Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(7)2023 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-37047398

RESUMEN

Inflammatory bowel disease (IBD) refers to chronic intestinal immune-mediated diseases including two main disease manifestations: ulcerative colitis (UC) and Crohn's disease (CD). Epidemiological, clinical, and preclinical evidence has highlighted the potential anti-inflammatory properties of naturally occurring alkaloids. In the present study, we investigated the potential anti-inflammatory activities of the tobacco alkaloids nicotine and anatabine in a dextran sulfate sodium (DSS)-induced UC mouse model with a fully humanized immune system. Our results show that nicotine significantly reduced all acute colitis symptoms and improved colitis-specific endpoints, including histopathologically assessed colon inflammation, tissue damage, and mononuclear cell infiltration. The tobacco alkaloid anatabine showed similar effectiveness trends, although they were generally weaker or not significant. Gene expression analysis in the context of biological network models of IBD further pinpointed a possible mechanism by which nicotine attenuated DSS-induced colitis in humanized mice. The current study enables further investigation of possible molecular mechanisms by which tobacco alkaloids attenuate UC symptoms.


Asunto(s)
Alcaloides , Antineoplásicos , Colitis Ulcerosa , Colitis , Enfermedades Inflamatorias del Intestino , Animales , Ratones , Nicotiana/efectos adversos , Nicotina/efectos adversos , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Modelos Animales de Enfermedad , Antiinflamatorios/uso terapéutico , Antineoplásicos/uso terapéutico , Alcaloides/farmacología , Alcaloides/metabolismo , Sistema Inmunológico/metabolismo , Sulfato de Dextran/toxicidad , Ratones Endogámicos C57BL , Colon/metabolismo
2.
J Nat Prod ; 84(4): 1012-1021, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33706515

RESUMEN

Natural alkaloids, a large class of plant-derived substances, have attracted considerable interest because of their pharmacological activities. In this study, the in vivo pharmacokinetics and anti-inflammatory profile of anatabine, a naturally occurring alkaloid, were characterized in rodents. Anatabine was found to be bioavailable and brain-penetrant following systemic administration. Following intraperitoneal (i.p.) administration (1, 2, and 5 mg/kg), anatabine caused a dose-dependent reduction in carrageenan-induced paw edema in rats; in mice, it inhibited the production of pro-inflammatory cytokines and simultaneously elevated the levels of an anti-inflammatory cytokine in a dose-dependent manner 2 h after lipopolysaccharide challenge. Furthermore, anatabine (∼10 and ∼20 mg/kg/day for 4 weeks; inhalation exposure) had effects in a murine model of multiple sclerosis, reducing neurological deficits and bodyweight loss. Comparative studies of the pharmacokinetics and anti-inflammatory activity of anatabine demonstrated its bioequivalence in rats following i.p. administration and inhalation exposure. This study not only provides the first detailed profile of anatabine pharmacokinetics in rodents but also comprehensively characterizes the anti-inflammatory activities of anatabine in acute and chronic inflammatory models. These findings provide a basis for further characterizing and optimizing the anti-inflammatory properties of anatabine.


Asunto(s)
Alcaloides/farmacología , Antiinflamatorios/farmacología , Piridinas/farmacología , Alcaloides/farmacocinética , Animales , Antiinflamatorios/farmacocinética , Encéfalo/metabolismo , Carragenina , Citocinas , Edema/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Piridinas/farmacocinética , Ratas , Ratas Wistar
3.
Chem Res Toxicol ; 33(10): 2550-2564, 2020 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-32638588

RESUMEN

Transcriptomic approaches can give insight into molecular mechanisms underlying chemical toxicity and are increasingly being used as part of toxicological assessments. To aid the interpretation of transcriptomic data, we have developed a systems toxicology method that relies on a computable biological network model. We created the first network model describing cardiotoxicity in zebrafish larvae-a valuable emerging model species in testing cardiotoxicity associated with drugs and chemicals. The network is based on scientific literature and represents hierarchical molecular pathways that lead from receptor activation to cardiac pathologies. To test the ability of our approach to detect cardiotoxic outcomes from transcriptomic data, we have selected three publicly available data sets that reported chemically induced heart pathologies in zebrafish larvae for five different chemicals. Network-based analysis detected cardiac perturbations for four out of five chemicals tested, for two of them using transcriptomic data collected up to 3 days before the onset of a visible phenotype. Additionally, we identified distinct molecular pathways that were activated by the different chemicals. The results demonstrate that the proposed integrational method can be used for evaluating the effects of chemicals on the zebrafish cardiac function and, together with observed cardiac apical end points, can provide a comprehensive method for connecting molecular events to organ toxicity. The computable network model is freely available and may be used to generate mechanistic hypotheses and quantifiable perturbation values from any zebrafish transcriptomic data.


Asunto(s)
Biología Computacional , Corazón/efectos de los fármacos , Animales , Cardiotoxicidad , Corazón/fisiopatología , Pez Cebra/embriología
4.
Arch Toxicol ; 94(6): 2179-2206, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32367274

RESUMEN

The use of flavoring substances is an important element in the development of reduced-risk products for adult smokers to increase product acceptance and encourage switching from cigarettes. In a first step towards characterizing the sub-chronic inhalation toxicity of neat flavoring substances, a study was conducted using a mixture of the substances in a base solution of e-liquid, where the standard toxicological endpoints of the nebulized aerosols were supplemented with transcriptomics analysis. The flavor mixture was produced by grouping 178 flavors into 26 distinct chemical groups based on structural similarities and potential metabolic and biological effects. Flavoring substances predicted to show the highest toxicological effect from each group were selected as the flavor group representatives (FGR). Following Organization for Economic Cooperation and Development Testing Guideline 413, rats were exposed to three concentrations of the FGR mixture in an e-liquid composed of nicotine (23 µg/L), propylene glycol (1520 µg/L), and vegetable glycerin (1890 µg/L), while non-flavored and no-nicotine mixtures were included as references to identify potential additive or synergistic effects between nicotine and the flavoring substances. The results indicated that the inhalation of an e-liquid containing the mixture of FGRs caused very minimal local and systemic toxic effects. In particular, there were no remarkable clinical (in-life) observations in flavored e-liquid-exposed rats. The biological effects related to exposure to the mixture of neat FGRs were limited and mainly nicotine-mediated, including changes in hematological and blood chemistry parameters and organ weight. These results indicate no significant additive biological changes following inhalation exposure to the nebulized FGR mixture above the nicotine effects measured in this sub-chronic inhalation study. In a subsequent study, e-liquids with FGR mixtures will be aerosolized by thermal treatment and assessed for toxicity.


Asunto(s)
Cigarrillo Electrónico a Vapor/toxicidad , Sistemas Electrónicos de Liberación de Nicotina , Aromatizantes/toxicidad , Perfilación de la Expresión Génica , Hígado/efectos de los fármacos , Sistema Respiratorio/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Vapeo/efectos adversos , Animales , Biomarcadores/sangre , Seguridad de Productos para el Consumidor , Femenino , Exposición por Inhalación , Hígado/metabolismo , Hígado/patología , Masculino , Ratas Sprague-Dawley , Sistema Respiratorio/inmunología , Sistema Respiratorio/metabolismo , Sistema Respiratorio/patología , Medición de Riesgo , Factores de Tiempo , Pruebas de Toxicidad
5.
BMC Bioinformatics ; 20(1): 451, 2019 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-31481014

RESUMEN

BACKGROUND: High-throughput gene expression technologies provide complex datasets reflecting mechanisms perturbed in an experiment, typically in a treatment versus control design. Analysis of these information-rich data can be guided based on a priori knowledge, such as networks of related proteins or genes. Assessing the response of a specific mechanism and investigating its biological basis is extremely important in systems toxicology; as compounds or treatment need to be assessed with respect to a predefined set of key mechanisms that could lead to toxicity. Two-layer networks are suitable for this task, and a robust computational methodology specifically addressing those needs was previously published. The NPA package ( https://github.com/philipmorrisintl/NPA ) implements the algorithm, and a data package of eight two-layer networks representing key mechanisms, such as xenobiotic metabolism, apoptosis, or epithelial immune innate activation, is provided. RESULTS: Gene expression data from an animal study are analyzed using the package and its network models. The functionalities are implemented using R6 classes, making the use of the package seamless and intuitive. The various network responses are analyzed using the leading node analysis, and an overall perturbation, called the Biological Impact Factor, is computed. CONCLUSIONS: The NPA package implements the published network perturbation amplitude methodology and provides a set of two-layer networks encoded in the Biological Expression Language.


Asunto(s)
Metodologías Computacionales , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Programas Informáticos , Algoritmos , Animales , Apoptosis/genética , Ciclo Celular/genética , Bases de Datos Genéticas , Matriz Extracelular/metabolismo , Ratones Endogámicos C57BL , Estrés Oxidativo , Transcriptoma/genética
6.
Inhal Toxicol ; 30(13-14): 553-567, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30849254

RESUMEN

We compared early biological changes in mice after inhalation exposures to cigarette smoke or e-vapor aerosols (MarkTen® cartridge with Carrier, Test-1, or Test-2 formulations; 4% nicotine). Female C57BL/6 mice were exposed to 3R4F cigarette smoke or e-vapor aerosols by nose-only inhalation for up to 4 hours/day, 5 days/week, for 3 weeks. The 3R4F and e-vapor exposures were set to match the target nose port aerosol nicotine concentration (∼41 µg/L). Only the 3R4F group showed postexposure clinical signs such as tremors and lethargy. At necropsy, the 3R4F group had significant increases in lung weight and changes in bronchoalveolar lavage parameters, as well as microscopic findings in the respiratory tract. The e-vapor groups had minimal microscopic changes, including squamous metaplasia in laryngeal epiglottis, and histiocytic infiltrates in the lung (Test-2 group only). The 3R4F group had a higher incidence and severity of microscopic findings compared to any e-vapor group. Transcriptomic analysis also showed that the 3R4F group had the highest number of differentially expressed genes compared to Sham Control. Among e-vapor groups, Test-2 group had more differentially expressed genes but the magnitude of gene expression-based network perturbations in all e-vapor groups was ∼94% less than the 3R4F group. On proteome analysis in the lung, differentially regulated proteins were detected in the 3R4F group only. In conclusion, 3-weeks of 3R4F exposure induced molecular and microscopic changes associated with smoking-related diseases in the respiratory tract, while e-vapor exposures showed substantially reduced biological activities.


Asunto(s)
Sistemas Electrónicos de Liberación de Nicotina , Sistema Respiratorio/efectos de los fármacos , Humo/efectos adversos , Productos de Tabaco/efectos adversos , Administración por Inhalación , Aerosoles , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Carboxihemoglobina/análisis , Femenino , Perfilación de la Expresión Génica , Ratones Endogámicos C57BL , Pruebas de Función Respiratoria , Fenómenos Fisiológicos Respiratorios/efectos de los fármacos , Sistema Respiratorio/metabolismo , Sistema Respiratorio/patología
7.
Regul Toxicol Pharmacol ; 81 Suppl 2: S151-S157, 2016 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-27845159

RESUMEN

As part of current harm reduction strategies, candidate modified risk tobacco products (MRTP) are developed to offer adult smokers who want to continue using tobacco product an alternative to cigarettes while potentially reducing individual risk and population harm compared to smoking cigarettes. One of these candidate MRTPs is the Tobacco Heating System (THS) 2.2 which does not burn tobacco, but instead heats it, thus producing significantly reduced levels of harmful and potentially harmful constituents (HPHC) compared with combustible cigarettes (CC). A controlled, parallel group, open-label clinical study was conducted with subjects randomized to three monitored groups: (1) switching from CCs to THS2.2; (2) continuous use of non-menthol CC brand (CC arm); or (3) smoking abstinence (SA arm) for five days. Exposure response was assessed by measuring biomarkers of exposure to selected HPHCs. To complement the classical exposure response measurements, we have used the previously reported whole blood derived gene signature that can distinguish current smokers from either non-smokers or former smokers with high specificity and sensitivity. We tested the small signature consisting of only 11 genes on the blood transcriptome of subjects enrolled in the clinical study and showed a reduced exposure response in subjects that either stopped smoking or switched to a candidate MRTP, the THS2.2, compared with subjects who continued smoking their regular tobacco product.


Asunto(s)
Sistemas Electrónicos de Liberación de Nicotina/efectos adversos , Reducción del Daño , Calor , Humo/efectos adversos , Fumar/efectos adversos , Biología de Sistemas , Industria del Tabaco , Productos de Tabaco/toxicidad , Pruebas de Toxicidad/métodos , Aerosoles , Biomarcadores/sangre , Seguridad de Productos para el Consumidor , Diseño de Equipo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Marcadores Genéticos , Genómica , Humanos , Exposición por Inhalación/efectos adversos , Polonia , Medición de Riesgo , Fumar/sangre , Fumar/genética , Cese del Hábito de Fumar/métodos , Prevención del Hábito de Fumar , Transcriptoma/efectos de los fármacos
8.
Regul Toxicol Pharmacol ; 81 Suppl 2: S82-S92, 2016 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-27866933

RESUMEN

Modified-risk tobacco products (MRTP) are designed to reduce the individual risk of tobacco-related disease as well as population harm compared to smoking cigarettes. Experimental proof of their benefit needs to be provided at multiple levels in research fields. Here, we examined microRNA (miRNA) levels in the lungs of rats exposed to a candidate modified-risk tobacco product, the Tobacco Heating System 2.2 (THS2.2) in a 90-day OECD TG-413 inhalation study. Our aim was to assess the miRNA response to THS2.2 aerosol compared with the response to combustible cigarettes (CC) smoke from the reference cigarette 3R4F. CC smoke exposure, but not THS2.2 aerosol exposure, caused global miRNA downregulation, which may be explained by the interference of CC smoke constituents with the miRNA processing machinery. Upregulation of specific miRNA species, such as miR-146a/b and miR-182, indicated that they are causal elements in the inflammatory response in CC-exposed lungs, but they were reduced after THS2.2 aerosol exposure. Transforming transcriptomic data into protein activity based on corresponding downstream gene expression, we identified potential mechanisms for miR-146a/b and miR-182 that were activated by CC smoke but not by THS2.2 aerosol and possibly involved in the regulation of those miRNAs. The inclusion of miRNA profiling in systems toxicology approaches increases the mechanistic understanding of the complex exposure responses.


Asunto(s)
Sistemas Electrónicos de Liberación de Nicotina/efectos adversos , Reducción del Daño , Calor , Pulmón/efectos de los fármacos , MicroARNs/genética , Neumonía/prevención & control , Fumar/efectos adversos , Industria del Tabaco , Productos de Tabaco/toxicidad , Pruebas de Toxicidad/métodos , Aerosoles , Animales , Biología Computacional , Seguridad de Productos para el Consumidor , Diseño de Equipo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Marcadores Genéticos , Genómica , Humanos , Exposición por Inhalación/efectos adversos , Pulmón/metabolismo , Masculino , MicroARNs/metabolismo , Neumonía/inducido químicamente , Neumonía/genética , Ratas Sprague-Dawley , Medición de Riesgo , Humo/efectos adversos , Fumar/genética , Factores de Tiempo , Toxicogenética , Transcriptoma/efectos de los fármacos
9.
Inflamm Res ; 64(7): 471-86, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25962837

RESUMEN

BACKGROUND: Mouse models are useful for studying cigarette smoke (CS)-induced chronic pulmonary pathologies such as lung emphysema. To enhance translation of large-scale omics data from mechanistic studies into pathophysiological changes, we have developed computational tools based on reverse causal reasoning (RCR). OBJECTIVE: In the present study we applied a systems biology approach leveraging RCR to identify molecular mechanistic explanations of pathophysiological changes associated with CS-induced lung emphysema in susceptible mice. METHODS: The lung transcriptomes of five mouse models (C57BL/6, ApoE (-/-) , A/J, CD1, and Nrf2 (-/-) ) were analyzed following 5-7 months of CS exposure. RESULTS: We predicted 39 molecular changes mostly related to inflammatory processes including known key emphysema drivers such as NF-κB and TLR4 signaling, and increased levels of TNF-α, CSF2, and several interleukins. More importantly, RCR predicted potential molecular mechanisms that are less well-established, including increased transcriptional activity of PU.1, STAT1, C/EBP, FOXM1, YY1, and N-COR, and reduced protein abundance of ITGB6 and CFTR. We corroborated several predictions using targeted proteomic approaches, demonstrating increased abundance of CSF2, C/EBPα, C/EBPß, PU.1, BRCA1, and STAT1. CONCLUSION: These systems biology-derived candidate mechanisms common to susceptible mouse models may enhance understanding of CS-induced molecular processes underlying emphysema development in mice and their relevancy for human chronic obstructive pulmonary disease.


Asunto(s)
Nicotiana , Enfisema Pulmonar/genética , Enfisema Pulmonar/patología , Humo , Animales , Apolipoproteínas E/genética , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Causalidad , Perfilación de la Expresión Génica , Exposición por Inhalación , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CFTR , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Proteómica , Enfisema Pulmonar/inducido químicamente , Fumar , Especificidad de la Especie
10.
BMC Bioinformatics ; 15: 238, 2014 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-25015298

RESUMEN

BACKGROUND: High-throughput measurement technologies such as microarrays provide complex datasets reflecting mechanisms perturbed in an experiment, typically a treatment vs. control design. Analysis of these information rich data can be guided based on a priori knowledge, such as networks or set of related proteins or genes. Among those, cause-and-effect network models are becoming increasingly popular and more than eighty such models, describing processes involved in cell proliferation, cell fate, cell stress, and inflammation have already been published. A meaningful systems toxicology approach to study the response of a cell system, or organism, exposed to bio-active substances requires a quantitative measure of dose-response at network level, to go beyond the differential expression of single genes. RESULTS: We developed a method that quantifies network response in an interpretable manner. It fully exploits the (signed graph) structure of cause-and-effect networks models to integrate and mine transcriptomics measurements. The presented approach also enables the extraction of network-based signatures for predicting a phenotype of interest. The obtained signatures are coherent with the underlying network perturbation and can lead to more robust predictions across independent studies. The value of the various components of our mathematically coherent approach is substantiated using several in vivo and in vitro transcriptomics datasets. As a proof-of-principle, our methodology was applied to unravel mechanisms related to the efficacy of a specific anti-inflammatory drug in patients suffering from ulcerative colitis. A plausible mechanistic explanation of the unequal efficacy of the drug is provided. Moreover, by utilizing the underlying mechanisms, an accurate and robust network-based diagnosis was built to predict the response to the treatment. CONCLUSION: The presented framework efficiently integrates transcriptomics data and "cause and effect" network models to enable a mathematically coherent framework from quantitative impact assessment and data interpretation to patient stratification for diagnosis purposes.


Asunto(s)
Modelos Biológicos , Biología de Sistemas/métodos , Animales , Antiinflamatorios/efectos adversos , Antiinflamatorios/uso terapéutico , Colitis Ulcerosa/tratamiento farmacológico , Relación Dosis-Respuesta a Droga , Perfilación de la Expresión Génica , Humanos , Fenotipo , Toxicología
11.
Bioinformatics ; 29(22): 2892-9, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23966112

RESUMEN

MOTIVATION: After more than a decade since microarrays were used to predict phenotype of biological samples, real-life applications for disease screening and identification of patients who would best benefit from treatment are still emerging. The interest of the scientific community in identifying best approaches to develop such prediction models was reaffirmed in a competition style international collaboration called IMPROVER Diagnostic Signature Challenge whose results we describe herein. RESULTS: Fifty-four teams used public data to develop prediction models in four disease areas including multiple sclerosis, lung cancer, psoriasis and chronic obstructive pulmonary disease, and made predictions on blinded new data that we generated. Teams were scored using three metrics that captured various aspects of the quality of predictions, and best performers were awarded. This article presents the challenge results and introduces to the community the approaches of the best overall three performers, as well as an R package that implements the approach of the best overall team. The analyses of model performance data submitted in the challenge as well as additional simulations that we have performed revealed that (i) the quality of predictions depends more on the disease endpoint than on the particular approaches used in the challenge; (ii) the most important modeling factor (e.g. data preprocessing, feature selection and classifier type) is problem dependent; and (iii) for optimal results datasets and methods have to be carefully matched. Biomedical factors such as the disease severity and confidence in diagnostic were found to be associated with the misclassification rates across the different teams. AVAILABILITY: The lung cancer dataset is available from Gene Expression Omnibus (accession, GSE43580). The maPredictDSC R package implementing the approach of the best overall team is available at www.bioconductor.org or http://bioinformaticsprb.med.wayne.edu/.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Técnicas de Diagnóstico Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Fenotipo , Enfermedad/genética , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/genética , Esclerosis Múltiple/diagnóstico , Esclerosis Múltiple/genética , Psoriasis/diagnóstico , Psoriasis/genética , Enfermedad Pulmonar Obstructiva Crónica/diagnóstico , Enfermedad Pulmonar Obstructiva Crónica/genética
12.
J Transl Med ; 12: 185, 2014 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-24965703

RESUMEN

BACKGROUND: Numerous inflammation-related pathways have been shown to play important roles in atherogenesis. Rapid and efficient assessment of the relative influence of each of those pathways is a challenge in the era of "omics" data generation. The aim of the present work was to develop a network model of inflammation-related molecular pathways underlying vascular disease to assess the degree of translatability of preclinical molecular data to the human clinical setting. METHODS: We constructed and evaluated the Vascular Inflammatory Processes Network (V-IPN), a model representing a collection of vascular processes modulated by inflammatory stimuli that lead to the development of atherosclerosis. RESULTS: Utilizing the V-IPN as a platform for biological discovery, we have identified key vascular processes and mechanisms captured by gene expression profiling data from four independent datasets from human endothelial cells (ECs) and human and murine intact vessels. Primary ECs in culture from multiple donors revealed a richer mapping of mechanisms identified by the V-IPN compared to an immortalized EC line. Furthermore, an evaluation of gene expression datasets from aortas of old ApoE-/- mice (78 weeks) and human coronary arteries with advanced atherosclerotic lesions identified significant commonalities in the two species, as well as several mechanisms specific to human arteries that are consistent with the development of unstable atherosclerotic plaques. CONCLUSIONS: We have generated a new biological network model of atherogenic processes that demonstrates the power of network analysis to advance integrative, systems biology-based knowledge of cross-species translatability, plaque development and potential mechanisms leading to plaque instability.


Asunto(s)
Aterosclerosis/patología , Vasos Sanguíneos/patología , Inflamación/patología , Modelos Cardiovasculares , Placa Aterosclerótica/patología , Transducción de Señal , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/metabolismo , Aterosclerosis/genética , Análisis por Conglomerados , Bases de Datos como Asunto , Humanos , Ratones , Oportunidad Relativa , Placa Aterosclerótica/genética , Programas Informáticos , Transcriptoma/genética , Investigación Biomédica Traslacional
13.
Int J Toxicol ; 33(6): 506-17, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25297719

RESUMEN

Exposure to cigarette smoke (CS) is linked to the development of respiratory diseases, and there is a need to understand the mechanisms whereby CS causes damage. Although animal models have provided valuable insights into smoking-related respiratory tract damage, modern toxicity testing calls for reliable in vitro models as alternatives for animal experimentation. We report on a repeated whole mainstream CS exposure of nasal and bronchial organotypic tissue cultures that mimic the morphological, physiological, and molecular attributes of the human respiratory tract. Despite the similar cellular staining and cytokine secretion in both tissue types, the transcriptomic analyses in the context of biological network models identified similar and diverse biological processes that were impacted by CS-exposed nasal and bronchial cultures. Our results demonstrate that nasal and bronchial tissue cultures are appropriate in vitro models for the assessment of CS-induced adverse effects in the respiratory system and promising alternative to animal experimentation.


Asunto(s)
Bronquios/efectos de los fármacos , Mucosa Nasal/efectos de los fármacos , Nicotiana/efectos adversos , Humo/efectos adversos , Técnicas de Cultivo de Tejidos , Anciano , Alternativas a las Pruebas en Animales , Bronquios/metabolismo , Citocinas/metabolismo , Células Epiteliales , Femenino , Fibroblastos , Perfilación de la Expresión Génica , Humanos , Masculino , Mucosa Nasal/metabolismo
14.
Toxicol Mech Methods ; 24(7): 470-87, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25046638

RESUMEN

Smoking has been associated with diseases of the lung, pulmonary airways and oral cavity. Cytologic, genomic and transcriptomic changes in oral mucosa correlate with oral pre-neoplasia, cancer and inflammation (e.g. periodontitis). Alteration of smoking-related gene expression changes in oral epithelial cells is similar to that in bronchial and nasal epithelial cells. Using a systems toxicology approach, we have previously assessed the impact of cigarette smoke (CS) seen as perturbations of biological processes in human nasal and bronchial organotypic epithelial culture models. Here, we report our further assessment using in vitro human oral organotypic epithelium models. We exposed the buccal and gingival organotypic epithelial tissue cultures to CS at the air-liquid interface. CS exposure was associated with increased secretion of inflammatory mediators, induction of cytochrome P450s activity and overall weak toxicity in both tissues. Using microarray technology, gene-set analysis and a novel computational modeling approach leveraging causal biological network models, we identified CS impact on xenobiotic metabolism-related pathways accompanied by a more subtle alteration in inflammatory processes. Gene-set analysis further indicated that the CS-induced pathways in the in vitro buccal tissue models resembled those in the in vivo buccal biopsies of smokers from a published dataset. These findings support the translatability of systems responses from in vitro to in vivo and demonstrate the applicability of oral organotypical tissue models for an impact assessment of CS on various tissues exposed during smoking, as well as for impact assessment of reduced-risk products.


Asunto(s)
Mucosa Bucal/efectos de los fármacos , Humo , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Humanos , Técnicas In Vitro , Mucosa Bucal/metabolismo , Nicotiana , Transcriptoma
15.
Toxicol Lett ; 393: 107-113, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38350531

RESUMEN

In the absence of epidemiological data, there is a need to develop computational models that convert in vitro findings to human disease risk predictions following toxicant exposure. In such efforts, in vitro data can be evaluated in the context of adverse outcome pathways (AOPs) that organize mechanistic knowledge based on empirical evidence into a sequence of molecular-, cellular-, tissue-, and organ-level key events that precede an adverse outcome (AO). Here we combined data from advanced in vitro organotypic airway models exposed to combustible cigarette (CC) smoke or Tobacco Heating System (THS) aerosol with an AOP for increased oxidative stress leads to decreased lung function. The mathematical modeling predicted reduced risk of decreased ciliary beating frequency (CBF) based on oxidative stress measurements and reduced risk of decreased mucociliary clearance (MCC) based on CBF measurements in THS aerosol- compared with CC smoke-exposed cultures. To extend the predictions to the AO of decreased lung function, we leveraged human MCC data from current smokers, nonsmokers, former smokers, and users of heated tobacco products. This approach provided a plausible prediction of diminished reduction in lung function in response to THS use compared with continued smoking. The current approach may also present a basis for an integrated approach to testing and assessment of tobacco products for future regulatory decision-making.


Asunto(s)
Rutas de Resultados Adversos , Productos de Tabaco , Humanos , Productos de Tabaco/toxicidad , Humo/efectos adversos , Medición de Riesgo , Pulmón/metabolismo , Aerosoles
16.
Toxicol Appl Pharmacol ; 272(3): 863-78, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23933166

RESUMEN

Exposure to biologically active substances such as therapeutic drugs or environmental toxicants can impact biological systems at various levels, affecting individual molecules, signaling pathways, and overall cellular processes. The ability to derive mechanistic insights from the resulting system responses requires the integration of experimental measures with a priori knowledge about the system and the interacting molecules therein. We developed a novel systems biology-based methodology that leverages mechanistic network models and transcriptomic data to quantitatively assess the biological impact of exposures to active substances. Hierarchically organized network models were first constructed to provide a coherent framework for investigating the impact of exposures at the molecular, pathway and process levels. We then validated our methodology using novel and previously published experiments. For both in vitro systems with simple exposure and in vivo systems with complex exposures, our methodology was able to recapitulate known biological responses matching expected or measured phenotypes. In addition, the quantitative results were in agreement with experimental endpoint data for many of the mechanistic effects that were assessed, providing further objective confirmation of the approach. We conclude that our methodology evaluates the biological impact of exposures in an objective, systematic, and quantifiable manner, enabling the computation of a systems-wide and pan-mechanistic biological impact measure for a given active substance or mixture. Our results suggest that various fields of human disease research, from drug development to consumer product testing and environmental impact analysis, could benefit from using this methodology.


Asunto(s)
Redes Reguladoras de Genes/genética , Mucosa Respiratoria/fisiología , Transcriptoma/genética , Animales , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Simulación por Computador , Humanos , Ratones , Ratones Noqueados , Contaminación por Humo de Tabaco/efectos adversos
17.
Int J Inflam ; 2022: 4071472, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35126992

RESUMEN

Virtually any stressor that alters the cellular homeostatic state may result in an inflammatory response. As a critical component of innate immunity, inflammasomes play a prominent role in the inflammatory response. The information on inflammasome biology is rapidly growing, thus creating the need for structuring it into a model that can help visualize and enhance the understanding of underlying biological processes. Causal biological network (CBN) models provide predictive power for novel disease mechanisms and treatment outcomes. We assembled the available literature information on inflammasome activation into the CBN model and scored it with publicly available transcriptomic datasets that address viral infection of the lungs, osteo- and rheumatoid arthritis, psoriasis, and aging. The scoring inferred pathway activation leading to NLRP3 inflammasome activation in these diverse conditions, demonstrating that the CBN model provides a platform for interpreting transcriptomic data in the context of inflammasome activation.

18.
Sci Rep ; 12(1): 4205, 2022 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-35273209

RESUMEN

Astrocytes play a central role in the neuroimmune response by responding to CNS pathologies with diverse molecular and morphological changes during the process of reactive astrogliosis. Here, we used a computational biological network model and mathematical algorithms that allow the interpretation of high-throughput transcriptomic datasets in the context of known biology to study reactive astrogliosis. We gathered available mechanistic information from the literature into a comprehensive causal biological network (CBN) model of astrocyte reactivity. The CBN model was built in the Biological Expression Language, which is both human-readable and computable. We characterized the CBN with a network analysis of highly connected nodes and demonstrated that the CBN captures relevant astrocyte biology. Subsequently, we used the CBN and transcriptomic data to identify key molecular pathways driving the astrocyte phenotype in four CNS pathologies: samples from mouse models of lipopolysaccharide-induced endotoxemia, Alzheimer's disease, and amyotrophic lateral sclerosis; and samples from multiple sclerosis patients. The astrocyte CBN provides a new tool to identify causal mechanisms and quantify astrogliosis based on transcriptomic data.


Asunto(s)
Gliosis , Enfermedades Neuroinflamatorias , Animales , Astrocitos/metabolismo , Gliosis/patología , Humanos , Inflamación/patología , Ratones , Análisis de Sistemas
19.
Front Genet ; 12: 652632, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34211495

RESUMEN

Adverse outcomes that result from chemical toxicity are rarely caused by dysregulation of individual proteins; rather, they are often caused by system-level perturbations in networks of molecular events. To fully understand the mechanisms of toxicity, it is necessary to recognize the interactions of molecules, pathways, and biological processes within these networks. The developing brain is a prime example of an extremely complex network, which makes developmental neurotoxicity one of the most challenging areas in toxicology. We have developed a systems toxicology method that uses a computable biological network to represent molecular interactions in the developing brain of zebrafish larvae. The network is curated from scientific literature and describes interactions between biological processes, signaling pathways, and adverse outcomes associated with neurotoxicity. This allows us to identify important signaling hubs, pathway interactions, and emergent adverse outcomes, providing a more complete understanding of neurotoxicity. Here, we describe the construction of a zebrafish developmental neurotoxicity network and its validation by integration with publicly available neurotoxicity-related transcriptomic datasets. Our network analysis identified consistent regulation of tumor suppressors p53 and retinoblastoma 1 (Rb1) as well as the oncogene Krüppel-like factor (Klf8) in response to chemically induced developmental neurotoxicity. The developed network can be used to interpret transcriptomic data in a neurotoxicological context.

20.
Sci Rep ; 11(1): 11519, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34075172

RESUMEN

The molecular mechanisms of IBD have been the subject of intensive exploration. We, therefore, assembled the available information into a suite of causal biological network models, which offer comprehensive visualization of the processes underlying IBD. Scientific text was curated by using Biological Expression Language (BEL) and compiled with OpenBEL 3.0.0. Network properties were analysed by Cytoscape. Network perturbation amplitudes were computed to score the network models with transcriptomic data from public data repositories. The IBD network model suite consists of three independent models that represent signalling pathways that contribute to IBD. In the "intestinal permeability" model, programmed cell death factors were downregulated in CD and upregulated in UC. In the "inflammation" model, PPARG, IL6, and IFN-associated pathways were prominent regulatory factors in both diseases. In the "wound healing" model, factors promoting wound healing were upregulated in CD and downregulated in UC. Scoring of publicly available transcriptomic datasets onto these network models demonstrated that the IBD models capture the perturbation in each dataset accurately. The IBD network model suite can provide better mechanistic insights of the transcriptional changes in IBD and constitutes a valuable tool in personalized medicine to further understand individual drug responses in IBD.


Asunto(s)
Colitis Ulcerosa/inmunología , Enfermedad de Crohn/inmunología , Mucosa Intestinal/inmunología , Modelos Inmunológicos , Biología de Sistemas , Transcriptoma/inmunología , Colitis Ulcerosa/patología , Enfermedad de Crohn/patología , Humanos , Mucosa Intestinal/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA