Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Clin Exp Allergy ; 52(2): 250-264, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34757674

RESUMEN

BACKGROUND: Excessive production of IgE plays a major role in the pathology of food allergy. In an attempt to identify anti-IgE natural products, Arctium Lappa was one of the most effective herbs among approximately 300 screened medicinal herbs. However, little is known about its anti-IgE compounds. OBJECTIVE: To identify compounds from Arctium Lappa for targeted therapy on IgE production and explore their underlying mechanisms. METHODS: Liquid-liquid extraction and column chromatographic methods were used to purify the compounds. IgE inhibitory effects were determined on IgE-producing human myeloma U266 cells, peanut-allergic murine model and PBMCs from food-allergic patients. Genes involved in IgE inhibition in PBMCs were studied by RNA sequencing. RESULTS: The main compounds isolated were identified as arctiin and arctigenin. Both compounds significantly inhibited IgE production in U266 cells, with arctigenin the most potent (IC50=5.09µg/mL). Arctigenin (at a dose of 13 mg/kg) markedly reduced peanut-specific IgE levels, blocked hypothermia and histamine release in a peanut-allergic mouse model. Arctigenin also significantly reduced IgE production and Th2 cytokines (IL-5, IL-13) by PBMCs. We found 479 differentially expressed genes in PBMCs with arctigenin treatment (p < .001 and fold-change ≥1.5), involving 24 gene ontology terms (p < .001, FDR <0.05); cell division was the most significant. Eleven genes including UBE2C and BCL6 were validated by qPCR. CONCLUSION: Arctigenin markedly inhibited IgE production in U266 cells, peanut-allergic murine model and PBMCs from allergic patients by down-regulating cell division, cell cycle-related genes and up-regulating anti-inflammatory factors.


Asunto(s)
Hipersensibilidad a los Alimentos , Hipersensibilidad al Cacahuete , Animales , Anticuerpos Antiidiotipos , Hipersensibilidad a los Alimentos/tratamiento farmacológico , Furanos , Humanos , Lignanos , Ratones , Hipersensibilidad al Cacahuete/tratamiento farmacológico , Extractos Vegetales/química , Transcriptoma
2.
FASEB J ; 35(4): e21360, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33749932

RESUMEN

The novel coronavirus disease, COVID-19, has grown into a global pandemic and a major public health threat since its breakout in December 2019. To date, no specific therapeutic drug or vaccine for treating COVID-19 and SARS has been FDA approved. Previous studies suggest that berberine, an isoquinoline alkaloid, has shown various biological activities that may help against COVID-19 and SARS, including antiviral, anti-allergy and inflammation, hepatoprotection against drug- and infection-induced liver injury, as well as reducing oxidative stress. In particular, berberine has a wide range of antiviral activities such as anti-influenza, anti-hepatitis C, anti-cytomegalovirus, and anti-alphavirus. As an ingredient recommended in guidelines issued by the China National Health Commission for COVID-19 to be combined with other therapy, berberine is a promising orally administered therapeutic candidate against SARS-CoV and SARS-CoV-2. The current study comprehensively evaluates the potential therapeutic mechanisms of berberine in preventing and treating COVID-19 and SARS using computational modeling, including target mining, gene ontology enrichment, pathway analyses, protein-protein interaction analysis, and in silico molecular docking. An orally available immunotherapeutic-berberine nanomedicine, named NIT-X, has been developed by our group and has shown significantly increased oral bioavailability of berberine, increased IFN-γ production by CD8+ T cells, and inhibition of mast cell histamine release in vivo, suggesting a protective immune response. We further validated the inhibition of replication of SARS-CoV-2 in lung epithelial cells line in vitro (Calu3 cells) by berberine. Moreover, the expression of targets including ACE2, TMPRSS2, IL-1α, IL-8, IL-6, and CCL-2 in SARS-CoV-2 infected Calu3 cells were significantly suppressed by NIT-X. By supporting protective immunity while inhibiting pro-inflammatory cytokines; inhibiting viral infection and replication; inducing apoptosis; and protecting against tissue damage, berberine is a promising candidate in preventing and treating COVID-19 and SARS. Given the high oral bioavailability and safety of berberine nanomedicine, the current study may lead to the development of berberine as an orally, active therapeutic against COVID-19 and SARS.


Asunto(s)
Antivirales/farmacología , Berberina/farmacología , Tratamiento Farmacológico de COVID-19 , COVID-19 , Regulación de la Expresión Génica/efectos de los fármacos , Modelos Biológicos , SARS-CoV-2/metabolismo , Síndrome Respiratorio Agudo Grave , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/metabolismo , Administración Oral , COVID-19/metabolismo , Línea Celular , Simulación por Computador , Humanos , Pandemias , Síndrome Respiratorio Agudo Grave/tratamiento farmacológico , Síndrome Respiratorio Agudo Grave/metabolismo
3.
J Allergy Clin Immunol ; 138(2): 536-543.e4, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27130858

RESUMEN

BACKGROUND: Treatments to reverse peanut allergy remain elusive. Current clinical approaches using peanut oral/sublingual immunotherapy are promising, but concerns about safety and long-term benefit remain a barrier to wide use. Improved methods of delivering peanut-specific immunotherapy are needed. OBJECTIVE: We sought to investigate the efficacy and safety of peanut oral immunotherapy using CpG-coated poly(lactic-co-glycolic acid) nanoparticles containing peanut extract (CpG/PN-NPs) in a murine model of peanut allergy. METHODS: C3H/HeJ mice were rendered peanut allergic by means of oral sensitization with peanut and cholera toxin. Mice were then subjected to 4 weekly gavages with CpG/PN-NPs, vehicle (PBS), nanoparticles alone, peanut alone, CpG nanoparticles, or peanut nanoparticles. Untreated mice served as naive controls. After completing therapy, mice underwent 5 monthly oral peanut challenges. Anaphylaxis was evaluated by means of visual assessment of symptom scores and measurement of body temperature and plasma histamine levels. Peanut-specific serum IgE, IgG1, and IgG2a levels were measured by using ELISA, as were cytokine recall responses in splenocyte cultures. RESULTS: Mice with peanut allergy treated with CpG/PN-NPs but not vehicle or other treatment components were significantly protected from anaphylaxis to all 5 oral peanut challenges, as indicated by lower symptom scores, less change in body temperature, and a lower increase of plasma histamine levels. Importantly, CpG/PN-NP treatment did not cause anaphylactic reactions. Treatment was associated with a sustained and significant decrease in peanut-specific IgE/IgG1 levels and an increase in peanut-specific IgG2a levels. Compared with vehicle control animals, peanut recall responses in splenocyte cultures from nanoparticle-treated mice showed significantly decreased levels of TH2 cytokines (IL-4, IL-5, and IL-13) but increased IFN-γ levels in cell supernatants. CONCLUSIONS: Preclinical findings indicate that peanut oral immunotherapy with CpG/PN-NPs might be a valuable strategy for peanut-specific immunotherapy in human subjects.


Asunto(s)
Alérgenos/inmunología , Arachis/efectos adversos , Desensibilización Inmunológica , Ácido Láctico , Nanopartículas , Hipersensibilidad al Cacahuete/inmunología , Ácido Poliglicólico , Alérgenos/administración & dosificación , Animales , Citocinas/sangre , Citocinas/metabolismo , Desensibilización Inmunológica/métodos , Modelos Animales de Enfermedad , Femenino , Histamina/sangre , Inmunización , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Ratones , Hipersensibilidad al Cacahuete/diagnóstico , Hipersensibilidad al Cacahuete/metabolismo , Hipersensibilidad al Cacahuete/terapia , Extractos Vegetales/administración & dosificación , Extractos Vegetales/química , Extractos Vegetales/inmunología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico
4.
Ann Allergy Asthma Immunol ; 114(1): 43-51.e1, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25465920

RESUMEN

BACKGROUND: Maternal asthma is a risk factor for asthma in offspring; however, transmission of the risk for allergic asthma without direct offspring sensitization has not been explored. OBJECTIVE: To determine whether offspring from mothers with ovalbumin (OVA)-sensitized asthma would develop airway disease at first-ever exposure to OVA and whether preconception maternal treatment with the Antiasthma Simplified Herbal Medicine Intervention (ASHMI) or dexamethasone (DEX) could modify this risk in offspring. METHODS: Female BALB/c mice (F0) with OVA-induced asthma were generated using established protocols. Mice with asthma were treated with ASHMI, DEX, or water for 6 to 7 weeks. Naive mice served as controls. Subsequently, mice were mated. Twelve-day-old F1 offspring received 3 consecutive intranasal low- or high-dose OVA exposures without sensitization. Forty-eight hours later, airway inflammation, mucus hypersecretion, serum antibodies, and cytokines were evaluated. RESULTS: Offspring from OVA-sensitized mothers, but not naive mothers, showed eosinophilic and neutrophilic airway inflammation, and mucus hyperplasia after OVA exposure and he presence of OVA-specific IgG1 and IgG2a. Offspring of ASHMI- and DEX-treated mothers showed decreased airway inflammation and mucus hypersecretion after low-dose OVA (P < .05-.001 for the 2 comparisons vs offspring of OVA/Sham mothers). Offspring of ASHMI-treated, but not DEX-treated, mothers were protected after the high-dose OVA challenge (P < .05-.01 vs offspring OVA/Sham). Maternal ASHMI therapy was associated with increased IgG2a (P < .01 vs offspring of OVA/Sham mothers) and decreased bronchoalveolar lavage fluid CXCL-1 and eotaxin-1 levels (P < .01 and P < .05, respectively, vs offspring of OVA/Sham mothers). CONCLUSION: Offspring of mothers with OVA-induced asthma developed airway inflammation and mucus to first-ever OVA exposure without prior sensitization. Maternal therapy with ASHMI was superior to DEX in decreasing offspring susceptibility to airway disease and could be a strategy to lower asthma prevalence.


Asunto(s)
Asma/terapia , Medicamentos Herbarios Chinos/administración & dosificación , Inmunidad Materno-Adquirida/efectos de los fármacos , Pulmón/efectos de los fármacos , Neumonía/prevención & control , Hijos Adultos , Animales , Anticuerpos/sangre , Asma/inmunología , Quimiocina CCL11/genética , Quimiocina CCL11/metabolismo , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Citocinas/sangre , Dexametasona/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Inmunidad Innata/efectos de los fármacos , Pulmón/inmunología , Masculino , Exposición Materna/efectos adversos , Ratones , Ratones Endogámicos BALB C , Embarazo
5.
J Allergy Clin Immunol ; 134(6): 1339-1345.e7, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25441650

RESUMEN

BACKGROUND: Although maternal atopy is a risk factor for the development of peanut allergy, this phenomenon has not been well characterized experimentally, and the mechanisms underlying offspring risk are unclear. OBJECTIVE: We sought to determine whether offspring of mothers with peanut allergy (O-PAM mice) are more susceptible to peanut allergy than offspring of naive mothers (O-NM mice) in a murine model and, if so, whether the susceptibility is linked to TH2-biased epigenetic alterations. METHODS: Five-week-old O-PAM and O-NM mice were intragastrically sensitized to and challenged with peanut. Serum peanut-specific IgE levels, plasma histamine levels, anaphylactic reactions, and splenocyte and MLN cell cytokine production were measured. DNA methylation levels of the Il4 gene promoter from splenocytes and MLN cells from sensitized offspring and splenocytes from unsensitized neonatal offspring were determined by means of pyrosequencing. RESULTS: O-PAM mice exhibited 3-fold higher peanut-specific IgE levels after peanut sensitization, as well as 5-fold higher histamine levels and significantly higher anaphylactic symptom scores after challenge than O-NM mice (P < .05-.01). Cultured splenocytes and MLNs from O-PAM mice produced significantly more TH2 cytokines than cells from O-NM mice (P < .05-.01). Cells from O-PAM mice exhibited significantly reduced DNA methylation at CpG sites of the Il4 gene promoter than cells from O-NM mice. DNA methylation levels were inversely correlated with IL-4 and IgE production. O-PAM neonatal splenocyte hypomethylation of the Il4 gene promoter was also present. CONCLUSION: This study is the first to demonstrate that increased susceptibility to peanut allergy in O-PAM mice is associated with epigenetic alteration of the Il4 gene promoter. This finding might provide insight into preventing the development of early-life allergy.


Asunto(s)
Citocinas/genética , Citocinas/inmunología , Epigénesis Genética , Hipersensibilidad al Cacahuete/inmunología , Células Th2/inmunología , Anafilaxia/etiología , Anafilaxia/genética , Anafilaxia/inmunología , Animales , Arachis/efectos adversos , Arachis/inmunología , Islas de CpG , Metilación de ADN , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Ganglios Linfáticos/citología , Masculino , Ratones , Ratones Endogámicos C3H , Hipersensibilidad al Cacahuete/genética , Embarazo , Regiones Promotoras Genéticas , Bazo/citología
6.
Ann Allergy Asthma Immunol ; 112(4): 339-47.e1-2, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24679734

RESUMEN

BACKGROUND: Neutrophil-predominant asthma is less responsive to steroids and associated with poorer disease control. The effects of Antiasthma Simplified Herbal Medicine Intervention (ASHMI), a traditional Chinese medicine formula reported to be efficacious in asthmatic patients and murine asthma models, on neutrophil predominant asthma are unknown. OBJECTIVE: To determine the effects of standard ASHMI and refined formula ASHMI (ASHMI(II)) in a neutrophil-predominant murine model of ragweed (RW) asthma and explore underlying mechanisms. METHODS: BALB/c mice were systemically sensitized, intranasally challenged with RW extract, and orally treated with ASHMI, ASHMI(II), or vehicle (water). In a separate experiment, some RW sensitized mice were treated with dexamethasone before challenge. After RW challenge, airway hyperreactivity (AHR), total and differential bronchoalveolar lavage fluid leukocyte counts, lung histologic features, and bronchoalveolar lavage fluid cytokine and chemokine levels were assessed. RW stimulation of the murine macrophage cell line RAW264.7 was used to determine effects of ASHMI active compound ganoderic acid C1 (GAC1) on tumor necrosis factor α (TNF-α) production and regulation of phosphorylated IκB and histone deacetylase 2 (HDAC2) levels. RESULTS: ASHMI and ASHMI(II) markedly reduced AHR, mucous production, neutrophilic inflammation, and TNF-α, interleukin 8, and interleukin 17 levels and decreased eosinophilic inflammation and TH2 responses in vivo (P < .01-.001 for all). GAC1 inhibited TNF-α production in RW-stimulated RAW264.7 cells in association with suppression of phosphorylated IκB and increased HDAC2 expression. Dexamethasone failed to reduce AHR and neutrophilic inflammation. CONCLUSION: ASHMI treatment was efficacious in a murine model of neutrophil-predominant asthma via modulation of innate chemokines, TH2 responses, nuclear factor-κB, and HDAC2. ASHMI, and/or its constituent GAC1, may be a valuable option for treating neutrophil-predominant asthma.


Asunto(s)
Ambrosia/efectos adversos , Asma/terapia , Medicamentos Herbarios Chinos/administración & dosificación , Macrófagos/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Neumonía/terapia , Administración Oral , Alérgenos/inmunología , Animales , Antígenos de Plantas/inmunología , Línea Celular , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/metabolismo , Humanos , Proteínas I-kappa B/metabolismo , Recuento de Leucocitos , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Neutrófilos/inmunología , Triterpenos/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
7.
J Inflamm Res ; 17: 2547-2561, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38686360

RESUMEN

Introduction: Neutrophil predominant airway inflammation is associated with severe and steroid-resistant asthma clusters. Previously, we reported efficacy of ASHMI, a three-herb TCM asthma formula in a steroid-resistant neutrophil-dominant murine asthma model and further identified Ganoderic Acid C1 (GAC1) as a key ASHMI active compound in vitro. The objective of this study is to investigate GAC1 effect on neutrophil-dominant, steroid-resistant asthma in a murine model. Methods: In this study, Balb/c mice were systematically sensitized with ragweed (RW) and alum and intranasally challenged with ragweed. Unsensitized/PBS challenged mice served as normal controls. Post sensitization, mice were given 4 weeks of oral treatment with GAC1 or acute dexamethasone (Dex) treatment at 48 hours prior to challenge. Pulmonary cytokines were measured by ELISA, and lung sections were processed for histology by H&E staining. Furthermore, GAC1 effect on MUC5AC expression and on reactive oxygen species (ROS) production in human lung epithelial cell line (NCI-H292) was determined by qRT-PCR and ROS assay kit, respectively. Computational analysis was applied to select potential targets of GAC1 in steroid-resistant neutrophil-dominant asthma. Molecular docking was performed to predict binding modes between GAC1 and Dex with TNF-α. Results: The result of the study showed that chronic GAC1 treatment, significantly reduced pulmonary inflammation (P < 0.01-0.001 vs Sham) and airway neutrophilia (P < 0.01 vs Sham), inhibited TNF-α, IL-4 and IL-5 levels (P < 0.05-0.001 vs Sham). Acute Dex treatment reduced eosinophilic inflammation and IL-4, IL-5 levels, but had no effect on neutrophilia and TNF-α production. GAC1 treated H292 cells showed decreased MUC5AC gene expression and production of ROS (P < 0.001 vs stimulated/untreated cells). Molecular docking results showed binding energy of complex GAC1-TNF was -10.8 kcal/mol. Discussion: GAC1 may be a promising anti-asthma botanical drug for treatment of steroid-resistant asthma.

8.
Front Immunol ; 15: 1299484, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38380329

RESUMEN

Introduction: Peanut allergy is an immunoglobulin E (IgE) mediated food allergy. Rubia cordifolia L. (R. cordifolia), a Chinese herbal medicine, protects against peanut-induced anaphylaxis by suppressing IgE production in vivo. This study aims to identify IgE-inhibitory compounds from the water extract of R. cordifolia and investigate the underlying mechanisms using in vitro and in vivo models. Methods: Compounds were isolated from R. cordifolia water extract and their bioactivity on IgE production was assessed using a human myeloma U266 cell line. The purified active compound, xanthopurpurin (XPP), was identified by LC-MS and NMR. Peanut-allergic C3H/HeJ mice were orally administered with or without XPP at 200µg or 400µg per mouse per day for 4 weeks. Serum peanut-specific IgE levels, symptom scores, body temperatures, and plasma histamine levels were measured at challenge. Cytokines in splenocyte cultures were determined by ELISA, and IgE + B cells were analyzed by flow cytometry. Acute and sub-chronic toxicity were evaluated. IL-4 promoter DNA methylation, RNA-Seq, and qPCR analysis were performed to determine the regulatory mechanisms of XPP. Results: XPP significantly and dose-dependently suppressed the IgE production in U266 cells. XPP significantly reduced peanut-specific IgE (>80%, p <0.01), and plasma histamine levels and protected the mice against peanut-allergic reactions in both early and late treatment experiments (p < 0.05, n=9). XPP showed a strong protective effect even 5 weeks after discontinuing the treatment. XPP significantly reduced the IL-4 level without affecting IgG or IgA and IFN-γ production. Flow cytometry data showed that XPP reduced peripheral and bone marrow IgE + B cells compared to the untreated group. XPP increased IL-4 promoter methylation. RNA-Seq and RT-PCR experiments revealed that XPP regulated the gene expression of CCND1, DUSP4, SDC1, ETS1, PTPRC, and IL6R, which are related to plasma cell IgE production. All safety testing results were in the normal range. Conclusions: XPP successfully protected peanut-allergic mice against peanut anaphylaxis by suppressing IgE production. XPP suppresses murine IgE-producing B cell numbers and inhibits IgE production and associated genes in human plasma cells. XPP may be a potential therapy for IgE-mediated food allergy.


Asunto(s)
Anafilaxia , Hipersensibilidad a los Alimentos , Hipersensibilidad al Cacahuete , Ratones , Humanos , Animales , Hipersensibilidad al Cacahuete/terapia , Anafilaxia/prevención & control , Histamina , Interleucina-4 , Médula Ósea , Ratones Endogámicos C3H , Inmunoglobulina E , Agua
9.
Am J Physiol Lung Cell Mol Physiol ; 305(12): L1002-10, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24163140

RESUMEN

Our previous studies have shown that the anti-asthma traditional Chinese medicine herbal formula ASHMI (anti-asthma simplified herbal medicine intervention) inhibits acetylcholine-induced contractions of tracheal rings from ovalbumin-sensitized and naive mice in a ß-adrenoceptor-independent manner. We sought to determine whether acute in vivo ASHMI administration inhibits airway hyperreactivity (AHR) in a murine model of allergic asthma and acetylcholine-induced tracheal ring constriction ex vivo and to elucidate the cellular mechanisms underlying these effects. Ovalbumin-sensitized mice received a single oral ASHMI dose 2 h before intravenous acetylcholine challenge. AHR was determined by invasive airway measurements. Myography was used to determine the effects of ASHMI on acetylcholine-induced constriction of tracheal rings from asthmatic mice with or without epithelial denudation. The effect of cyclooxygenase inhibition and EP2/EP4 receptor blockade on ASHMI attenuation of acetylcholine contractions was evaluated. Tracheal cAMP and PGE2 levels were measured by ELISA. A single acute oral dose of ASHMI dramatically reduced AHR in response to acetylcholine provocation in ovalbumin-sensitized mice (P < 0.001). In ex vivo experiments, ASHMI significantly and dose-dependently reduced tracheal ring constriction to acetylcholine (P < 0.05-0.001), which was epithelium independent and associated with elevated cAMP levels. This effect was abrogated by cyclooxygenase inhibition or EP2/EP4 receptor blockade. ASHMI also inhibited contraction to high K(+) (P < 0.001). ASHMI increased tracheal ring PGE2 release in response to acetylcholine or high K(+) (P < 0.05 for both). ASHMI produced direct and acute inhibition of AHR in vivo and blocked acetylcholine-induced tracheal ring constriction via the EP2/EP4 receptor pathway, identifying the mechanism by which ASHMI is an orally active bronchoprotective agent.


Asunto(s)
Antiasmáticos/uso terapéutico , Dinoprostona/metabolismo , Medicamentos Herbarios Chinos/uso terapéutico , Contracción Muscular/efectos de los fármacos , Músculo Liso/metabolismo , Receptores de Prostaglandina E/metabolismo , Animales , Asma/tratamiento farmacológico , Medicamentos Herbarios Chinos/administración & dosificación , Medicina de Hierbas/métodos , Ratones , Músculo Liso/efectos de los fármacos , Ovalbúmina/farmacología , Tráquea/efectos de los fármacos
10.
Comput Biol Med ; 164: 107311, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37552916

RESUMEN

Chest or upper body auscultation has long been considered a useful part of the physical examination going back to the time of Hippocrates. However, it did not become a prevalent practice until the invention of the stethoscope by Rene Laennec in 1816, which made the practice suitable and hygienic. Pulmonary disease is a kind of sickness that affects the lungs and various parts of the respiratory system. Lung diseases are the third largest cause of death in the world. According to the World Health Organization (WHO), the five major respiratory diseases, namely chronic obstructive pulmonary disease (COPD), tuberculosis, acute lower respiratory tract infection (LRTI), asthma, and lung cancer, cause the death of more than 3 million people each year worldwide. Respiratory sounds disclose significant information regarding the lungs of patients. Numerous methods are developed for analyzing the lung sounds. However, clinical approaches require qualified pulmonologists to diagnose such kind of signals appropriately and are also time consuming. Hence, an efficient Fractional Water Cycle Swarm Optimizer-based Deep Residual Network (Fr-WCSO-based DRN) is developed in this research for detecting the pulmonary abnormalities using respiratory sounds signals. The proposed Fr-WCSO is newly designed by the incorporation of Fractional Calculus (FC) and Water Cycle Swarm Optimizer WCSO. Meanwhile, WCSO is the combination of Water Cycle Algorithm (WCA) with Competitive Swarm Optimizer (CSO). The respiratory input sound signals are pre-processed and the important features needed for the further processing are effectively extracted. With the extracted features, data augmentation is carried out for minimizing the over fitting issues for improving the overall detection performance. Once data augmentation is done, feature selection is performed using proposed Fr-WCSO algorithm. Finally, pulmonary abnormality detection is performed using DRN where the training procedure of DRN is performed using the developed Fr-WCSO algorithm. The developed method achieved superior performance by considering the evaluation measures, namely True Positive Rate (TPR), True Negative Rate (TNR) and testing accuracy with the values of 0.963(96.3%), 0.932,(93.2%) and 0.948(94.8%), respectively.


Asunto(s)
Asma , Enfermedad Pulmonar Obstructiva Crónica , Humanos , Ruidos Respiratorios/diagnóstico , Pulmón , Enfermedad Pulmonar Obstructiva Crónica/diagnóstico , Asma/diagnóstico , Auscultación/métodos
11.
Front Immunol ; 14: 1174907, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37575233

RESUMEN

Background: Gut microbiota influence food allergy. We showed that the natural compound berberine reduces IgE and others reported that BBR alters gut microbiota implying a potential role for microbiota changes in BBR function. Objective: We sought to evaluate an oral Berberine-containing natural medicine with a boiled peanut oral immunotherapy (BNP) regimen as a treatment for food allergy using a murine model and to explore the correlation of treatment-induced changes in gut microbiota with therapeutic outcomes. Methods: Peanut-allergic (PA) mice, orally sensitized with roasted peanut and cholera toxin, received oral BNP or control treatments. PA mice received periodic post-therapy roasted peanut exposures. Anaphylaxis was assessed by visualization of symptoms and measurement of body temperature. Histamine and serum peanut-specific IgE levels were measured by ELISA. Splenic IgE+B cells were assessed by flow cytometry. Fecal pellets were used for sequencing of bacterial 16S rDNA by Illumina MiSeq. Sequencing data were analyzed using built-in analysis platforms. Results: BNP treatment regimen induced long-term tolerance to peanut accompanied by profound and sustained reduction of IgE, symptom scores, plasma histamine, body temperature, and number of IgE+ B cells (p <0.001 vs Sham for all). Significant differences were observed for Firmicutes/Bacteroidetes ratio across treatment groups. Bacterial genera positively correlated with post-challenge histamine and PN-IgE included Lachnospiraceae, Ruminococcaceae, and Hydrogenanaerobacterium (all Firmicutes) while Verrucromicrobiacea. Caproiciproducens, Enterobacteriaceae, and Bacteroidales were negatively correlated. Conclusions: BNP is a promising regimen for food allergy treatment and its benefits in a murine model are associated with a distinct microbiota signature.


Asunto(s)
Berberina , Hipersensibilidad a los Alimentos , Microbiota , Hipersensibilidad al Cacahuete , Ratones , Animales , Arachis , Hipersensibilidad al Cacahuete/diagnóstico , Berberina/farmacología , Berberina/uso terapéutico , Histamina , Modelos Animales de Enfermedad , Desensibilización Inmunológica , Inmunoglobulina E
12.
Front Immunol ; 14: 1081121, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36825019

RESUMEN

Introduction: Food allergy is a significant public health problem with limited treatment options. As Food Allergy Herbal Formula 2 (FAHF-2) showed potential as a food allergy treatment, we further developed a purified version named EBF-2 and identified active compounds. We investigated the mechanisms of EBF-2 on IgE-mediated peanut (PN) allergy and its active compound, berberine, on IgE production. Methods: IgE plasma cell line U266 cells were cultured with EBF-2 and FAHF-2, and their effects on IgE production were compared. EBF-2 was evaluated in a murine PN allergy model for its effect on PN-specific IgE production, number of IgE+ plasma cells, and PN anaphylaxis. Effects of berberine on IgE production, the expression of transcription factors, and mitochondrial glucose metabolism in U266 cells were evaluated. Results: EBF-2 dose-dependently suppressed IgE production and was over 16 times more potent than FAHF-2 in IgE suppression in U266 cells. EBF-2 significantly suppressed PN-specific IgE production (70%, p<0.001) and the number of IgE-producing plasma cells in PN allergic mice, accompanied by 100% inhibition of PN-induced anaphylaxis and plasma histamine release (p<0.001) without affecting IgG1 or IgG2a production. Berberine markedly suppressed IgE production, which was associated with suppression of XBP1, BLIMP1, and STAT6 transcription factors and a reduced rate of mitochondrial oxidation in an IgE-producing plasma cell line. Conclusions: EBF-2 and its active compound berberine are potent IgE suppressors, associated with cellular regulation of immunometabolism on IgE plasma cells, and may be a potential therapy for IgE-mediated food allergy and other allergic disorders.


Asunto(s)
Anafilaxia , Berberina , Hipersensibilidad a los Alimentos , Hipersensibilidad al Cacahuete , Ratones , Animales , Inmunoglobulina E , Anafilaxia/prevención & control , Berberina/farmacología , Berberina/uso terapéutico , Interferón gamma/metabolismo , Hipersensibilidad a los Alimentos/tratamiento farmacológico , Inmunoglobulina G , Factores de Transcripción
13.
Front Endocrinol (Lausanne) ; 14: 1054674, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37033234

RESUMEN

Goals: To assess the efficacy and safety of Chinese Medicine Prescription "W-LHIT" in subjects with simple obesity, and to explore its potential mechanism of action. Methods: Thirty-seven patients aged 18 to 60 from Wei-En hospital (Weifang City, Shandong, China), participated in a double blinded, placebo-controlled study. Subjects were randomly divided into 2 groups, 18 in treatment and 19 in placebo group. The treatment group took the "W-LHIT" capsules for two months, while the control group received placebo capsules. Both groups accepted healthy lifestyle education materials. After a 2-month treatment, the placebo group transferred to open-label treatment after unblinding. Results: 72.22% participants in the treatment group lost more than 5% of their body weight, compared with 36.84% in the placebo group (p < 0.001). Body weight loss and body mass index reduction of the treatment group were also significantly higher than those of the placebo group (p < 0.05). These changes were accompanied by increased abundance of Akkermansia muciniphila and Enterococcus faecium, and decreased abundance of Proteobacteria in gut microbiota. Furthermore, the treatment group also showed improvement in obesity-related comorbidities such as hypertension and elevation of liver enzymes. No serious adverse reactions were found during the study period. Weight did not rebound at a follow-up visit 2 months after treatment. Conclusion: W-LHIT significantly improved body weight and comorbid conditions without obvious adverse reaction or rebound weight gain. These effects were associated with increased abundance of probiotics in gut microbiota. W-LHIT may have a potential for treating obesity in conjunction with healthy lifestyle modifications.


Asunto(s)
Microbioma Gastrointestinal , Humanos , Obesidad/complicaciones , Obesidad/tratamiento farmacológico , Pérdida de Peso , Resultado del Tratamiento , Estilo de Vida
14.
Ann Allergy Asthma Immunol ; 108(5): 351-358.e1, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22541407

RESUMEN

BACKGROUND: Food Allergy Herbal Formula-2 (FAHF-2) prevents anaphylaxis in a murine model of peanut allergy. Multiple food allergies (MFA) are common and associated with a higher risk of anaphylaxis. No well-characterized murine model of sensitization to multiple food allergens exists, and no satisfactory therapy for MFA is currently available. OBJECTIVE: To determine the effect of FAHF-2 in a murine model of MFA. METHODS: C3H/HeJ mice were orally sensitized to peanut, codfish, and egg concurrently. Oral FAHF-2 treatment commenced 1 day after completing sensitization and continued daily for 7 weeks. Mice were subsequently orally challenged with each allergen. RESULTS: Antibodies in sera from mice simultaneously sensitized with peanut, codfish, and egg recognized major allergens of all 3 foods, demonstrating sensitization to multiple unrelated food allergens (MFA mice). Sham-treated MFA mice exhibited anaphylactic symptoms accompanied by elevation of plasma histamine and hypothermia. In contrast, FAHF-2-treated MFA mice showed no anaphylactic symptoms, normal body temperature, and histamine levels after challenge with each allergen. Protection was accompanied by reduction in allergen-specific immunoglobulin E levels. Allergen-stimulated Th2 cytokine interleukin-4 and interleukin-13 production levels decreased, whereas the Th1 cytokine interferon-γ levels were elevated in cultured splenocytes and mesenteric lymph node cells in FAHF-2-treated mice. CONCLUSION: We established the first murine model of MFA. FAHF-2 prevents peanut, egg, and fish-induced anaphylactic reactions in this model, suggesting that FAHF-2 may have potential for treating human MFA.


Asunto(s)
Anafilaxia/prevención & control , Hipersensibilidad al Huevo/prevención & control , Peces/inmunología , Hipersensibilidad a los Alimentos/prevención & control , Hipersensibilidad al Cacahuete/prevención & control , Extractos Vegetales/uso terapéutico , Alérgenos/administración & dosificación , Alérgenos/efectos adversos , Alérgenos/inmunología , Anafilaxia/etiología , Anafilaxia/inmunología , Animales , Arachis/inmunología , Modelos Animales de Enfermedad , Hipersensibilidad al Huevo/inmunología , Femenino , Hipersensibilidad a los Alimentos/etiología , Hipersensibilidad a los Alimentos/inmunología , Humanos , Ratones , Ratones Endogámicos C3H , Hipersensibilidad al Cacahuete/inmunología , Extractos Vegetales/inmunología , Resultado del Tratamiento
15.
Comput Biol Med ; 150: 106123, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36228465

RESUMEN

The recent investigation has started for evaluating the human respiratory sounds, like voice recorded, cough, and breathing from hospital confirmed Covid-19 tools, which differs from healthy person's sound. The cough-based detection of Covid-19 also considered with non-respiratory and respiratory sounds data related with all declared situations. Covid-19 is respiratory disease, which is usually produced by Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2). However, it is more indispensable to detect the positive cases for reducing further spread of virus, and former treatment of affected patients. With constant rise in the COVID-19 cases, there has been a constant rise in the need of efficient and safe ways to detect an infected individual. With the cases multiplying constantly, the current detecting devices like RT-PCR and fast testing kits have become short in supply. An effectual Covid-19 detection model using devised hybrid Honey Badger Optimization-based Deep Neuro Fuzzy Network (HBO-DNFN) is developed in this paper. Here, the audio signal is considered as input for detecting Covid-19. The gaussian filter is applied to input signal for removing the noises and then feature extraction is performed. The substantial features, like spectral roll-off, spectral bandwidth, Mel frequency cepstral coefficients (MFCC), spectral flatness, zero crossing rate, spectral centroid, mean square energy and spectral contract are extracted for further processing. Finally, DNFN is applied for detecting Covid-19 and the deep leaning model is trained by designed hybrid HBO algorithm. Accordingly, the developed Hybrid HBO method is newly designed by incorporating Honey Badger optimization Algorithm (HBA) and Jaya algorithm. The performance of developed Covid-19 detection model is evaluated using three metrics, like testing accuracy, sensitivity and specificity. The developed Hybrid HBO-based DNFN is outpaced than other existing approaches in terms of testing accuracy, sensitivity and specificity of "0.9176, 0.9218 and 0. 9219". All the test results are validated with the k-fold cross validation method in order to make an assessment of the generalizability of these results. When k-fold value is 9, sensitivity of existing techniques and developed JHBO-based DNFN is 0.8982, 0.8816, 0.8938, and 0.9207. The sensitivity of developed approach is improved by means of gaussian filtering model. The specificity of DCNN is 0.9125, BI-AT-GRU is 0.8926, and XGBoost is 0.9014, while developed JHBO-based DNFN is 0.9219 in k-fold value 9.


Asunto(s)
COVID-19 , Aprendizaje Profundo , Mustelidae , Humanos , Animales , COVID-19/diagnóstico , SARS-CoV-2 , Tos , Ruidos Respiratorios
16.
J Inflamm Res ; 15: 5527-5540, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36176352

RESUMEN

Background: Eosinophilic Esophagitis (EoE) is an increasingly common chronic inflammatory disease. The pathological mechanisms underlying EoE are largely unknown. Objective: We sought to understand the mechanisms underlying aeroallergen-induced EoE in Sharpin gene deficient (Sharpin-/-) mice that is prone to inflammatory response. Methods: Sharpin-/-mice were exposed with Aspergillus fumigatus and ovalbumin intranasally every alternate day for 4 weeks. Wild type (WT) naïve mice, WT exposed, and un-exposed Sharpin-/- mice were controls. Histopathological analysis was performed by H&E, trichrome and major basic protein staining. Total and specific IgE, IgG, and IgA levels were measured by ELISA and Th2 cytokine and CCL11 chemokine gene expression were determined. Results: Airborne allergen exposed Sharpin-/- mice showed severe eosinophilic inflammation in the esophagus (p < 0.001), and markedly increased epithelial thickening (p < 0.0001) compared to WT normal controls, whereas airborne allergen exposed WT mice and unexposed Sharpin-/- mice only showed mild eosinophilic inflammation in the esophagus. These exposed Sharpin-/- mice also showed over 7-fold increase in blood eosinophils (p < 0.0001), 60-fold increase in eosinophils in bronchoalveolar lavage fluid (p < 0.0001) and 4-fold increase in eosinophils in the skin (p < 0.0001) compared to normal controls. Surprisingly, exposed Sharpin-/- mice did not show elevation of serum total or antigen-specific IgE levels but reduced total IgA and IgG levels than normal controls There was a marked increase in IL-4, IL-13 and CCL11 gene expression in esophageal tissue (p < 0.001) in exposed Sharpin-/- mice compared to WT normal mice. Conclusion: Th2 cytokines and chemokines, but not IgE may play an important pathologic role in aeroallergen-induced EoE. This study may provide insight into new therapeutics for EoE.

17.
J Asthma Allergy ; 15: 645-664, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35603013

RESUMEN

Purpose: Food allergy-induced autism-like behavior has been increasing for decades, but the causal drivers of this association are unclear. We sought to test the association of gut microbiota and mammalian/mechanistic target of rapamycin (mTOR) signaling with cow's milk allergy (CMA)-induced autism pathogenesis. Methods: Mice were sensitized intragastrically with whey protein containing cholera toxin before sensitization on intraperitoneal injection with whey-containing alum, followed by intragastric allergen challenge to induce experimental CMA. The food allergic immune responses, ASD-like behavioral tests and changes in the mTOR signaling pathway and gut microbial community structure were performed. Results: CMA mice showed autism-like behavioral abnormalities and several distinct biomarkers. These include increased levels of 5-hydroxymethylcytosine (5-hmC) in the hypothalamus; c-Fos were predominantly located in the region of the lateral orbital prefrontal cortex (PFC), but not ventral; decreased serotonin 1A in amygdala and PFC. CMA mice exhibited a specific microbiota signature characterized by coordinate changes in the abundance of taxa of several bacterial genera, including the Lactobacillus. Interestingly, the changes were accompanied by promoted mTOR signaling in the brain of CMA mice. Conclusion: We found that disease-associated microbiota and mTOR activation may thus play a pathogenic role in the intestinal, immunological, and psychiatric Autism Spectrum Disorder (ASD)-like symptoms seen in CAM associated autism. However, this is only a preliminary study, and their mechanisms require further investigation.

18.
Front Pharmacol ; 13: 1042756, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36793921

RESUMEN

Introduction: Cytochrome P450 (CYP) 3A4 is a major drug metabolizing enzyme for corticosteroids (CS). Epimedium has been used for asthma and variety of inflammatory conditions with or without CS. It is unknown whether epimedium has an effect on CYP 3A4 and how it interacts with CS. We sought to determine the effects of epimedium on CYP3A4 and whether it affects the anti-inflammatory function of CS and identify the active compound responsible for this effect. Methods: The effect of epimedium on CYP3A4 activity was evaluated using the Vivid CYP high-throughput screening kit. CYP3A4 mRNA expression was determined in human hepatocyte carcinoma (HepG2) cells with or without epimedium, dexamethasone, rifampin, and ketoconazole. TNF-α levels were determined following co-culture of epimedium with dexamethasone in a murine macrophage cell line (Raw 264.7). Active compound (s) derived from epimedium were tested on IL-8 and TNF-α production with or without corticosteroid, on CYP3A4 function and binding affinity. Results: Epimedium inhibited CYP3A4 activity in a dose-dependent manner. Dexamethasone enhanced the expression of CYP3A4 mRNA, while epimedium inhibited the expression of CYP3A4 mRNA and further suppressed dexamethasone enhancement of CYP3A4 mRNA expression in HepG2 cells (p < 0.05). Epimedium and dexamethasone synergistically suppressed TNF-α production by RAW cells (p < 0.001). Eleven epimedium compounds were screened by TCMSP. Among the compounds identified and tested only kaempferol significantly inhibited IL-8 production in a dose dependent manner without any cell cytotoxicity (p < 0.01). Kaempferol in combination with dexamethasone showed complete elimination of TNF-α production (p < 0.001). Furthermore, kaempferol showed a dose dependent inhibition of CYP3A4 activity. Computer docking analysis showed that kaempferol significantly inhibited the catalytic activity of CYP3A4 with a binding affinity of -44.73kJ/mol. Discussion: Inhibition of CYP3A4 function by epimedium and its active compound kaempferol leads to enhancement of CS anti-inflammatory effect.

19.
J Allergy Clin Immunol ; 126(6): 1208-17.e3, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21134573

RESUMEN

BACKGROUND: Mast cells and basophils are key effector cells of IgE-mediated anaphylactic reactions. The Chinese herbal formula, food allergy herbal formula 2 (FAHF-2), protects against peanut anaphylaxis in mice. However, the mechanisms underlying this effect are not fully elucidated. OBJECTIVE: To investigate whether FAHF-2 inhibits mast cell/basophil numbers and IgE-mediated activation. METHODS: Mice with peanut allergy (PNA mice) were treated with FAHF-2 intragastrically for 7 weeks and challenged intragastrically with peanut 1 day and 4 weeks posttreatment. Peripheral blood basophil numbers and peritoneal mast cell numbers and FcεRI expression were determined. Direct effects of FAHF-2 on the murine mast cell line MC/9, and effects of 4 fractions and 3 compounds isolated from FAHF-2 on rat basophilic leukemia cells (RBL-2H3) and human skin mast cells degranulation and on the IgE-mediated spleen tyrosine kinase signaling pathway, were determined. RESULTS: Although all sham-treated PNA mice developed anaphylaxis, FAHF-2-treated PNA mice were protected against anaphylaxis after peanut challenge at 1 day and 4 weeks posttherapy. Reduction of peripheral blood basophils began after 1 week of treatment and continued for at least 4 weeks posttherapy. The number and FcεRI expression of peritoneal mast cells were also significantly decreased 4 weeks posttherapy. FAHF-2-treated MC/9 cells showed significantly reduced IgE-induced FcεRI expression, FcεRI γ mRNA subunit expression, proliferation, and histamine release on challenge. Fraction 2 from FAHF-2 inhibited RBL-2H3 cell and human mast cell degranulation. Three compounds from fraction 2-berberine, palmatine, and jatrorrhizine-inhibited RBL-2H3 cell degranulation via suppressing spleen tyrosine kinase phosphorylation. CONCLUSION: Food allergy herbal formula 2 reduction of basophils and mast cell numbers as well as suppression of IgE-mediated mast cell activation may contribute to FAHF-2's persistent protection against peanut anaphylaxis.


Asunto(s)
Basófilos/efectos de los fármacos , Hipersensibilidad a los Alimentos/tratamiento farmacológico , Hipersensibilidad a los Alimentos/inmunología , Mastocitos/efectos de los fármacos , Extractos Vegetales/administración & dosificación , Anafilaxia/prevención & control , Animales , Arachis/efectos adversos , Basófilos/inmunología , Basófilos/metabolismo , Basófilos/patología , Recuento de Células , Degranulación de la Célula/efectos de los fármacos , Degranulación de la Célula/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Hipersensibilidad a los Alimentos/patología , Hipersensibilidad a los Alimentos/fisiopatología , Humanos , Inmunoglobulina E/inmunología , Inmunoglobulina E/metabolismo , Mastocitos/inmunología , Mastocitos/metabolismo , Mastocitos/patología , Ratones , Ratones Endogámicos C3H , Extractos Vegetales/farmacología , Ratas , Receptores de IgE/genética , Receptores de IgE/metabolismo
20.
Artículo en Inglés | MEDLINE | ID: mdl-34055023

RESUMEN

Eczema is a complex chronic inflammatory skin disease impacted by environmental factors, infections, immune disorders, and deficiencies in skin barrier function. Shi Zhen Tea (SZT), derived from traditional Chinese medicine Xiao-Feng-San, has shown to be an effective integrative therapy for treating skin lesions, itching, and sleeping loss, and it facilitates reduction of topical steroid and antihistamine use in pediatric and adult patients with severe eczema. Yet, its active compounds and therapeutic mechanisms have not been elucidated. In this study, we sought to investigate the active compounds and molecular mechanisms of SZT in treating eczema using systems pharmacology and in silico docking analysis. SZT is composed of 4 medicinal herbs, Baizhu (Atractylodis macrocephalae rhizome), Jingjie (Schizonepetae herba), Kushen (Sophorae flavescentis radix), and Niubangzi (Arctii fructus). We first identified 51 active compounds from SZT and their 81 potential molecular targets by high-throughput computational analysis, from which we identified 4 major pathways including Th17 cell differentiation, metabolic pathways, pathways in cancer, and the PI3K-Akt signaling pathway. Through network analysis of the compound-target pathway, we identified hub molecular targets within these pathways including carbonic anhydrase II (CA2), peroxisome proliferator activated receptor γ (PPAR γ), retinoid X receptor α (RXRA), and vitamin D receptor (VDR). We further identified top 5 compounds including cynarine, stigmasterin, kushenol, ß-sitosterol, and (24S)-24-propylcholesta-5-ene-3ß-ol as putative key active compounds on the basis of their molecular docking scores with identified hub target proteins. Our study provides an insight into the therapeutic mechanism underlying multiscale benefits of SZT for eczema and paves the way for developing new and potentially more effective eczema therapies.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA