Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Molecules ; 27(18)2022 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-36144628

RESUMEN

α-Linolenic acid (ALA) is a natural essential fatty acid widely found in plant seed oils and beans, which shows positive anti-inflammatory and antiallergic effects. In our previous study, ALA was proven to bind tightly to the seven protein targets closely associated with allergic rhinitis (AR) by molecular docking, which indicates that ALA may have a potential role in the treatment of AR. A mouse model of AR induced by ovalbumin (OVA) was adopted in this study to explore the therapeutical effect and potential mechanism of ALA in treating AR. Results demonstrated that ALA remarkably relieved the nasal symptoms, reduced the OVA-sIgE level in the serum, relieved the histopathological injuries, and downregulated the mRNA expression levels of IL-6 and IL-1ß in the nasal mucosa. ALA also remarkably moderated the imbalance of Th1/Th2 cells, increased the mRNA expression levels of T-bet and STAT1, and reduced GATA3 and STAT6. ALA was proven to have a substantial therapeutic effect on mice with AR, and the underlying mechanism was likely to be the regulation of Th1/Th2 imbalance through the JAK/T-bet/STAT1 and JAK/GATA3/STAT6 pathways. This study provides a specific experimental basis for the clinical use and drug development of ALA in the treatment of AR.


Asunto(s)
Antialérgicos , Rinitis Alérgica , Animales , Antialérgicos/farmacología , Antiinflamatorios/farmacología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inflamación/tratamiento farmacológico , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos BALB C , Simulación del Acoplamiento Molecular , Mucosa Nasal , Ovalbúmina , Aceites de Plantas/farmacología , ARN Mensajero/metabolismo , Rinitis Alérgica/inducido químicamente , Rinitis Alérgica/tratamiento farmacológico , Rinitis Alérgica/metabolismo , Células Th2 , Ácido alfa-Linolénico/metabolismo , Ácido alfa-Linolénico/farmacología
2.
Phytomedicine ; 95: 153874, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34923232

RESUMEN

BACKGROUND: Although severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and human coronavirus 229E (HCoV-229E) pose a huge threat to human public health, no specific treatment is available. Jinzhen granule (JZ) is a traditional eight ingredients-Chinese medicine with prominent efficacy for treating viral-induced diseases. However, little is known about the antiviral effect and mechanism of JZ against SARS-CoV-2 and HCoV-229E. PURPOSE: This study aimed to reveal the antiviral effects of JZ against SARS-CoV-2 and HCoV-229E, and to further explore the underlying mechanisms regulating the host immune response. METHODS: The chromatographic separation of JZ was performed using a Shimadzu analytical high-performance liquid chromatograph with UV detection and Alltech ELSD 2000ES. We conducted cytopathic effect (CPE) and plaque reduction assays to evaluate the antiviral effect of JZ. A lethal human angiotensin converting enzyme 2 (hACE2) transgenic mouse model of SARS-CoV-2 was established to determine the protective effect of JZ on mortality and lung virus titers. Real-time quantitative PCR assays were used to analyze the expression of proinflammatory cytokines in vitro and in vivo. Western blotting was further performed to determine the activities on regulating the nuclear factor kappa B (NF-κB)/MAPK pathway. Finally, mitochondrial membrane potential assays, flow cytometry analysis and western blotting were used to assess the anti-apoptotic potency toward HCoV-229E infection. RESULTS: The results showed that 13 chemical components were identified and five peaks were determined and quantitated (gallic acid 1.97 mg/g, baicalin 20.69 mg/g, glycyrrhizic acid 4.92 mg/g, hyodeoxycholic acid 4.86 mg/g, cholic acid 4.07 mg/g). We found that JZ exerted inhibitory potency against SARS-CoV-2 and HCoV-229E in vitro by using CPE and plaque reduction assays, and it was further found that JZ protected mice infected by SARS-CoV-2 from death and inhibited lung virus titers. JZ also significantly decreased the induction of inflammatory cytokines (IL-1α, IL-6, CCL-5 and MIP-1ß), similar to the observed in vitro effect. Moreover, JZ suppressed the release of inflammatory cytokines in vitro and it decreased the protein expression of p-p38 MAPK, p-JNK, p-NF-κB p65 and p-IκBα induced by HCoV-229E and increased the expression of IκBα. Notably, JZ significantly protected HCoV-229E-infected Huh-7 cells from mitochondrial damage and decreased apoptotic cells. The activation of the mitochondria-mediated apoptotic pathway was inhibited by JZ, as shown by the reduced expression of cleaved caspase-9, caspase-3 and p-PARP. CONCLUSIONS: In conclusion, JZ (gallic acid 1.97 mg/g, baicalin 20.69 mg/g, glycyrrhizic acid 4.92 mg/g, hyodeoxycholic acid 4.86 mg/g, cholic acid 4.07 mg/g) exhibited antiviral activities against SARS-CoV-2 and HCoV-229E by regulating the NF-κB/MAPK pathway and the mitochondria-mediated apoptotic pathway. These findings demonstrated the efficacy of JZ against CoVs and suggested JZ treatment as a novel clinical therapeutic strategy for COVID-19.


Asunto(s)
Antivirales , Coronavirus Humano 229E , Medicamentos Herbarios Chinos/farmacología , SARS-CoV-2/efectos de los fármacos , Animales , Antivirales/farmacología , COVID-19 , Coronavirus Humano 229E/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , FN-kappa B
3.
Phytomedicine ; 93: 153808, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34753027

RESUMEN

BACKGROUND: Qingwenjiere Mixture (QJM) is a traditional Chinese medicine (TCM) that has been shown to have remarkable clinical efficacy against COVID-19. However, little is known about the antiviral and anti-inflammatory activities of QJM against a wider range of human coronavirus (HCoV) strains. PURPOSE: The study aims to investigate the antiviral and anti-inflammatory activities of QJM, as well as the underlying mechanisms against HCoV infections. METHODS: The chemical compositions from QJM were analyzed by LC-MS. The inhibitory effect of QJM on infections of HCoV-OC43, HCoV-229E, HCoV-NL63, and SARS-CoV-2 was evaluated in HRT-18 cells, Huh7 cells, LLC-MK2 cells, and Vero-E6 cells, respectively, by using cytopathic effect (CPE) inhibition assay or RT-qPCR detection of viral n, s, or RdRp/Hel genes. The expression of pro-inflammatory cytokines induced by HCoV-OC43, HCoV-229E, and SARS-CoV-2, as well as the host ace2 gene was also determined by RT-qPCR assay. Furthermore, the expression of key molecules in the NF-κB/MAPKs signaling pathways was determined by western blot. RESULTS: In alcohol-extraction groups of QJM and reference decoction pieces, 53 similar ion peaks were identified, the majority of which were phenylpropanoids, iridoids, and flavonoids. In addition, QJM reduced CPE caused by HCoVs and the expression of viral n genes or N protein. Pretreatment with QJM also exerted inhibitory effect on viral n gene expression. QJM also inhibited the expression of RdRp/Hel and s genes of SARS-CoV-2, as well as the host ace2 gene. Besides, QJM markedly reduced virus-induced mRNA expression of a panel of pro-inflammatory cytokines, such as IL-6, CXCL-8/IL-8, CXCL-10/IP-10, CCL-5/RANTES, TNF-α, IFN-α, CCL-2/MCP-1, CXCL-9/MIG, and IL1-α. We further showed that QJM inhibited the phosphorylation of NF-κB p65, and JNK, ERK 1/2, and p38 MAPKs in HCoV-OC43-infected HRT-18 cells. CONCLUSIONS: QJM has broad antiviral and anti-inflammatory activity against both common and newly emerged HCoVs possibly by inhibiting the activation of key components in NF-κB/MAPKs signaling pathway. QJM also has a prevention effect against HCoV infections and inhibits the host receptor required for virus entry. These results indicate that QJM may have the therapeutic potential in the treatment of diseases caused by a broad range of HCoVs.


Asunto(s)
COVID-19 , Coronavirus Humano OC43 , Antiinflamatorios/farmacología , Antivirales/farmacología , Humanos , SARS-CoV-2
4.
Artículo en Inglés | MEDLINE | ID: mdl-34497658

RESUMEN

BACKGROUND: Luofushan-Baicao Oil (LBO) is an essential oil-rich traditional Chinese medicine (TCM) formula that is commonly used to treat cold, cough, headache, sore throat, swelling, and pain. However, the anti-influenza activities of LBO and the underlying mechanism remain to be investigated. METHODS: The in vitro anti-influenza activity of LBO was tested with methyl thiazolyl tetrazolium (MTT) and plaque assays. The effects of LBO on the expressions of viral nucleoprotein and cytokines were evaluated. In the polyinosinic-polycytidylic acid- (Poly I: C-) induced inflammation model, the influences of LBO on the expression of cytokines and the activation of NF-κB P65 (P65) and interferon regulatory factor 3 (IRF3) were tested. After influenza A virus (IVA) infection, mice were administered with LBO for 5 days. The lung index, histopathologic change, the expression of viral protein, P65, and IRF3 in the lung tissue were measured. The levels of proinflammatory cytokines in serum were examined. RESULTS: In vitro, LBO could significantly inhibit the infection of IVA, decrease the formation of plaques, and reduce the expression of viral nucleoprotein and cytokines. LBO could also effectively downregulate the expression of interleukin-1ß (IL-1ß), interleukin-6 (IL-6), and interferon-ß and the activation of P65 and IRF3 in Poly I:C-treated cells. In the IVA-infected mice model, inhalation of LBO with atomizer could decrease the lung index, alleviate the pathological injury in the lung tissue, and reduce the serum levels of IL-1ß and IL-6. LBO could significantly downregulate the expression of viral protein (nucleoprotein, PB2, and matrix 2 ion channel) and the phosphorylation of P65 and IRF3 in the lungs of mice. CONCLUSION: The therapeutic effects of LBO on treating influenza might result from the regulation of the immune response of IVA infection. LBO can be developed as an alternative therapeutic agent for influenza prevention.

5.
J Ethnopharmacol ; 279: 114367, 2021 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-34174375

RESUMEN

BACKGROUND: Although the rapid emergence of coronavirus disease 2019 (COVID-19) poses a considerable threat to global public health, no specific treatment is available for COVID-19. ReDuNing injection (RDN) is a traditional Chinese medicine known to exert antibacterial, antiviral, antipyretic, and anti-inflammatory effects. In addition, RDN has been recommended in the diagnosis and treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-associated pneumonia by the National Health Council and the National Administration of Chinese Medicine. However, there is no information regarding its efficacy against COVID-19. AIM OF STUDY: This study was designed to determine the clinical efficacy of RDN in patients with COVID-19 and characterize its antiviral activity against SARS-CoV-2 in vitro. MATERIALS AND METHODS: A total of 50 adults with COVID-19 were included in this study, and the primary endpoint was recovery from clinical symptoms following 14 days of treatment. General improvements were defined as the disappearance of the major symptoms of infection including fever, fatigue, and cough. The secondary endpoints included the proportion of patients who achieved clinical symptom amelioration on days 7 and 10, time to clinical recovery, time to a negative nucleic acid test result, duration of hospitalization, and time to defervescence. Plaque reduction and cytopathic effect assays were also performed in vitro, and reverse-transcription quantitative PCR was performed to evaluate the expression of inflammatory cytokines (TNF-α, IP-10, MCP-1, IL-6, IFN-α, IFN-γ, IL-2 and CCL-5) during SARS-CoV-2 infection. RESULTS: The RDN group exhibited a shorter median time for the resolution of clinical symptoms (120 vs. 220 h, P < 0.0001), less time to a negative PCR test result (215 vs. 310 h, P = 0.0017), shorter hospitalization (14.8 vs. 18.5 days, P = 0.0002), and lower timeframe for defervescence (24.5 vs. 75 h, P = 0.0001) than the control group. In addition, time to improved imaging was also shorter in the RDN group than in the control group (6 vs.8.9 days, P = 0.0273); symptom resolution rates were higher in the RDN group than in the control group at 7 (96.30% vs. 39.13%, P < 0.0001) and 10 days (96.30% vs. 56.52%, P = 0.0008). No allergic reactions or anaphylactic responses were reported in this trial. RDN markedly inhibited SARS-CoV-2 proliferation and viral plaque formation in vitro. In addition, RDN significantly reduced inflammatory cytokine production in infected cells. CONCLUSIONS: RDN relieves clinical symptoms in patients with COVID-19 and reduces SARS-CoV-2 infection by regulating inflammatory cytokine-related disorders, suggestion that this medication might be a safe and effective treatment for COVID-19.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19 , Citocinas/análisis , Medicamentos Herbarios Chinos , SARS-CoV-2 , Antivirales/administración & dosificación , Antivirales/efectos adversos , COVID-19/epidemiología , COVID-19/inmunología , Prueba de Ácido Nucleico para COVID-19/métodos , Línea Celular , China/epidemiología , Pruebas Inmunológicas de Citotoxicidad/métodos , Monitoreo de Drogas/métodos , Medicamentos Herbarios Chinos/administración & dosificación , Medicamentos Herbarios Chinos/efectos adversos , Femenino , Hospitalización/estadística & datos numéricos , Humanos , Masculino , Persona de Mediana Edad , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/aislamiento & purificación , Evaluación de Síntomas/métodos , Resultado del Tratamiento
6.
J Ethnopharmacol ; 277: 114066, 2021 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-33766755

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Liu Shen Wan (LSW) is a traditional Chinese medicine (TCM) with detoxification and antiphlogistic activity; it is composed of bezoar, toad venom, musk, pearl powder, borneol and realgar. In recent years, LSW has been widely used in traditional medicine for the treatment of influenza, tonsillitis, pharyngitis, mumps, cancer and leukaemia. AIM OF STUDY: The anti-influenza virus properties of LSW and its inhibition of the inflammatory response was demonstrated in our previous research; however, the effect and potential mechanism of LSW against influenza induced secondary bacteria have remained obscure. Therefore, in the present study, a model of influenza virus PR8 with secondary infection by Staphylococcus aureus (S. aureus) in vitro and in mice was established to examine the effect and potential mechanism by which LSW inhibits bacterial adhesion and subsequent severe pneumonia after viral infection. MATERIALS AND METHODS: We investigated the effect of LSW on the PR8-induced adhesion of live S. aureus in A549 cells. RT-qPCR was used to detect the expression of adhesion molecules. Western blotting was used to determine the expression of CEACAM1, RIG-1, MDA5, p-NF-κB, and NF-κB in A549 cells. Inflammatory cytokines were detected using a Bio-Plex Pro Human Cytokine Screening Panel (R&D) in A549 cells and Mouse Magnetic Luminex Assays (R&D) in mice infected with PR8 virus and secondarily with S. aureus, respectively. Moreover, the survival rate, lung index, viral titre, bacterial loads and pathological changes in the lung tissue of mice infected with PR8 and S. aureus were investigated to estimate the effect of LSW in inhibiting severe pneumonia. RESULTS: LSW significantly decreased S. aureus adhesion following influenza virus infection in A549 cells, which may have occurred by suppressing expression of the adhesion molecule CEACAM1. In addition, treatment with LSW dramatically suppressed the induction of proinflammatory cytokines (CCL2/MCP-1 and CXCL-9/MIG) and chemokines (IL-6 and TNF-α) by PR8 infection following secondary LPS stimulation in A549 cells. Upregulation of related signalling proteins (RIG-I, MDA5 and NF-κB) induced by viruses and bacteria was suppressed by LSW in A549 cells. LSW significantly decreased the viral titres and bacterial load, prolonged survival time, and ameliorated lung inflammation and injury in mice with S. aureus infection secondary to PR8 infection. CONCLUSIONS: We demonstrated that LSW prevents S. aureus adherence to influenza virus-infected A549 cells, perhaps by inhibiting the expression of the adhesion molecule CEACAM1. The upregulation of proinflammatory cytokines and related signalling proteins induced by viruses and bacteria was suppressed by LSW in A549 cells. LSW significantly ameliorated lung injury caused by viral and secondary bacterial infection. These findings provide a further evaluation of LSW and suggest a beneficial effect of LSW for the prevention of secondary bacterial infection and related complications.


Asunto(s)
Mezclas Complejas/farmacología , Gripe Humana/complicaciones , Enfermedades Pulmonares/tratamiento farmacológico , Infecciones Estafilocócicas/tratamiento farmacológico , Células A549 , Animales , Citocinas/metabolismo , Perros , Femenino , Humanos , Gripe Humana/tratamiento farmacológico , Enfermedades Pulmonares/microbiología , Enfermedades Pulmonares/virología , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos BALB C , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/aislamiento & purificación , Tasa de Supervivencia
7.
J Ethnopharmacol ; 264: 112800, 2021 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-32224195

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: The San Wu Huangqin Decoction (SWHD), which is made from the dried root of Sophora flavescens Aiton (Kushen in Chinese), the dried root of Scutellaria baicalensis Georgi (Huangqin in Chinese), and the dried root tuber of Rehmannia glutinosa (Gaertn.) DC. (Dihuang in Chinese), is a traditional Chinese formula used to treat prolonged fever and inflammatory diseases in clinics and proven to inhibit influenza virus effectively in our previous study. AIM OF THE STUDY: This work was performed to study the regulation of SWHD on inflammation and immune dysfunction induced by the influenza virus and the underlying mechanism in the treatment of SWHD. METHODS: In this study, the influenza virus A/PR/8/34 (H1N1)-infected mouse model was used to investigate the regulation of SWHD on inflammation and immune dysfunction induced by H1N1. The pathological changes, the capacity of proliferation of T and B lymphocytes, the cytotoxicity of natural killer (NK) cells, and the levels of IL-6, TNF-α, IL-1ß, IL-4, and IFN-γ in the serum, bronchoalveolar lavage fluid (BALF), and lung were analyzed. The effects of type 1 T helper cell (Th1) and type 2 T helper cell (Th2) immune responses were discussed indirectly. In addition, the expression levels of p-p65, p65, IKKα/ß, p-IκBα, and IκBα in relation to the NF-κB pathway were measured using Western blot analysis, or immunohistochemical assay. RESULTS: SWHD decreased the pathological changes in lung tissues, promoted the proliferation of T and B lymphocytes, enhanced NK cell activity, and accelerated the phagocytic function of macrophages in H1N1-infected mice. At the same time, SWHD decreased the levels of IL-6, TNF-α, IL-1ß, IFN-γ, and increased the level of IL-4 in the serum, BALF, and lung of model mice. Moreover, the p-p65, p65, and IκBα protein expression levels were inhibited, whereas the p-IκBα protein expression levels were improved in the lungs of H1N1-infected mice. CONCLUSIONS: SWHD can inhibit the replication of the H1N1 virus and reduced the excessive inflammation and immune dysfunction induced by the H1N1 virus in the body. This work provides rich experimental basis for further anti-inflammation research of SWHD and sets the foundation for the development of a viral inflammation drug of traditional Chinese medicine.


Asunto(s)
Antivirales/uso terapéutico , Medicamentos Herbarios Chinos/uso terapéutico , Inmunidad Celular/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , FN-kappa B/fisiología , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Animales , Antivirales/farmacología , Relación Dosis-Respuesta a Droga , Medicamentos Herbarios Chinos/farmacología , Femenino , Inmunidad Celular/fisiología , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
8.
Phytomedicine ; 78: 153296, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32890913

RESUMEN

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has extensively and rapidly spread in the world, causing an outbreak of acute infectious pneumonia. However, no specific antiviral drugs or vaccines can be used. Phillyrin (KD-1), a representative ingredient of Forsythia suspensa, possesses anti-inflammatory, anti-oxidant, and antiviral activities. However, little is known about the antiviral abilities and mechanism of KD-1 against SARS-CoV-2 and human coronavirus 229E (HCoV-229E). PURPOSE: The study was designed to investigate the antiviral and anti-inflammatory activities of KD-1 against the novel SARS-CoV-2 and HCoV-229E and its potential effect in regulating host immune response in vitro. METHODS: The antiviral activities of KD-1 against SARS-CoV-2 and HCoV-229E were assessed in Vero E6 cells using cytopathic effect and plaque-reduction assay. Proinflammatory cytokine expression levels upon infection with SARS-CoV-2 and HCoV-229E infection in Huh-7 cells were measured by real-time quantitative PCR assays. Western blot assay was used to determine the protein expression of nuclear factor kappa B (NF-κB) p65, p-NF-κB p65, IκBα, and p-IκBα in Huh-7 cells, which are the key targets of the NF-κB pathway. RESULTS: KD-1 could significantly inhibit SARS-CoV-2 and HCoV-229E replication in vitro. KD-1 could also markedly reduce the production of proinflammatory cytokines (TNF-α, IL-6, IL-1ß, MCP-1, and IP-10) at the mRNA levels. Moreover, KD-1 could significantly reduce the protein expression of p-NF-κB p65, NF-κB p65, and p-IκBα, while increasing the expression of IκBα in Huh-7 cells. CONCLUSIONS: KD-1 could significantly inhibit virus proliferation in vitro, the up-regulated expression of proinflammatory cytokines induced by SARS-CoV-2 and HCoV-229E by regulating the activity of the NF-кB signaling pathway. Our findings indicated that KD-1 protected against virus attack and can thus be used as a novel strategy for controlling the coronavirus disease 2019.


Asunto(s)
Antiinflamatorios/farmacología , Antivirales/farmacología , Betacoronavirus/efectos de los fármacos , Coronavirus Humano 229E/efectos de los fármacos , Infecciones por Coronavirus , Glucósidos/farmacología , FN-kappa B/metabolismo , Pandemias , Neumonía Viral , Animales , COVID-19 , Chlorocebus aethiops , Coronavirus/efectos de los fármacos , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/virología , Citocinas/metabolismo , Forsythia/química , Humanos , Fitoterapia , Extractos Vegetales/farmacología , Neumonía Viral/metabolismo , Neumonía Viral/virología , SARS-CoV-2 , Síndrome Respiratorio Agudo Grave/virología , Transducción de Señal/efectos de los fármacos , Células Vero , Replicación Viral/efectos de los fármacos
10.
Pharmacol Res ; 158: 104850, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32360580

RESUMEN

Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has spread worldwide through person-to-person contact, causing a public health emergency of international concern. At present, there is no specific antiviral treatment recommended for SARS-CoV-2 infection. Liu Shen capsule (LS), a traditional Chinese medicine, has been proven to have a wide spectrum of pharmacological properties, such as anti-inflammatory, antiviral and immunomodulatory activities. However, little is known about the antiviral effect of LS against SARS-CoV-2. Herein, the study was designed to investigate the antiviral activity of SARS-CoV-2 and its potential effect in regulating the host's immune response. The inhibitory effect of LS against SARS-CoV-2 replication in Vero E6 cells was evaluated by using the cytopathic effect (CPE) and plaque reduction assay. The number of virions of SARS-CoV-2 was observed under transmission electron microscope after treatment with LS. Proinflammatory cytokine expression levels upon SARS-CoV-2 infection in Huh-7 cells were measured by real-time quantitative PCR assays. The results showed that LS could significantly inhibit SARS-CoV-2 replication in Vero E6 cells, and reduce the number of virus particles and it could markedly reduce pro-inflammatory cytokines (TNF-α, IL-6, IL-1ß, IL-8, CCL-2/MCP-1 and CXCL-10/IP-10) production at the mRNA levels. Moreover, the expression of the key proteins in the NF-κB/MAPK signaling pathway was detected by western blot and it was found that LS could inhibit the expression of p-NF-κB p65, p-IκBα and p-p38 MAPK, while increasing the expression of IκBα. These findings indicate that LS could inhibit SARS-CoV-2 virus infection via downregulating the expression of inflammatory cytokines induced virus and regulating the activity of NF-κB/MAPK signaling pathway in vitro, making its promising candidate treatment for controlling COVID-19 disease.


Asunto(s)
Betacoronavirus/efectos de los fármacos , Mezclas Complejas/farmacología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Alanina/análogos & derivados , Alanina/farmacología , Animales , Antiinflamatorios/farmacología , Antivirales/farmacología , COVID-19 , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Chlorocebus aethiops , Infecciones por Coronavirus/virología , Humanos , Mediadores de Inflamación/metabolismo , Pandemias , Neumonía Viral/virología , SARS-CoV-2 , Virión/efectos de los fármacos
11.
J Ethnopharmacol ; 252: 112584, 2020 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-31972325

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Liu Shen Wan (LSW), first prescribed in "Lei Yunshang Song Fen Tang Fang", traditional Chinese medicine (TCM), is used to cure influenza, tonsillitis, pharyngitis and mumps for more than one hundred years. AIM OF STUDY: LSW was proved extensive pharmacological properties, for instance, anti-inflammatory, anticancer, antiviral, analgesic, antibacterial and immunomodulatory activities. Nevertheless, the mechanism of this process and the evaluation of this product is still ambiguous. Hence, the study was designed to investigate the antiviral and anti-inflammatory activities of LSW against the influenza virus in vitro and vivo. MATERIALS AND METHODS: The antiviral activities of LSW were assayed in virus-infected cells and mice. To study the antiviral effects of LSW against influenza A/PR/8/34 virus (PR8), we employed CPE inhibition assay with different concentrations of LSW at different times of infection in vitro. The mice were intranasally infected with virus to induce viral pneumonia, then treated with different doses of LSW. The death protection of the mice, the lung index, virus titer and pathological changes in the lung tissue of mice were investigated to estimate the anti-virus effect of LSW. Moreover, RT-qPCR was used to determine the mRNA expression of TNF-α, IL-1ß, IL-6, and IFN-γ in the A549 cells and the supernatant of lung tissues, and the concentrations of these four cytokines in serum of mice were determined with ELISA. Western blot was used to determine the expression of TLR4, p-NF-κB p65, NF-κB p65, p-IκBα and IκBα in the A549 cells and lung tissues, which are the key targets of TLR4/NF-κB pathway. Moreover, the immunohistochemical assay was used to determine the expression of the NF-κB p65 in the mice lungs. RESULTS: LSW could significantly inhibit influenza virus at different stages of viral replication (at the process of the pre-, post-, and co-virus infection) in vitro. And LSW (100 mg/kg and 50 mg/kg) could effectively increase the survival time of mice. The virus titres, lung index, pathological changes in the mice lungs also decreased. Moreover, LSW could significantly reduce the contents of IL-1ß, TNF-α, IFN-γ and IL-6 in the infected cells and the infected-mice. In addition, LSW could significantly reduce the expression of TLR4, p-NF-κB p65, NF-κB p65 and p-IκBα, while increase the IκBα in the infected cells and in the lung of mice. CONCLUSIONS: LSW could significantly not only inhibit virus replication and proliferation in vitro, but also ameliorate pneumonia damage in vivo. The antiviral effect was attributed to down-regulating the expression of inflammatory cytokines induced by influenza virus via regulating the activity of TLR4/NF-кB signaling pathway.


Asunto(s)
Antiinflamatorios/uso terapéutico , Antivirales/uso terapéutico , Mezclas Complejas/uso terapéutico , Virus de la Influenza A/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Receptor Toll-Like 4/antagonistas & inhibidores , Células A549 , Animales , Antiinflamatorios/farmacología , Antivirales/farmacología , Mezclas Complejas/farmacología , Citocinas/genética , Perros , Femenino , Humanos , Virus de la Influenza A/fisiología , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/virología , Células de Riñón Canino Madin Darby , Medicina Tradicional China , Ratones Endogámicos BALB C , FN-kappa B/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/inmunología , Replicación Viral/efectos de los fármacos
12.
Front Biosci (Landmark Ed) ; 24(4): 616-627, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30844702

RESUMEN

Sophocarpine is one of the major ingredients of Sophorae flavescentis which could inhibits many kinds of cancers. However, the effect of sophocarpine on gastric cancer (GC) and the mechanism involved remain unknown. The present study aims to explore the effects of the sophocarpine on the proliferation and apoptosis of GC cells and elucidates the relevant molecular mechanisms. After treatment with sophocarpine, GC cells were evaluated on their proliferation, autophagy, cell cycle progress and apoptosis. The protein levels of LC3-I, LC3-II, Beclin, p62, PTEN, PI3K, p53, Bax, Bcl-2, AKT and p-AKT were detected by western blot. Sophocarpine inhibited the proliferation of GC cells both in vitro and in vivo dose-dependently. Sophocarpine not only caused cell apoptosis and cell cycle arrest in G0/G1 phase but also induced cell autophagy. Moreover, sophocarpine dose-dependently suppressed PI3K/AKT signaling pathway and activated apoptosis in gastric cancer cells. Thus, sophocarpine significantly inhibited the growth of GC cells through multiple mechanisms such as induction of autophagy, activation of cell apoptosis and down-regulation of cell survival signaling pathway.


Asunto(s)
Alcaloides/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Extractos Vegetales/farmacología , Neoplasias Gástricas/patología , Caspasas/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Medicina Tradicional China , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Neoplasias Gástricas/tratamiento farmacológico
13.
Viruses ; 10(3)2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29522425

RESUMEN

The San Wu Huangqin Decoction (SWHD), a traditional Chinese medicine formula, is used to treat colds caused by exposure to wind-pathogen, hyperpyrexia, infectious diseases and cancer; moreover, it is used for detoxification. The individual herbs of SWHD, such as Sophora flavescens and Scutellaria baicalensis, exhibit a wide spectrum of antiviral, anti-inflammatory, antibacterial, anticancer and other properties. The Chinese compound formula of SWHD is composed of S. flavescens, S. baicalensis and Rehmannia glutinosa. However, the effect of SWHD on the influenza virus (IFV) and its mechanism remain unknown. The aim of this study was to evaluate, for the first time, whether SWHD could be used to treat influenza. Results showed that SWHD could effectively inhibit influenza A/PR/8/34 (H1N1) virus at different stages of viral replication (confirmed through antiviral effect assay, penetration assay, attachment assay and internalization assay) in vitro. It could reduce the infection of the virus in a dose- and time-dependent manner, as confirmed by observing the cell cytopathic effect and calculating the cell viability (p < 0.05). SWHD demonstrated better antiviral activity than oseltamivir in the evaluation of antiviral prophylaxis on influenza (p < 0.05). The antiviral activity of SWHD may be related to its regulation ability on the immune system. Western blot, real-time polymerase chain reaction and indirect immunofluorescence assay showed that the expression of the four target viral proteins of the IFV (namely, haemagglutinin (HA), neuraminidase (NA), nucleoprotein (NP) and matrix-2 (M2)) reduced significantly (p < 0.05). Moreover, SWHD (23.40 and 11.70 g/kg) significantly alleviated the clinical signs, reduced the mortality and increased the survival time of infected mice (p < 0.05). The lung index, virus titres, pathological changes in lung tissues and the expression of key proteins of the IFV in mice also decreased (p < 0.05). In conclusion, SWHD possessed anti-influenza activity. This work provided a new view of complementary therapy and drug discovery for clinical treatment.


Asunto(s)
Antivirales/farmacología , Medicamentos Herbarios Chinos/farmacología , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Infecciones por Orthomyxoviridae/prevención & control , Infecciones del Sistema Respiratorio/prevención & control , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Antivirales/química , Núcleo Celular/metabolismo , Efecto Citopatogénico Viral/efectos de los fármacos , Perros , Relación Dosis-Respuesta a Droga , Medicamentos Herbarios Chinos/química , Genes Virales/genética , Pulmón/patología , Pulmón/virología , Células de Riñón Canino Madin Darby , Ratones , Estructura Molecular , Nucleoproteínas/metabolismo , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Infecciones del Sistema Respiratorio/patología , Infecciones del Sistema Respiratorio/virología , Análisis de Supervivencia , Transcripción Genética/efectos de los fármacos , Carga Viral/efectos de los fármacos
14.
J Ethnopharmacol ; 209: 24-31, 2017 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-28684300

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: The liquid oral formulation of Poria cocos polysaccharides is composed of polysaccharides of Lentinusedodes, Ganodermalucidum and Poria cocos(1:1:2), which are all fungi used in traditional Chinese medicine. Polysaccharides extracted from these fungi have been reported to exhibit an antitumor effect by modulating the immune system. AIM OF THE STUDY: The present study aimed to clarify the antitumor mechanism of an orally administered liquid containing Poriacocos and to further provide clinical guidance. MATERIALS AND METHODS: In this study, the effects of an orally administered liquid containing Poriacocos polysaccharides on the solid tumors formed from sarcoma 180 cells in mice were evaluated. The protein expression of Bcl-2, caspase-3, and caspase-9in the thymus, spleen and liver tissues in the mice was determined by Western blot analysis. In addition, hematoxylin-eosin(H&E)staining and immunohistochemistry were performed on thymus, spleen and liver tissue and the positive staining rate was calculated for the three protein expression. RESULTS: The liquid oral formulation of Poriacocos polysaccharides reduced Bcl-2 protein levels and increased caspase-3 and -9 protein levels in sarcoma 180 cells. CONCLUSION: The mechanism underlying the antitumor effects of the oral liquid formulation of Poriacocos polysaccharides involved inhibition of Bcl-2 expression and activation of caspase-9 expression in sarcoma 180 cells. Furthermore, the downstream caspase-3 promoter cascade was activated and cell apoptosis was activated in sarcoma 180 cells.


Asunto(s)
Antineoplásicos/uso terapéutico , Polisacáridos Fúngicos/uso terapéutico , Sarcoma/tratamiento farmacológico , Wolfiporia/química , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Femenino , Polisacáridos Fúngicos/administración & dosificación , Polisacáridos Fúngicos/química , Polisacáridos Fúngicos/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/tratamiento farmacológico
15.
Viruses ; 9(4)2017 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-28417913

RESUMEN

Shuang-Huang-Lian injectable powder (SHL)-a classical purified herbal preparation extracted from Scutellaria baicalensis, Lonicera japonica, and Forsythia suspense-has been used against human adenovirus III (HAdV3) for many years. The combination herb and its major bioactive compounds, including chlorogenic acid, baicalin, and forsythia glycosides A, are effective inhibitors of the virus. However, no comprehensive studies are available on the antiviral effects of SHL against HAdV3. Moreover, it remains unclear whether the mixture of chlorogenic acid, baicalin, and forsythia glycosides A (CBF) has enhanced antiviral activity compared with SHL. Therefore, a comparative study was performed to investigate the combination which is promising for further antiviral drug development. To evaluate their antivirus activity in parallel, the combination ratio and dose of CBF were controlled and consistent with SHL. First, the fingerprint and the ratio of CBF in SHL were determined by high performance liquid chromatography. Then, a plaque reduction assay, reverse transcription polymerase chain reaction (PCR), real-time polymerase chain reaction (qPCR), and enzyme-linked immunosorbent assay (ELISA) were used to explore its therapeutic effects on viral infection and replication, respectively. The results showed that SHL and CBF inhibited dose- and time-dependently HAdV3-induced plaque formation in A549 and HEp-2 cells. SHL was more effective than CBF when supplemented prior to and after viral inoculation. SHL prevented viral attachment, internalization, and replication at high concentration and decreased viral levels within and out of cells at non-toxic concentrations in both cell types. Moreover, the expression of tumor necrosis factor alpha (TNF)-α, interleukin (IL)-1ß, and IL-6 was lower and the expression of interferon (IFN)-γ was higher in both cell types treated with SHL than with CBF. In conclusion, SHL is much more effective and slightly less toxic than CBF.


Asunto(s)
Adenovirus Humanos/efectos de los fármacos , Antivirales/farmacología , Mezclas Complejas/farmacología , Medicamentos Herbarios Chinos/farmacología , Extractos Vegetales/farmacología , Adenovirus Humanos/fisiología , Antivirales/química , Antivirales/aislamiento & purificación , Línea Celular , Cromatografía Líquida de Alta Presión , Mezclas Complejas/química , Mezclas Complejas/aislamiento & purificación , Citocinas/antagonistas & inhibidores , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/aislamiento & purificación , Ensayo de Inmunoadsorción Enzimática , Forsythia/química , Humanos , Lonicera/química , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Scutellaria baicalensis/química , Carga Viral , Ensayo de Placa Viral , Acoplamiento Viral/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
16.
J Ethnopharmacol ; 179: 356-66, 2016 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-26719286

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: The Mahuang-Xingren (MX) herb pair, the combination of Herba Ephedrae (Mahuang in Chinese) and Semen Armeniacae Amarum (Xingren in Chinese), is a core component of traditional Chinese medicine formulations used to treat asthma and bronchitis. Although Xingren is considered to be toxic, MX is widely used in the clinic and has few adverse effects. The mechanism underlying detoxification of Xingren by Mahuang in MX remains unknown and merits investigation. AIM OF THE STUDY: To determine the mechanism underlying detoxification of Xingren by Mahuang in MX. MATERIALS AND METHODS: Acute toxic effects were evaluated in mice after oral administration of Mahuang, Xingren, and MX aqueous extracts. Synergism, additivity, and antagonism were quantified by determining the CI (combination index) and DRI (dose-reduction index), which were calculated by the median effect method. High performance liquid chromatography analysis of bioactive compounds (ephedrine, pseudoephedrine and amygdalin) in aqueous extracts and data from previous pharmacokinetic studies in rats were combined to explore the potential mechanism of toxicity antagonism by the components of MX. Moreover, the cytotoxic effects of amygdalin and amygdalin activated by ß-glucosidase (including different proportions of l-amygdalin and d-amygdalin) were also investigated. RESULTS: Mahuang prevented and antagonized the acute toxicity of Xingren and allowed escalation of the Xingren dose. Pearson correlation analysis indicated that the proportion of d-amygdalin was closely correlated with the antagonism of Xingren toxicity. The antagonism of its acute toxicity was primarily attributed to stereoselective metabolism of amygdalin. Interestingly, the process was facilitated by Mahuang, which led to reduced levels of the d-prunasin in vivo and thus reduced toxicity. Furthermore, the mechanism was also evaluated by testing the cytotoxicity of amygdalin. Metabolism of d-amygdalin was a major cause of cytotoxicity and no stereoselective metabolism occurred in culture medium. CONCLUSIONS: A comprehensive study of Xingren detoxification in the context of the MX combination suggested that stereoselective metabolism of amygdalin facilitated by Mahuang may be the crucial mechanism underlying detoxification of Xingren in the MX combination. Therefore, Mahuang acts to enhance and control the effects of Xingren in the MX combination. These results illustrate the rationale behind the combination of Mahuang and Xingren.


Asunto(s)
Amigdalina/metabolismo , Amigdalina/toxicidad , Medicamentos Herbarios Chinos/farmacología , Ephedra sinica , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Combinación de Medicamentos , Medicamentos Herbarios Chinos/metabolismo , Medicamentos Herbarios Chinos/toxicidad , Nitrilos/metabolismo , Estereoisomerismo
17.
J Pharm Biomed Anal ; 109: 67-73, 2015 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-25766850

RESUMEN

The Mahuang-Xingren herb-pair (MX), the combination of Herba Ephedrae (Mahuang in Chinese) and Semen Armeniacae Amarum (Xingren in Chinese), is a classical combination used in traditional Chinese Medicine to treat asthma and bronchitis. A simple and reliable ultra-performance liquid chromatography-tandem mass spectrometry method was developed to simultaneously quantify and compare the pharmacokinetics of 5 ephedra alkaloids and epimers of amygdalin and prunasin in rat plasma after oral administration of Mahuang, Xingren, and MX aqueous extracts. Samples were pretreated by a single-step protein precipitation with acetonitrile, and diphenhydramine hydrochloride and puerarin were used as internal standards. Pharmacokinetic parameters were investigated using DAS 3.2.2 (Mathematical Pharmacology Professional Committee of China, Shanghai, China). The validated method demonstrated adequate sensitivity, selectivity, and process efficiency for the bioanalysis of 8 compounds, including 3 pairs of epimers. MX administration improved the bioavailability of amygdalin and prunasin. Furthermore, MX facilitated intake of lower doses of ephedra alkaloids and increased elimination rates in comparison with Mahuang alone. These results illustrate the rationale behind the preferred use of the combination of Mahuang and Xingren. To our knowledge, this is the first report of stereo-selective metabolism of amygdalin. Further, the metabolic mechanism underlying this phenomenon merits future research attention.


Asunto(s)
Asteraceae/química , Medicamentos Herbarios Chinos/análisis , Medicamentos Herbarios Chinos/farmacocinética , Ephedra/química , Amigdalina/análisis , Amigdalina/farmacocinética , Animales , Disponibilidad Biológica , Cromatografía Líquida de Alta Presión , Masculino , Nitrilos/análisis , Nitrilos/farmacocinética , Control de Calidad , Ratas , Ratas Sprague-Dawley , Estándares de Referencia , Reproducibilidad de los Resultados , Estereoisomerismo , Espectrometría de Masas en Tándem
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA