Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 180(9): 1197-1209, 2023 05.
Article in English | MEDLINE | ID: mdl-36470846

ABSTRACT

BACKGROUND AND PURPOSE: In recent decades, a focus on the most critical and fundamental concepts has proven highly advantageous to students and educators in many science disciplines. Pharmacology, unlike microbiology, biochemistry, or physiology, lacks a consensus list of such core concepts. EXPERIMENTAL APPROACH: We sought to develop a research-based, globally relevant list of core concepts that all students completing a foundational pharmacology course should master. This two-part project consisted of exploratory and refinement phases. The exploratory phase involved empirical data mining of the introductory sections of five key textbooks, in parallel with an online survey of over 200 pharmacology educators from 17 countries across six continents. The refinement phase involved three Delphi rounds involving 24 experts from 15 countries across six continents. KEY RESULTS: The exploratory phase resulted in a consolidated list of 74 candidate core concepts. In the refinement phase, the expert group produced a consensus list of 25 core concepts of pharmacology. CONCLUSION AND IMPLICATIONS: This list will allow pharmacology educators everywhere to focus their efforts on the conceptual knowledge perceived to matter most by experts within the discipline. Next steps for this project include defining and unpacking each core concept and developing resources to help pharmacology educators globally teach and assess these concepts within their educational contexts.

2.
J Biol Chem ; 298(10): 102325, 2022 10.
Article in English | MEDLINE | ID: mdl-35926710

ABSTRACT

Neurite outgrowth is an integrated whole cell response triggered by the cannabinoid-1 receptor. We sought to identify the many different biochemical pathways that contribute to this whole cell response. To understand underlying mechanisms, we identified subcellular processes (SCPs) composed of one or more biochemical pathways and their interactions required for this response. Differentially expressed genes and proteins were obtained from bulk transcriptomics and proteomic analysis of extracts from cells stimulated with a cannabinoid-1 receptor agonist. We used these differentially expressed genes and proteins to build networks of interacting SCPs by combining the expression data with prior pathway knowledge. From these SCP networks, we identified additional genes that when ablated, experimentally validated the SCP involvement in neurite outgrowth. Our experiments and informatics modeling allowed us to identify diverse SCPs such as those involved in pyrimidine metabolism, lipid biosynthesis, and mRNA splicing and stability, along with more predictable SCPs such as membrane vesicle transport and microtubule dynamics. We find that SCPs required for neurite outgrowth are widely distributed among many biochemical pathways required for constitutive cellular functions, several of which are termed 'deep', since they are distal to signaling pathways and the key SCPs directly involved in extension of the neurite. In contrast, 'proximal' SCPs are involved in microtubule growth and membrane vesicle transport dynamics required for neurite outgrowth. From these bioinformatics and dynamical models based on experimental data, we conclude that receptor-mediated regulation of subcellular functions for neurite outgrowth is both distributed, that is, involves many different biochemical pathways, and deep.


Subject(s)
Cannabinoid Receptor Agonists , Neurites , Neuronal Outgrowth , Proteomics , Receptor, Cannabinoid, CB1 , Neurites/drug effects , Neurites/metabolism , Neuronal Outgrowth/drug effects , Signal Transduction , Receptor, Cannabinoid, CB1/metabolism , Cannabinoid Receptor Agonists/pharmacology , Humans
3.
Sci Data ; 9(1): 18, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35058449

ABSTRACT

Drug Toxicity Signature Generation Center (DToxS) at the Icahn School of Medicine at Mount Sinai is one of the centers for the NIH Library of Integrated Network-Based Cellular Signatures (LINCS) program. Its key aim is to generate proteomic and transcriptomic signatures that can predict cardiotoxic adverse effects of kinase inhibitors approved by the Food and Drug Administration. Towards this goal, high throughput shotgun proteomics experiments (308 cell line/drug combinations +64 control lysates) have been conducted. Using computational network analyses, these proteomic data can be integrated with transcriptomic signatures, generated in tandem, to identify cellular signatures of cardiotoxicity that may predict kinase inhibitor-induced toxicity and enable possible mitigation. Both raw and processed proteomics data have passed several quality control steps and been made publicly available on the PRIDE database. This broad protein kinase inhibitor-stimulated human cardiomyocyte proteomic data and signature set is valuable for prediction of drug toxicities.


Subject(s)
Antineoplastic Agents , Proteomics , Antineoplastic Agents/pharmacology , Cardiotoxicity , Humans , Protein Kinase Inhibitors/adverse effects , Transcriptome
4.
J Virol ; 96(2): e0106321, 2022 01 26.
Article in English | MEDLINE | ID: mdl-34669512

ABSTRACT

COVID-19 affects multiple organs. Clinical data from the Mount Sinai Health System show that substantial numbers of COVID-19 patients without prior heart disease develop cardiac dysfunction. How COVID-19 patients develop cardiac disease is not known. We integrated cell biological and physiological analyses of human cardiomyocytes differentiated from human induced pluripotent stem cells (hiPSCs) infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the presence of interleukins (ILs) with clinical findings related to laboratory values in COVID-19 patients to identify plausible mechanisms of cardiac disease in COVID-19 patients. We infected hiPSC-derived cardiomyocytes from healthy human subjects with SARS-CoV-2 in the absence and presence of IL-6 and IL-1ß. Infection resulted in increased numbers of multinucleated cells. Interleukin treatment and infection resulted in disorganization of myofibrils, extracellular release of troponin I, and reduced and erratic beating. Infection resulted in decreased expression of mRNA encoding key proteins of the cardiomyocyte contractile apparatus. Although interleukins did not increase the extent of infection, they increased the contractile dysfunction associated with viral infection of cardiomyocytes, resulting in cessation of beating. Clinical data from hospitalized patients from the Mount Sinai Health System show that a significant portion of COVID-19 patients without history of heart disease have elevated troponin and interleukin levels. A substantial subset of these patients showed reduced left ventricular function by echocardiography. Our laboratory observations, combined with the clinical data, indicate that direct effects on cardiomyocytes by interleukins and SARS-CoV-2 infection might underlie heart disease in COVID-19 patients. IMPORTANCE SARS-CoV-2 infects multiple organs, including the heart. Analyses of hospitalized patients show that a substantial number without prior indication of heart disease or comorbidities show significant injury to heart tissue, assessed by increased levels of troponin in blood. We studied the cell biological and physiological effects of virus infection of healthy human iPSC-derived cardiomyocytes in culture. Virus infection with interleukins disorganizes myofibrils, increases cell size and the numbers of multinucleated cells, and suppresses the expression of proteins of the contractile apparatus. Viral infection of cardiomyocytes in culture triggers release of troponin similar to elevation in levels of COVID-19 patients with heart disease. Viral infection in the presence of interleukins slows down and desynchronizes the beating of cardiomyocytes in culture. The cell-level physiological changes are similar to decreases in left ventricular ejection seen in imaging of patients' hearts. These observations suggest that direct injury to heart tissue by virus can be one underlying cause of heart disease in COVID-19.


Subject(s)
COVID-19/immunology , Induced Pluripotent Stem Cells , Interleukin-10/immunology , Interleukin-1beta/immunology , Interleukin-6/immunology , Myocytes, Cardiac , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/immunology , Induced Pluripotent Stem Cells/pathology , Induced Pluripotent Stem Cells/virology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , Myocytes, Cardiac/virology
5.
Stem Cell Reports ; 16(12): 3036-3049, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34739849

ABSTRACT

A library of well-characterized human induced pluripotent stem cell (hiPSC) lines from clinically healthy human subjects could serve as a useful resource of normal controls for in vitro human development, disease modeling, genotype-phenotype association studies, and drug response evaluation. We report generation and extensive characterization of a gender-balanced, racially/ethnically diverse library of hiPSC lines from 40 clinically healthy human individuals who range in age from 22 to 61 years. The hiPSCs match the karyotype and short tandem repeat identities of their parental fibroblasts, and have a transcription profile characteristic of pluripotent stem cells. We provide whole-genome sequencing data for one hiPSC clone from each individual, genomic ancestry determination, and analysis of mendelian disease genes and risks. We document similar transcriptomic profiles, single-cell RNA-sequencing-derived cell clusters, and physiology of cardiomyocytes differentiated from multiple independent hiPSC lines. This extensive characterization makes this hiPSC library a valuable resource for many studies on human biology.


Subject(s)
Health , Induced Pluripotent Stem Cells/cytology , Adult , Calcium Signaling , Cell Differentiation , Cell Line , Clone Cells , Ethnicity , Female , Gene Expression Profiling , Gene Expression Regulation , Genetic Predisposition to Disease , Genetic Variation , Heart Atria/cytology , Heart Ventricles/cytology , Humans , Male , Middle Aged , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Risk Factors , Young Adult
6.
medRxiv ; 2020 Nov 16.
Article in English | MEDLINE | ID: mdl-33200140

ABSTRACT

COVID-19 affects multiple organs. Clinical data from the Mount Sinai Health System shows that substantial numbers of COVID-19 patients without prior heart disease develop cardiac dysfunction. How COVID-19 patients develop cardiac disease is not known. We integrate cell biological and physiological analyses of human cardiomyocytes differentiated from human induced pluripotent stem cells (hiPSCs) infected with SARS-CoV-2 in the presence of interleukins, with clinical findings, to investigate plausible mechanisms of cardiac disease in COVID-19 patients. We infected hiPSC-derived cardiomyocytes, from healthy human subjects, with SARS-CoV-2 in the absence and presence of interleukins. We find that interleukin treatment and infection results in disorganization of myofibrils, extracellular release of troponin-I, and reduced and erratic beating. Although interleukins do not increase the extent, they increase the severity of viral infection of cardiomyocytes resulting in cessation of beating. Clinical data from hospitalized patients from the Mount Sinai Health system show that a significant portion of COVID-19 patients without prior history of heart disease, have elevated troponin and interleukin levels. A substantial subset of these patients showed reduced left ventricular function by echocardiography. Our laboratory observations, combined with the clinical data, indicate that direct effects on cardiomyocytes by interleukins and SARS-CoV-2 infection can underlie the heart disease in COVID-19 patients.

7.
Nat Commun ; 11(1): 4809, 2020 09 23.
Article in English | MEDLINE | ID: mdl-32968055

ABSTRACT

Kinase inhibitors (KIs) represent an important class of anti-cancer drugs. Although cardiotoxicity is a serious adverse event associated with several KIs, the reasons remain poorly understood, and its prediction remains challenging. We obtain transcriptional profiles of human heart-derived primary cardiomyocyte like cell lines treated with a panel of 26 FDA-approved KIs and classify their effects on subcellular pathways and processes. Individual cardiotoxicity patient reports for these KIs, obtained from the FDA Adverse Event Reporting System, are used to compute relative risk scores. These are then combined with the cell line-derived transcriptomic datasets through elastic net regression analysis to identify a gene signature that can predict risk of cardiotoxicity. We also identify relationships between cardiotoxicity risk and structural/binding profiles of individual KIs. We conclude that acute transcriptomic changes in cell-based assays combined with drug substructures are predictive of KI-induced cardiotoxicity risk, and that they can be informative for future drug discovery.


Subject(s)
Cardiotoxicity/genetics , Cardiotoxicity/metabolism , Gene Expression Profiling/methods , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacology , Transcriptome , Antineoplastic Agents/pharmacology , Cardiotoxicity/drug therapy , Cell Line , Dose-Response Relationship, Drug , Drug Approval , Female , Gene Expression/drug effects , Humans , Male , Myocytes, Cardiac/drug effects , Regression Analysis , Risk Assessment , Risk Factors , Sequence Alignment , United States , United States Food and Drug Administration
8.
Mol Oncol ; 13(8): 1725-1743, 2019 08.
Article in English | MEDLINE | ID: mdl-31116490

ABSTRACT

The ability to predict responsiveness to drugs in individual patients is limited. We hypothesized that integrating molecular information from databases would yield predictions that could be experimentally tested to develop transcriptomic signatures for specific drugs. We analyzed lung adenocarcinoma patient data from The Cancer Genome Atlas and identified a subset of patients in which xanthine dehydrogenase (XDH) expression correlated with decreased survival. We tested allopurinol, an FDA-approved drug that inhibits XDH, on human non-small-cell lung cancer (NSCLC) cell lines obtained from the Broad Institute Cancer Cell Line Encyclopedia and identified sensitive and resistant cell lines. We utilized the transcriptomic profiles of these cell lines to identify six-gene signatures for allopurinol-sensitive and allopurinol-resistant cell lines. Transcriptomic networks identified JAK2 as an additional target in allopurinol-resistant lines. Treatment of resistant cell lines with allopurinol and CEP-33779 (a JAK2 inhibitor) resulted in cell death. The effectiveness of allopurinol alone or allopurinol and CEP-33779 was verified in vivo using tumor formation in NCR-nude mice. We utilized the six-gene signatures to predict five additional allopurinol-sensitive NSCLC cell lines and four allopurinol-resistant cell lines susceptible to combination therapy. We searched the transcriptomic data from a library of patient-derived NSCLC tumors from the Jackson Laboratory to identify tumors that would be predicted to be sensitive to allopurinol or allopurinol + CEP-33779 treatment. Patient-derived tumors showed the predicted drug sensitivity in vivo. These data indicate that we can use integrated molecular information from cancer databases to predict drug responsiveness in individual patients and thus enable precision medicine.


Subject(s)
Allopurinol/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Genomics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Systems Analysis , Allopurinol/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Lung Neoplasms/pathology , Mice, Nude , Phenotype , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Triazoles/pharmacology , Triazoles/therapeutic use , Xenograft Model Antitumor Assays
9.
Essays Biochem ; 62(4): 487-500, 2018 10 26.
Article in English | MEDLINE | ID: mdl-30287586

ABSTRACT

Systems biology is an integrative discipline connecting the molecular components within a single biological scale and also among different scales (e.g. cells, tissues and organ systems) to physiological functions and organismal phenotypes through quantitative reasoning, computational models and high-throughput experimental technologies. Systems biology uses a wide range of quantitative experimental and computational methodologies to decode information flow from genes, proteins and other subcellular components of signaling, regulatory and functional pathways to control cell, tissue, organ and organismal level functions. The computational methods used in systems biology provide systems-level insights to understand interactions and dynamics at various scales, within cells, tissues, organs and organisms. In recent years, the systems biology framework has enabled research in quantitative and systems pharmacology and precision medicine for complex diseases. Here, we present a brief overview of current experimental and computational methods used in systems biology.


Subject(s)
Systems Biology/methods , Artificial Intelligence , Gene Regulatory Networks , High-Throughput Screening Assays , Models, Biological
10.
Int Immunopharmacol ; 27(2): 224-31, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26004313

ABSTRACT

Asthma is a heterogeneous airway inflammatory disease, which is associated with Th2 cytokine-driven inflammation and non-Th2, TNF-α mediated inflammation. Unlike Th2 mediated inflammation, TNF-α mediated asthma inflammation is generally insensitive to inhaled corticosteroids (ICS). ASHMITM, aqueous extract of three medicinal herbs-Ganoderma lucidum (G. lucidum), Sophora flavescens Ait (S. flavescens) and Glycyrrhiza uralensis Fischer (G. uralensis), showed a high safety profile and was clinically beneficial in asthma patients. It also suppresses both Th2 and TNF-α associated inflammation in murine asthma models. We previously determined that G. uralensis flavonoids are the key active compounds responsible for ASHMITM suppression of Th2 mediated inflammation. Until now, there are limited studies on anti-TNF-α compounds presented in ASHMITM. The objective of this study was to isolate and identify TNF-α inhibitory compounds in ASHMITM. Here we report that G. lucidum, but not the other two herbal extracts, S. flavescens or G. uralensis inhibited TNF-α production by murine macrophages; and that the methylene chloride (MC)-triterpenoid-enriched fraction, but not the polysaccharide-enriched fraction, contained the inhibitory compounds. Of the 15 triterpenoids isolated from the MC fraction, only ganoderic acid C1 (GAC1) significantly reduced TNF-α production by murine macrophages (RAW 264.7 cells) and peripheral blood mononuclear cells (PBMCs) from asthma patients. Inhibition was associated with down-regulation of NF-κB expression, and partial suppression of MAPK and AP-1 signaling pathways. Ganoderic acid C1 may have potential for treating TNF-α mediated inflammation in asthma and other inflammatory diseases.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Drugs, Chinese Herbal/chemistry , Reishi/chemistry , Triterpenes/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Apoptosis/drug effects , Asthma/metabolism , Cell Line , Cell Proliferation/drug effects , Cells, Cultured , Female , Glycyrrhiza uralensis/chemistry , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Sophora/chemistry , Transcription Factor AP-1/metabolism , Triterpenes/isolation & purification , Tumor Necrosis Factor-alpha/metabolism
11.
Sci Transl Med ; 5(206): 206ra140, 2013 Oct 09.
Article in English | MEDLINE | ID: mdl-24107779

ABSTRACT

Drugs are designed for therapy, but medication-related adverse events are common, and risk/benefit analysis is critical for determining clinical use. Rosiglitazone, an efficacious antidiabetic drug, is associated with increased myocardial infarctions (MIs), thus limiting its usage. Because diabetic patients are often prescribed multiple drugs, we searched for usage of a second drug ("drug B") in the Food and Drug Administration's Adverse Event Reporting System (FAERS) that could mitigate the risk of rosiglitazone ("drug A")-associated MI. In FAERS, rosiglitazone usage is associated with increased occurrence of MI, but its combination with exenatide significantly reduces rosiglitazone-associated MI. Clinical data from the Mount Sinai Data Warehouse support the observations from FAERS. Analysis for confounding factors using logistic regression showed that they were not responsible for the observed effect. Using cell biological networks, we predicted that the mitigating effect of exenatide on rosiglitazone-associated MI could occur through clotting regulation. Data we obtained from the db/db mouse model agreed with the network prediction. To determine whether polypharmacology could generally be a basis for adverse event mitigation, we analyzed the FAERS database for other drug combinations wherein drug B reduced serious adverse events reported with drug A usage such as anaphylactic shock and suicidality. This analysis revealed 19,133 combinations that could be further studied. We conclude that this type of crowdsourced approach of using databases like FAERS can help to identify drugs that could potentially be repurposed for mitigation of serious adverse events.


Subject(s)
Drug Combinations , Drug-Related Side Effects and Adverse Reactions/prevention & control , Systems Biology , Adverse Drug Reaction Reporting Systems , Animals , Blood Coagulation/drug effects , Confounding Factors, Epidemiologic , Databases as Topic , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/physiopathology , Drug Interactions , Drug-Related Side Effects and Adverse Reactions/physiopathology , Exenatide , Heart Function Tests , Humans , Mice , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/etiology , Myocardial Infarction/physiopathology , Peptides/adverse effects , Peptides/therapeutic use , Plasminogen Activator Inhibitor 1/metabolism , Rosiglitazone , Thiazolidinediones/adverse effects , Thiazolidinediones/therapeutic use , Thrombelastography , Ultrasonography , United States , United States Food and Drug Administration , Venoms/adverse effects , Venoms/therapeutic use
12.
Phytochemistry ; 95: 259-267, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23993294

ABSTRACT

Asthma is a serious health problem worldwide, particularly in industrialized countries. Despite a better understanding of the pathophysiology of asthma, there are still considerable gaps in knowledge as well as a need for classes of drugs. ASHMI™ (Anti-asthma Herbal Medicine Intervention) is an aqueous extract of Ganoderma lucidum (Fr.) P. Karst (Ling Zhi), Sophora flavescens Aiton (Ku Shen) and Glycyrrhiza uralensis Fisch. ex DC (Gan Cao). It prevents allergic asthma airway hyper-reactivity in mice and inhibits acetylcholine (ACh) induced airway smooth muscle (ASM) contraction in tracheal rings from allergic asthmatic mice. The purpose of this research was to identify individual herb(s) and their active compound(s) that inhibit ASM contraction. It was found that S. flavescens, but not G. lucidum or G. uralensis aqueous extracts, inhibited ASM contraction in tracheal rings from asthmatic mice. Bioassay-guided isolation and identification of flavonoid fractions/compound(s) via methylene chloride extraction, preparative HPLC fractionation, and LC-MS and NMR spectroscopic analyses showed that trifolirhizin is an active constituent that inhibits acetylcholine mediated ASM contraction or directly relaxes pre-contracted ASM independent of ß2-adrenoceptors.


Subject(s)
Asthma/physiopathology , Flavonoids/pharmacology , Glucosides/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Sophora/chemistry , Trachea/drug effects , Acetylcholine/metabolism , Animals , Asthma/drug therapy , Asthma/metabolism , Ganoderma , Glycyrrhiza , Mice , Phytotherapy , Plant Extracts/pharmacology
13.
J Integr Med ; 11(3): 195-205, 2013 May.
Article in English | MEDLINE | ID: mdl-23743163

ABSTRACT

OBJECTIVE: Anti-asthma herbal medicine intervention (ASHMI(TM)), a combination of three traditional Chinese medicinal herbs developed in our laboratory, has demonstrated efficacy in both mouse models of allergic asthma, and a double-blind placebo-controlled clinical trial in patients with asthma. This study was designed to determine if the anti-inflammatory effects of individual herbal constituents of ASHMI(TM) exhibited synergy. METHODS: Effects of ASHMI and its components aqueous extracts of Lingzhi (Ganoderma lucidum), Kushen (Sophora flavescens) and Gancao (Glycyrrhiza uralensis), on Th2 cytokine secretion by murine memory Th2 cells (D10.G4.1) and eotaxin-1 secretion by human lung fibroblast (HLF-1) cells were determined by measuring levels in culture supernatants by enzyme-linked immunosorbent assay. Potential synergistic effects were determined by computing interaction indices from concentration-effect curve parameters. RESULTS: Individual Lingzhi, Kushen and Gancao extracts and ASHMI (the combination of individual extracts) inhibited production of interleukin (IL)-4 and IL-5 by murine memory Th2 cells and eotaxin-1 production by HLF-1 cells. The mean 25%-inhibitory-concentration (IC25) values (mg/mL) for ASHMI, Lingzhi, Kushen and Gancao for IL-4 production were 30.9, 79.4, 123, and 64.6, respectively; for IL-5 production were 30.2, 263, 123.2 and 100, respectively; for eotaxin-1 were 13.2, 16.2, 30.2, and 25.1, respectively. The IC50 values (mg/mL) for ASHMI, Lingzhi, Kushen and Gancao for IL-4 production were 158.5, 239.9, 446.7, and 281.8, respectively; for eotaxin-1 were 38.1, 33.1, 100, and 158.5, respectively. The interaction indices of ASHMI constituents at IC25 were 0.35 for IL-4, 0.21 for IL-5 and 0.59 for eotaxin-1. The interaction indices at IC50 values were 0.50 for IL-4 and 0.62 for eotaxin-1 inhibition. Inhibition of IL-5 did not reach IC50 values. All interaction indices were below 1 which indicated synergy. CONCLUSION: By comparing the interaction index values, we find that constituents in ASHMI(TM) synergistically inhibited eotaxin-1 production as well as Th2 cytokine production.


Subject(s)
Asthma/drug therapy , Asthma/metabolism , Chemokine CCL11/metabolism , Drugs, Chinese Herbal/pharmacology , Fibroblasts/drug effects , Interleukin-4/metabolism , Interleukin-5/immunology , Th2 Cells/metabolism , Animals , Cell Line , Down-Regulation/drug effects , Drug Synergism , Drugs, Chinese Herbal/analysis , Fibroblasts/metabolism , Humans , Interleukin-5/genetics , Mice , Plants, Medicinal/chemistry , Th2 Cells/drug effects
14.
Phytother Res ; 27(9): 1381-91, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23165939

ABSTRACT

Allergic asthma is associated with Th2-mediated inflammation. Several flavonoids were isolated from Glycyrrhiza uralensis, one of the herbs in the anti-asthma herbal medicine intervention. The aim of this investigation was to determine whether Glycyrrhiza uralensis flavonoids have inhibitory effects on memory Th2 responses in vitro and antigen-induced Th2 inflammation in vivo. The effects of three Glycyrrhiza uralensis flavonoids on effector memory Th2 cells, D10.G4.1 (D10 cells), were determined by measuring Th2 cytokine production. Isoliquiritigenin, 7, 4'-dihydroxyflavone (7, 4'-DHF) and liquiritigenin significantly suppressed IL-4 and IL-5 production in a dose-dependent manner, 7, 4'-DHF being most potent. It was also evaluated for effects on D10 cell proliferation, GATA-3 expression and IL-4 mRNA expression, which were suppressed, with no loss of cell viability. Chronic treatment with 7, 4'-DHF in a murine model of allergic asthma not only significantly reduced eosinophilic pulmonary inflammation, serum IgE levels, IL-4 and IL-13 levels, but also increased IFN-γ production in lung cell cultures in response to antigen stimulation.


Subject(s)
Asthma/drug therapy , Flavonoids/pharmacology , Glycyrrhiza uralensis/chemistry , Th2 Cells/drug effects , Animals , Asthma/immunology , Cell Line , Chalcones/pharmacology , Disease Models, Animal , Female , Flavanones/pharmacology , GATA3 Transcription Factor/metabolism , Humans , Immunologic Memory/drug effects , Interferon-gamma/immunology , Interleukin-4 , Interleukin-5/immunology , Lung/cytology , Mice , Mice, Inbred BALB C , Phytotherapy , Plants, Medicinal/chemistry , Th2 Cells/immunology
16.
Phytother Res ; 24(7): 1047-55, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19998324

ABSTRACT

Allergic asthma is a chronic and progressive inflammatory disease for which there is no satisfactory treatment. Studies reported tolerability and efficacy of an anti-asthma herbal medicine intervention (ASHMI) for asthma patients, developed from traditional Chinese medicine. To investigate the pharmacological actions of ASHMI on early- and late-phase airway responses (EAR and LAR), Ovalbumin (OVA)-sensitized mice received 6 weeks of ASHMI treatment beginning 24 h following the first intratracheal OVA challenge. EAR were determined 30 min following the fourth challenge and LAR 48 h following the last challenge. ASHMI effects on cytokine secretion, murine tracheal ring contraction and human bronchial smooth muscle cell prostaglandin (PG) production were also determined.ASHMI abolished EAR, which was associated with significantly reduced histamine, leukotriene C4, and OVA-specific IgE levels, as well as LAR, which was associated with significantly reduced bronchoalveolar lavage fluid (BALF) eosinophils, decreased airway remodeling, and lower Th2 cytokine levels in BALF and splenocyte cultures. Furthermore, ASHMI inhibited contraction of murine tracheal rings and increased production of the potent smooth muscle relaxer PGI(2). ASHMI abrogation of allergic airway responses is associated with broad effects on asthma pathological mechanisms.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Drugs, Chinese Herbal/pharmacology , Airway Remodeling , Animals , Asthma/immunology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Chromatography, High Pressure Liquid , Cytokines/metabolism , Eosinophils/drug effects , Eosinophils/immunology , Histamine/blood , Humans , Immunoglobulin E/blood , In Vitro Techniques , Leukotriene C4/blood , Lung/pathology , Mice , Mice, Inbred BALB C , Molecular Structure , Myocytes, Smooth Muscle/drug effects , Ovalbumin/pharmacology , Prostaglandins/metabolism , Trachea/drug effects
17.
J Allergy Clin Immunol ; 123(2): 443-51, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19203662

ABSTRACT

BACKGROUND: Food allergy is a serious and sometimes fatal condition for which there is no cure. We previously reported that Food Allergy Herbal Formula (FAHF)-2) protected peanut-allergic mice against anaphylactic reactions as long as 4 weeks posttherapy. This formula is now in clinical trials in the United States. OBJECTIVE: We sought to determine whether FAHF-2-mediated protection could be extended long-term and explored the mechanisms underlying its persistent immunomodulatory effects. METHODS: Peanut-allergic mice received FAHF-2 daily orally by gavage for 7 weeks, and then received 7 oral peanut challenges at intervals of 4 to 10 weeks over a period of 36 weeks. For mechanistic studies, some mice received CD4(+) or CD8(+) T-cell-depleting antibodies or IFN-gamma-neutralizing antibodies. Anaphylactic symptoms, body temperatures, and plasma histamine levels were recorded after each challenge, and peanut-specific immunoglobulin levels and cytokine profiles of splenocytes, mesenteric lymph node cells, and purified CD4(+) and CD8(+) T cells were determined. RESULTS: Food Allergy Herbal Formula-2 treatment protected mice from anaphylaxis for more than 36 weeks after discontinuing treatment. Peanut-specific IgE levels were reduced as much as 50%, whereas IgG(2a) levels were increased as much as 60%, and these effects persisted over time. T(H)2 cytokine production by CD4(+) T cells from FAHF-2-treated mice was reduced as much as 75%, whereas CD8(+) T-cell IFN-gamma production was markedly increased by as much as 85% at the final challenge. Neutralization of INF-gamma and depletion of CD8(+) T cells markedly attenuated FAHF-2 efficacy. CONCLUSIONS: Food Allergy Herbal Formula-2 provides long-term protection from anaphylaxis by inducing a beneficial shift in allergen-specific immune responses mediated largely by elevated CD8(+) T-cell IFN-gamma production.


Subject(s)
Anaphylaxis/prevention & control , CD8-Positive T-Lymphocytes/immunology , Interferon-gamma/immunology , Peanut Hypersensitivity/prevention & control , Plant Extracts/therapeutic use , Allergens/administration & dosage , Allergens/immunology , Allergens/pharmacology , Anaphylaxis/immunology , Animals , Arachis/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cytokines/biosynthesis , Cytokines/drug effects , Cytokines/immunology , Disease Models, Animal , Female , Histamine/blood , Immunoglobulin E/blood , Interferon-gamma/metabolism , Lymphocyte Depletion , Mice , Mice, Inbred C3H , Peanut Hypersensitivity/immunology , Plant Extracts/pharmacology , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism
18.
J Agric Food Chem ; 57(3): 820-5, 2009 Feb 11.
Article in English | MEDLINE | ID: mdl-19132888

ABSTRACT

Glycyrrhiza uralensis (Gan-Cao), commonly called "licorice", is one of the most commonly used herbs in traditional Chinese medicine (TCM). In the United States, licorice products are most often consumed as flavoring and sweetening agents in food products. The licorice triterpenoid glycyrrhizin has several biological activities, including anti-inflammatory activity. Other potential anti-inflammatory constituents in G. uralensis have not been fully investigated. Airway eosinophilic inflammation is a major feature of allergic asthma. Eotaxin-1 (eotaxin) is involved in the recruitment of eosinophils to sites of antigen-induced inflammation in asthmatic airways. Because human lung fibroblasts are the major source of eotaxin, inhibition of eosinophil recruitment by suppression of fibroblast eotaxin production is a potentially valuable approach for the pharmacological intervention in asthma. A systematic bioassay-guided purification of G. uralensis yielded five flavonoids: liquiritin, liquiritigenin, isoliquiritigenin, 7,4'-dihydroxyflavone, and isoononin. The structures of the compounds were established by (1)H and (13)C nuclear magnetic resonance (NMR) and liquid chromatography-mass spectrometry (LC-MS) studies. The potential ability of these isolated pure compounds and glycyrrhizin to inhibit secretion of eotaxin-1 by human fetal lung fibroblasts (HFL-1) was tested. Liquiritigenin, isoliquiritigenin, and 7,4'-dihydroxyflavone were more effective than liquiritin, isoononin, and glycyrrhizin in suppressing eotaxin secretion. A concentration-response study showed the IC(50) concentrations of liquiritigenin, isoliquiritigenin, and 7,4'-dihydroxyflavone were 4.2, 0.92, and 0.21 microg/mL, respectively, demonstrating that Glycyrrhiza flavonoids inhibit eotaxin-1 secretion in vitro.


Subject(s)
Chemokine CCL11/metabolism , Fibroblasts/metabolism , Flavonoids/pharmacology , Glycyrrhiza/chemistry , Chemokine CCL11/antagonists & inhibitors , Chromatography, High Pressure Liquid , Fetus , Fibroblasts/drug effects , Flavonoids/analysis , Flavonoids/chemistry , Glycyrrhizic Acid/pharmacology , Humans , Lung , Mass Spectrometry
19.
Phytother Res ; 22(5): 651-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18389474

ABSTRACT

It was previously shown that a Chinese herbal formula, Food Allergy Herbal Formula 2 (FAHF-2) composed of nine herbs, blocked peanut-induced anaphylaxis in a murine model. The current study was designed to investigate the pharmacological actions of individual herbs comprising FAHF-2 on peanut-induced anaphylactic reactions in a murine model of peanut allergy and to determine if all nine herbs are necessary to prevent an anaphylactic reaction, or if a simplified formula containing fewer herbs would be equally effective. Some individual herbs reduced peanut-induced anaphylactic symptoms but no single herb offered full protection from anaphylactic symptoms equivalent to FAHF-2. The herbs had highly variable effects on histamine release, as well as peanut-specific serum IgE and IgG2a levels. The herbs also had variable effects on IL-4, IL-5 and IFN-gamma levels. A simplified formula comprising the most efficacious tested individual herbs showed only partial efficacy and was not able to reproduce comparably the effects of FAHF-2, suggesting that component herbs of FAHF-2 may work synergistically to produce the curative therapeutic effects produced by the whole formula, which appears to be the best option for future clinical trials.


Subject(s)
Peanut Hypersensitivity/drug therapy , Peanut Hypersensitivity/immunology , Plant Extracts/pharmacology , Anaphylaxis/blood , Anaphylaxis/drug therapy , Anaphylaxis/immunology , Animals , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Interferon-gamma/blood , Interferon-gamma/immunology , Interleukin-4/blood , Interleukin-4/immunology , Interleukin-5/blood , Interleukin-5/immunology , Medicine, Chinese Traditional/methods , Mice , Peanut Hypersensitivity/blood
20.
Mt Sinai J Med ; 74(1): 27-32, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17516560

ABSTRACT

The global relationship between drugs that are approved for therapeutic use and the human genome is not known. We employed graph-theory methods to analyze the Federal Food and Drug Administration (FDA) approved drugs and their known molecular targets. We used the FDA Approved Drug Products with Therapeutic Equivalence Evaluations 26(th) Edition Electronic Orange Book (EOB) to identify all FDA approved drugs and their active ingredients. We then connected the list of active ingredients extracted from the EOB to those known human protein targets included in the DrugBank database and constructed a bipartite network. We computed network statistics and conducted Gene Ontology analysis on the drug targets and drug categories. We find that drug to drug-target relationship in the bipartite network is scale-free. Several classes of proteins in the human genome appear to be better targets for drugs since they appear to be selectively enriched as drug targets for the currently FDA approved drugs. These initial observations allow for development of an integrated research methodology to identify general principles of the drug discovery process.


Subject(s)
Computational Biology , Databases, Factual , Drug Approval , United States Food and Drug Administration , Humans , Systems Biology , United States
SELECTION OF CITATIONS
SEARCH DETAIL
...