viral load. COVID-19 infection reduces the ACE2 expression and induces post-COVID-19 complications like pneumonia and lung injury. The modulation of the ACE2-Ang (1-7)-Mas (AAM) axis is also being explored as a modality to treat post-COVID-19 complications. Evidence indicates that specific food components may modulate the AAM axis. The variations in the susceptibility to COVID-19 infection and the post-COVID its complications are being correlated with varied dietary habits. Some of the food substances have emerged to have supportive roles in treating post-COVID-19 complications and are being considered as adjuvants to the COVID-19 therapy. It is possible that some of their active ingredients may emerge as the direct treatment for the COVID-19.


Asunto(s)
Angiotensina I/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/complicaciones , COVID-19/dietoterapia , Fragmentos de Péptidos/metabolismo , Proto-Oncogenes Mas/metabolismo , Enfermedades Cardiovasculares/patología , Enfermedades Cardiovasculares/virología , Proteínas en la Dieta/farmacología , Flavonoides/farmacología , Humanos , Pulmón/patología , Pulmón/virología , Aceites de Plantas/farmacología , Polifenoles/farmacología , Terpenos/farmacología , Internalización del Virus , Vitaminas/farmacología
6.
J Ethnopharmacol ; 285: 114838, 2022 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-34788645

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Keguan-1, a new traditional Chinese medicine (TCM) prescription contained seven Chinese herbs, is developed to treat coronavirus disease 19 (COVID-19). The first internationally registered COVID-19 randomised clinical trial on integrated therapy demonstrated that Keguan-1 significantly reduced the incidence of ARDS and inhibited the severe progression of COVID-19. AIM OF THE STUDY: To investigate the protective mechanism of Keguan-1 on ARDS, a lipopolysaccharide (LPS)-induced acute lung injury (ALI) model was used to simulate the pathological state of ARDS in patients with COVID-19, focusing on its effect and mechanism on ALI. MATERIALS AND METHODS: Mice were challenged with LPS (2 mg/kg) by intratracheal instillation (i.t.) and were orally administered Keguan-1 (low dose, 1.25 g/kg; medium dose, 2.5 g/kg; high dose, 5 g/kg) after 2 h. Bronchoalveolar lavage fluid (BALF) and lung tissue were collected 6 h and 24 h after i.t. administration of LPS. The levels of inflammatory factors tumour necrosis factor alpha (TNF-α), interleukin (IL)-6, IL-1ß, keratinocyte-derived chemokine (KC or mCXCL1), macrophage inflammatory protein 2 (MIP2 or mCXCL2), angiotensin II (Ang II), and endothelial cell junction-associated proteins were analysed using ELISA or western blotting. RESULTS: Keguan-1 improved the survival rate, respiratory condition, and pathological lung injury; decreased the production of proinflammatory factors (TNF-α, IL-6, IL-1ß, KC, and MIP2) in BALF and the number of neutrophils in the lung tissues; and ameliorated inflammatory injury in the lung tissues of the mice with LPS-induced ALI. Keguan-1 also reduced the expression of Ang II and the adhesion molecule ICAM-1; increased tight junction proteins (JAM-1 and claudin-5) and VE-cadherin expression; and alleviated pulmonary vascular endothelial injury in LPS-induced ALI. CONCLUSION: These results demonstrate that Keguan-1 can improve LPS-induced ALI by reducing inflammation and pulmonary vascular endothelial injury, providing scientific support for the clinical treatment of patients with COVID-19. Moreover, it also provides a theoretical basis and technical support for the scientific use of TCMs in emerging infectious diseases.


Asunto(s)
Lesión Pulmonar Aguda , Antivirales/farmacología , Líquido del Lavado Bronquioalveolar , COVID-19 , Medicamentos Herbarios Chinos/farmacología , Pulmón , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Líquido del Lavado Bronquioalveolar/virología , COVID-19/complicaciones , COVID-19/inmunología , COVID-19/virología , Cápsulas , Quimiocina CXCL2/análisis , Coix , Forsythia , Interleucina-1beta/análisis , Interleucina-6/análisis , Lonicera , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Pulmón/virología , Ratones , Mortalidad , Morus , Fragmentos de Péptidos/análisis , Prunus armeniaca , Respiración/efectos de los fármacos , SARS-CoV-2 , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/análisis
7.
Front Immunol ; 12: 704391, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34858393

RESUMEN

Cystic fibrosis (CF) is an autosomal recessive gene disorder that affects tens of thousands of patients worldwide. Individuals with CF often succumb to progressive lung disease and respiratory failure following recurrent infections with bacteria. Viral infections can also damage the lungs and heighten the CF patient's susceptibility to bacterial infections and long-term sequelae. Vitamin A is a key nutrient important for immune health and epithelial cell integrity, but there is currently no consensus as to whether vitamin A should be monitored in CF patients. Here we evaluate previous literature and present results from a CF mouse model, showing that oral vitamin A supplements significantly reduce lung lesions that would otherwise persist for 5-6 weeks post-virus exposure. Based on these results, we encourage continued research and suggest that programs for the routine monitoring and regulation of vitamin A levels may help reduce virus-induced lung pathology in CF patients.


Asunto(s)
Fibrosis Quística/metabolismo , Pulmón/patología , Infecciones por Respirovirus/metabolismo , Virus Sendai/fisiología , Vitamina A/metabolismo , Animales , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Suplementos Dietéticos , Modelos Animales de Enfermedad , Proteínas de Unión a Ácidos Grasos/genética , Humanos , Pulmón/virología , Ratones , Ratones Endogámicos CFTR , Ratones Transgénicos , Regiones Promotoras Genéticas , Vitamina A/administración & dosificación
8.
Indian J Pharmacol ; 53(5): 394-402, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34854410

RESUMEN

The severe acute respiratory syndrome coronavirus 2 is spreading like wildfire with no specific recommended treatment in sight. While some risk factors such as the presence of comorbidities, old age, and ethnicity have been recognized, not a lot is known about who the virus will strike first or impact more. In this hopeless scenario, exploration of time-tested facts about viral infections, in general, seems to be a sound basis to prop further research upon. The fact that immunity and its various determinants (e.g., micronutrients, sleep, and hygiene) have a crucial role to play in the defense against invading organisms, may be a good starting point for commencing research into these as yet undisclosed territories. Herein, the excellent immunomodulatory, antiviral, and anti-inflammatory roles of Vitamin D necessitate thorough investigation, particularly in COVID-19 perspective. This article reviews mechanisms and evidence suggesting the role Vitamin D plays in people infected by the newly identified COVID-19 virus. For this review, we searched the databases of Medline, PubMed, and Embase. We studied several meta-analyses and randomized controlled trials evaluating the role of Vitamin D in influenza and other contagious viral infections. We also reviewed the circumstantial and anecdotal evidence connecting Vitamin D with COVID-19 emerging recently. Consequently, it seems logical to conclude that the immune-enhancing, antiviral, anti-inflammatory, and lung-protective role of Vitamin D can be potentially lifesaving. Hence, Vitamin D deserves exhaustive exploration through rigorously designed and controlled scientific trials. Using Vitamin D as prophylaxis and/or chemotherapeutic treatment of COVID-19 infection is an approach worth considering. In this regard, mass assessment and subsequent supplementation can be tried, especially considering the mechanistic evidence in respiratory infections, low potential for toxicity, and widespread prevalence of the deficiency of Vitamin D affecting many people worldwide.


Asunto(s)
Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Inmunidad/efectos de los fármacos , Agentes Inmunomoduladores/uso terapéutico , Pulmón/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Deficiencia de Vitamina D/tratamiento farmacológico , Vitamina D/uso terapéutico , Vitaminas/uso terapéutico , Animales , Antivirales/efectos adversos , COVID-19/inmunología , COVID-19/fisiopatología , COVID-19/virología , Interacciones Huésped-Patógeno , Humanos , Agentes Inmunomoduladores/efectos adversos , Pulmón/inmunología , Pulmón/fisiopatología , Pulmón/virología , Medición de Riesgo , Factores de Riesgo , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , Resultado del Tratamiento , Vitamina D/efectos adversos , Vitamina D/sangre , Deficiencia de Vitamina D/inmunología , Deficiencia de Vitamina D/fisiopatología , Vitaminas/efectos adversos
9.
J Tradit Chin Med ; 41(6): 982-984, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34939396

RESUMEN

OBJECTIVE: To study the possible role of traditional Chinese medicine (TCM) of Huangqi (Radix Astragali Mongolici), Gancao (Radix Glycyrrhizae), Jinyinhua (Flos Lonicerae), and Lianqiao (Fructus Forsythiae Suspensae) in absorption of lung lesions in Corona Virus Disease 2019 (COVID-19) patients. METHODS: A cohort of COVID-19 cases was recruited. During hospitalization, chest computed tomographic (CT) scan and real time polymerase chain reaction (RT-PCR) test were performed every three days. Comparison was held (Western Medicine, WM vs WM plus TCM) on absorption of lung lesions, time interval from admission to negative test result of RT-PCR (ATN), and medical expense. Multivariate cox regression models were built to identify the possible prognostic factor of delayed absorption of lung lesion. RESULTS: The medical expenditure (1163 ± 379 vs 1137 ± 498, P = 0.863) and ATN (13 ± 4 vs 10 ± 4, P = 0.055) were comparable between cases treated with WM plus TCM and cases only received WM. Multivariate cox regression model showed that cases receiving extra TCM had lower risk of delayed absorption of lung lesions [Hazard ratio = 0.24, 95% confidence Interval (0.06, 0.96), P = 0.043]. CONCLUSION: Compared to WM, the treatment of WM plus TCM facilitates the recovery of pulmonary infiltration on COVID-19 cases without significantly increasing medical expense.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Medicamentos Herbarios Chinos/uso terapéutico , Pulmón/patología , Adulto , Astragalus propinquus , Femenino , Forsythia , Glycyrrhiza , Hospitalización , Humanos , Lonicera , Pulmón/virología , Masculino , Medicina Tradicional China , Persona de Mediana Edad , Extractos Vegetales
10.
Int J Mol Sci ; 22(24)2021 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-34948390

RESUMEN

Since the start of the COVID-19 outbreak, pharmaceutical companies and research groups have focused on the development of vaccines and antiviral drugs against SARS-CoV-2. Here, we apply a drug repurposing strategy to identify drug candidates that are able to block the entrance of the virus into human cells. By combining virtual screening with in vitro pseudovirus assays and antiviral assays in Human Lung Tissue (HLT) cells, we identify entrectinib as a potential antiviral drug.


Asunto(s)
Benzamidas/farmacología , Tratamiento Farmacológico de COVID-19 , Indazoles/farmacología , SARS-CoV-2/efectos de los fármacos , Animales , Antivirales/farmacología , Benzamidas/metabolismo , COVID-19/metabolismo , Línea Celular , Chlorocebus aethiops , Evaluación Preclínica de Medicamentos , Reposicionamiento de Medicamentos/métodos , Humanos , Indazoles/metabolismo , Pulmón/patología , Pulmón/virología , Simulación del Acoplamiento Molecular , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidad , Células Vero , Acoplamiento Viral/efectos de los fármacos
11.
Nat Commun ; 12(1): 6097, 2021 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-34671037

RESUMEN

Effective treatments against Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) are urgently needed. Monoclonal antibodies have shown promising results in patients. Here, we evaluate the in vivo prophylactic and therapeutic effect of COVA1-18, a neutralizing antibody highly potent against the B.1.1.7 isolate. In both prophylactic and therapeutic settings, SARS-CoV-2 remains undetectable in the lungs of treated hACE2 mice. Therapeutic treatment also causes a reduction in viral loads in the lungs of Syrian hamsters. When administered at 10 mg kg-1 one day prior to a high dose SARS-CoV-2 challenge in cynomolgus macaques, COVA1-18 shows very strong antiviral activity in the upper respiratory compartments. Using a mathematical model, we estimate that COVA1-18 reduces viral infectivity by more than 95% in these compartments, preventing lymphopenia and extensive lung lesions. Our findings demonstrate that COVA1-18 has a strong antiviral activity in three preclinical models and could be a valuable candidate for further clinical evaluation.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Neutralizantes/administración & dosificación , Antivirales/administración & dosificación , Tratamiento Farmacológico de COVID-19 , SARS-CoV-2/inmunología , Enzima Convertidora de Angiotensina 2/genética , Animales , Anticuerpos Monoclonales/farmacocinética , Antivirales/farmacocinética , COVID-19/sangre , COVID-19/inmunología , COVID-19/virología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Pulmón/metabolismo , Pulmón/virología , Macaca fascicularis , Masculino , Mesocricetus , Ratones , Ratones Transgénicos , SARS-CoV-2/aislamiento & purificación , Distribución Tisular , Carga Viral
12.
Phytomedicine ; 91: 153704, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34419736

RESUMEN

BACKGROUND: COVID-19 (Coronavirus Disease-2019) has spread widely around the world and impacted human health for millions. The lack of effective targeted drugs and vaccines forces scientific world to search for new effective antiviral therapeutic drugs. It has reported that flavonoids have potential inhibitory activity on SARS-CoV-2 Mpro and anti-inflammatory properties. Dihydromyricetin, as a flavonol, also has antiviral and anti-inflammatory potential. However, the inhibition of dihydromyricetin on SARS-CoV-2 Mpro and the protective effect of dihydromyricetin on pulmonary inflammation and fibrosis have not been proved and explained. PURPOSE: The coronavirus main protease (Mpro) is essential for SARS-CoV-2 replication and to be recognized as an attractive drug target, we expect to find the inhibitor of Mpro. Novel coronavirus infection can cause severe inflammation and even sequelae of pulmonary fibrosis in critically ill patients. We hope to find a drug that can not only inhibit virus replication but also alleviate inflammation and pulmonary fibrosis in patients. METHODS: FRET-based enzymatic assay was used to evaluate the inhibit activity of dihydromyricetin on SARS-CoV-2 Mpro. Molecular docking was used to identify the binding pose of dihydromyricetin with SARS-CoV-2 Mpro. The protective effects of dihydromyricetin against BLM-induced pulmonary inflammation and fibrosis were investigated in C57BL6 mice. BALF and lung tissue were collected for inflammation cells count, ELISA, masson and HE staining, western blotting and immunohistochemistry to analyze the effects of dihydromyricetin on pulmonary inflammation and fibrosis. MTT, western blotting, reverse transcription-polymerase chain reaction (RT-PCR) and wound healing were used to analyze the effects of dihydromyricetin on lung fibrosis mechanisms in Mlg cells. RESULTS: In this study, we found that dihydromyricetin is a potent inhibitor targeting the SARS-CoV-2 Mpro with a half-maximum inhibitory concentration (IC50) of 1.716 ± 0.419 µM, using molecular docking and the FRET-based enzymatic assay. The binding pose of dihydromyricetin with SARS-CoV-2 Mpro was identified using molecular docking method. In the binding pocket of SARS-CoV-2 Mpro, the dihydrochromone ring of dihydromyricetin interact with the imidazole side chain of His163 through π-π stacking. The 1-oxygen of dihydromyricetin forms a hydrogen bond with the backbone nitrogen of Glu166. The 3-, 7-, 3'- and 4'-hydroxyl of dihydromyricetin interact with Gln189, Leu141, Arg188 and Thr190 through hydrogen bonds. Moreover, our results showed that dihydromyricetin can significantly alleviate BLM-induced pulmonary inflammation by inhibiting the infiltration of inflammation cells and the secretion of inflammation factors in the early process and also ameliorate pulmonary fibrosis by improving pulmonary function and down-regulate the expression of α-SMA and fibronectin in vivo. Our results also showed that dihydromyricetin inhibits the migration and activation of myofibroblasts and extracellular matrix production via transforming growth factor (TGF)-ß1/Smad signaling pathways. CONCLUSION: Dihydromyricetin is an effective inhibitor for SARS-CoV-2 Mpro and it prevents BLM-induced pulmonary inflammation and fibrosis in mice. Dihydromyricetin will be a potential medicine for the treatment of COVID-19 and its sequelae.


Asunto(s)
Proteasas 3C de Coronavirus/antagonistas & inhibidores , Flavonoles/farmacología , Inhibidores de Proteasas , SARS-CoV-2 , Replicación Viral , Animales , Antivirales/farmacología , COVID-19 , Fibrosis , Humanos , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Inhibidores de Proteasas/farmacología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología , Replicación Viral/efectos de los fármacos
13.
Viruses ; 13(8)2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-34452298

RESUMEN

Influenza A viruses are serious zoonotic pathogens that continuously cause pandemics in several animal hosts, including birds, pigs, and humans. Indole derivatives containing an indole core framework have been extensively studied and developed to prevent and/or treat viral infection. This study evaluated the anti-influenza activity of several indole derivatives, including 3-indoleacetonitrile, indole-3-carboxaldehyde, 3-carboxyindole, and gramine, in A549 and MDCK cells. Among these compounds, 3-indoleacetonitrile exerts profound antiviral activity against a broad spectrum of influenza A viruses, as tested in A549 cells. Importantly, in a mouse model, 3-indoleacetonitrile with a non-toxic concentration of 20 mg/kg effectively reduced the mortality and weight loss, diminished lung virus titers, and alleviated lung lesions of mice lethally challenged with A/duck/Hubei/WH18/2015 H5N6 and A/Puerto Rico/8/1934 H1N1 influenza A viruses. The antiviral properties enable the potential use of 3-indoleacetonitrile for the treatment of IAV infection.


Asunto(s)
Antivirales/farmacología , Indoles/farmacología , Indoles/uso terapéutico , Virus de la Influenza A/efectos de los fármacos , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Células A549 , Animales , Antivirales/uso terapéutico , Antivirales/toxicidad , Perros , Femenino , Humanos , Indoles/toxicidad , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H5N1 del Virus de la Influenza A/fisiología , Virus de la Influenza A/fisiología , Pulmón/patología , Pulmón/virología , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Sulfuros/farmacología , Carga Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
14.
PLoS One ; 16(8): e0256429, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34415962

RESUMEN

INTRODUCTION: Coronavirus disease 2019 (COVID-19) has emerged as a global pandemic since its outbreak in Wuhan, China. It is an urgent task to prevent and treat COVID-19 effectively early. In China's experience combating the COVID-19 pandemic, Chinese herbal medicine (CHM) has played an indispensable role. A large number of epidemiological investigations have shown that mild to moderate COVID-19 accounts for the largest proportion of cases. It is of great importance to treat such COVID-19 cases, which can help control epidemic progression. Many trials have shown that CHM combined with conventional therapy in the treatment of mild to moderate COVID-19 was superior to conventional therapy alone. This review was designed to evaluate the add-on effect of CHM in the treatment of mild to moderate COVID-19. METHODS: Eight electronic databases including PubMed, EMBASE, Cochrane Central Register of Controlled Trials, the Clinical Trials.gov website, China National Knowledge Infrastructure (CNKI), China Science and Technology Journal Database (VIP), Wanfang Database and China Biology Medicine (CBM) were searched from December 2019 to March 2021 without language restrictions. Two reviewers searched and selected studies, and extracted data according to inclusion and exclusion criteria independently. Cochrane Risk of Bias (ROB) tool was used to assess the methodological quality of the included RCTs. Review Manager 5.3.0 software was used for statistical analysis. RESULTS: Twelve eligible RCTs including 1393 participants were included in this meta-analysis. Our meta-analyses found that lung CT parameters [RR = 1.26, 95% CI (1.15, 1.38), P<0.00001] and the clinical cure rate [RR = 1.26, 95%CI (1.16, 1.38), P<0.00001] of CHM combined with conventional therapy in the treatment of mild to moderate COVID-19 were better than those of conventional therapy. The rate of conversion to severe cases [RR = 0.48, 95%CI (0.32, 0.73), P = 0.0005], TCM symptom score of fever [MD = -0.62, 95%CI (-0.79, -0.45), P<0.00001], cough cases [RR = 1.43, 95%CI (1.16, 1.75), P = 0.0006], TCM symptom score of cough[MD = -1.07, 95%CI (-1.29, -0.85), P<0.00001], TCM symptom score of fatigue[MD = -0.66, 95%CI (-1.05, -0.28), P = 0.0007], and CRP[MD = -5.46, 95%CI (-8.19, -2.72), P<0.0001] of combination therapy was significantly lower than that of conventional therapy. The WBC count was significantly higher than that of conventional therapy[MD = 0.38, 95%CI (0.31, 0.44), P<0.00001]. Our meta-analysis results were robust through sensitivity analysis. CONCLUSION: Chinese herbal medicine combined with conventional therapy may be effective and safe in the treatment of mild to moderate COVID-19. More high-quality RCTs are needed in the future.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Medicamentos Herbarios Chinos/efectos adversos , Medicamentos Herbarios Chinos/uso terapéutico , COVID-19/diagnóstico por imagen , COVID-19/etiología , Tos/tratamiento farmacológico , Tos/virología , Diarrea/inducido químicamente , Medicamentos Herbarios Chinos/química , Fiebre/tratamiento farmacológico , Fiebre/virología , Humanos , Pulmón/diagnóstico por imagen , Pulmón/virología , Náusea/inducido químicamente , Tomografía Computarizada por Rayos X , Resultado del Tratamiento , Vómitos/inducido químicamente
15.
J Virol ; 95(18): e0060021, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34106002

RESUMEN

Coronaviruses are commonly characterized by a unique discontinuous RNA transcriptional synthesis strategy guided by transcription-regulating sequences (TRSs). However, the details of RNA synthesis in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have not been fully elucidated. Here, we present a time-scaled, gene-comparable transcriptome of SARS-CoV-2, demonstrating that ACGAAC functions as a core TRS guiding the discontinuous RNA synthesis of SARS-CoV-2 from a holistic perspective. During infection, viral transcription, rather than genome replication, dominates all viral RNA synthesis activities. The most highly expressed viral gene is the nucleocapsid gene, followed by ORF7 and ORF3 genes, while the envelope gene shows the lowest expression. Host transcription dysregulation keeps exacerbating after viral RNA synthesis reaches a maximum. The most enriched host pathways are metabolism related. Two of them (cholesterol and valine metabolism) affect viral replication in reverse. Furthermore, the activation of numerous cytokines emerges before large-scale viral RNA synthesis. IMPORTANCE SARS-CoV-2 is responsible for the current severe global health emergency that began at the end of 2019. Although the universal transcriptional strategies of coronaviruses are preliminarily understood, the details of RNA synthesis, especially the time-matched transcription level of each SARS-CoV-2 gene and the principles of subgenomic mRNA synthesis, are not clear. The coterminal subgenomic mRNAs of SARS-CoV-2 present obstacles in identifying the expression of most genes by PCR-based methods, which are exacerbated by the lack of related antibodies. Moreover, SARS-CoV-2-related metabolic imbalance and cytokine storm are receiving increasing attention from both clinical and mechanistic perspectives. Our transcriptomic research provides information on both viral RNA synthesis and host responses, in which the transcription-regulating sequences and transcription levels of viral genes are demonstrated, and the metabolic dysregulation and cytokine levels identified at the host cellular level support the development of novel medical treatment strategies.


Asunto(s)
COVID-19/genética , Células Epiteliales/metabolismo , Pulmón/metabolismo , ARN Mensajero/genética , SARS-CoV-2/aislamiento & purificación , Transcriptoma , Animales , COVID-19/metabolismo , COVID-19/virología , Células Cultivadas , Chlorocebus aethiops , Células Epiteliales/virología , Humanos , Pulmón/virología , ARN Mensajero/metabolismo , Células Vero , Replicación Viral
16.
Viruses ; 13(5)2021 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-34063247

RESUMEN

In late 2019, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic emerged to severely impact the global population, creating an unprecedented need for effective treatments. This study aims to investigate the potential of Scutellaria barbata D. Don (SB) as a treatment for SARS-CoV-2 infection through the inhibition of the proteases playing important functions in the infection by SARS-CoV-2. FRET assay was applied to investigate the inhibitory effects of SB on the two proteases involved in SARS-CoV-2 infection, Mpro and TMPRSS2. Additionally, to measure the potential effectiveness of SB treatment on infection inhibition, cellular models based on the Calu3 and VeroE6 cells and their TMPRSS2- expressing derivatives were assessed by viral pseudoparticles (Vpp) infection assays. The experimental approaches were conjugated with LC/MS analyses of the aqueous extracts of SB to identify the major constituent compounds, followed by a literature review to determine the potential active components of the inhibitory effects on protease activities. Our results showed that SB extracts inhibited the enzyme activities of Mpro and TMPRSS2. Furthermore, SB extracts effectively inhibited SARS-CoV-2 Vpp infection through a TMPRSS2-dependent mechanism. The aqueous extract analysis identified six major constituent compounds present in SB. Some of them have been known associated with inhibitory activities of TMPRSS2 or Mpro. Thus, SB may effectively prevent SARS-CoV-2 infection and replication through inhibiting Mpro and TMPRSS2 protease activities.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Proteasas 3C de Coronavirus/metabolismo , Extractos Vegetales/farmacología , Serina Endopeptidasas/metabolismo , Animales , COVID-19/metabolismo , Línea Celular , Chlorocebus aethiops , Proteasas 3C de Coronavirus/efectos de los fármacos , Humanos , Pulmón/virología , Pandemias , Péptido Hidrolasas , Peptidil-Dipeptidasa A/metabolismo , Extractos Vegetales/metabolismo , Proteolisis , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/patogenicidad , Scutellaria , Serina Endopeptidasas/efectos de los fármacos , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus/efectos de los fármacos
17.
Biomed Pharmacother ; 141: 111843, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34175821

RESUMEN

OBJECTIVE: To investigate the antiviral effect of Salvia plebeia R. Br. polysaccharides (SPP) against RSV and underlying mechanisms. METHODS: SPP was extracted via alcohol-precipitation method and extract was separated into various fractions using ultrafiltration method. The polysaccharide content was determined using UV-Vis. Antiviral effect of SPP and fractions was measured using MTT method and Reed-Muench method. Sixty Balb/c mice were randomly divided into 6 groups, and received either Ribavirin or SPP. Their body weight and food intake were recorded every day throughout the experiment period. The lung index inhibition ratio and pulmonary virus titer were determined followed by the histological analysis of lungs. Furthermore, time-of-addition and effective stage analysis were carried out to determine the mechanism of action. The TLR-3 and TLR-4 levels in the lungs were determined using qRT-PCR. The levels of IFN-γ, IL-2 and TNF-α in serum were determined using ELISA. RESULTS: The SPP content is 4.396%. SPP has shown a good anti-RSV effect both in vitro (TI = 123.041) and in vivo models. The antiviral activity of fractions with molecular weight ≥ 10,000 is found to possess more potent antiviral activity than other fractions. SPP inhibits the RSV proliferation and reduces the lung lesions induced by RSV. The mechanism of action involves the inhibition of TLR-3 and TLR-4 in lungs, up-regulation of IFN-γ and IL-2, and down-regulation of TNF-α in serum. It is also shown to improve the body's immune function. CONCLUSION: SPP has a potential to treat diseases caused by RSV.


Asunto(s)
Antivirales/farmacología , Antivirales/uso terapéutico , Polisacáridos/farmacología , Polisacáridos/uso terapéutico , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Salvia/química , Animales , Peso Corporal/efectos de los fármacos , Línea Celular , Citocinas/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Humanos , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Extractos Vegetales , Pruebas de Función Respiratoria , Ribavirina/uso terapéutico , Sales de Tetrazolio , Tiazoles , Receptores Toll-Like/metabolismo
18.
Pharm Biol ; 59(1): 696-703, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34110959

RESUMEN

CONTEXT: COVID-19 is a novel coronavirus that causes a severe infection in the respiratory system. Nigella sativa L. (Ranunculaceae) is an annual flowering plant used traditionally as a natural food supplement and multipurpose medicinal agent. OBJECTIVE: The possible beneficial effects of N. sativa, and its constituent, thymoquinone (TQ) on COVID-19 were reviewed. METHODS: The key words including, COVID-19, N. sativa, thymoquinone, antiviral effects, anti-inflammatory and immunomodulatory effects in different databases such as Web of Science (ISI), PubMed, Scopus, and Google Scholar were searched from 1990 up to February 2021. RESULTS: The current literature review showed that N. sativa and TQ reduced the level of pro-inflammatory mediators including, IL-2, IL-4, IL-6, and IL-12, while enhancing IFN-γ. Nigella sativa and TQ increased the serum levels of IgG1 and IgG2a, and improved pulmonary function tests in restrictive respiratory disorders. DISCUSSION AND CONCLUSIONS: These preliminary data of molecular docking, animal, and clinical studies propose N. sativa and TQ might have beneficial effects on the treatment or control of COVID-19 due to antiviral, anti-inflammatory and immunomodulatory properties as well as bronchodilatory effects. The efficacy of N. sativa and TQ on infected patients with COVID-19 in randomize clinical trials will be suggested.


Asunto(s)
Antiinflamatorios/farmacología , Antivirales/farmacología , Benzoquinonas/farmacología , Tratamiento Farmacológico de COVID-19 , Nigella sativa , Extractos Vegetales/farmacología , SARS-CoV-2/efectos de los fármacos , Animales , Antiinflamatorios/aislamiento & purificación , Antivirales/aislamiento & purificación , Benzoquinonas/aislamiento & purificación , COVID-19/inmunología , COVID-19/metabolismo , COVID-19/virología , Citocinas/metabolismo , Humanos , Sistema Inmunológico/efectos de los fármacos , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Sistema Inmunológico/virología , Mediadores de Inflamación/metabolismo , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/virología , Nigella sativa/química , Extractos Vegetales/aislamiento & purificación , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad
19.
J Nat Prod ; 84(4): 1261-1270, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33844528

RESUMEN

The coronaviruses disease 2019 (COVID-19) caused by a novel coronavirus (SARS-CoV-2) has become a major health problem, affecting more than 50 million people with over one million deaths globally. Effective antivirals are still lacking. Here, we optimized a high-content imaging platform and the plaque assay for viral output study using the legitimate model of human lung epithelial cells, Calu-3, to determine the anti-SARS-CoV-2 activity of Andrographis paniculata extract and its major component, andrographolide. SARS-CoV-2 at 25TCID50 was able to reach the maximal infectivity of 95% in Calu-3 cells. Postinfection treatment of A. paniculata and andrographolide in SARS-CoV-2-infected Calu-3 cells significantly inhibited the production of infectious virions with an IC50 of 0.036 µg/mL and 0.034 µM, respectively, as determined by the plaque assay. The cytotoxicity profile developed over the cell line representatives of major organs, including liver (HepG2 and imHC), kidney (HK-2), intestine (Caco-2), lung (Calu-3), and brain (SH-SY5Y), showed a CC50 of >100 µg/mL for A. paniculata extract and 13.2-81.5 µM for andrographolide, respectively, corresponding to a selectivity index of over 380. In conclusion, this study provided experimental evidence in favor of A. paniculata and andrographolide for further development as a monotherapy or in combination with other effective drugs against SARS-CoV-2 infection.


Asunto(s)
Andrographis , Diterpenos/farmacología , Extractos Vegetales/farmacología , SARS-CoV-2/efectos de los fármacos , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Epiteliales/virología , Humanos , Hidroxicloroquina/farmacología , Pulmón/virología
20.
J Ethnopharmacol ; 275: 114063, 2021 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-33813013

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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA