Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
2.
Front Bioeng Biotechnol ; 9: 616830, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33634087

RESUMEN

Evolution has endowed the lung with exceptional design providing a large surface area for gas exchange area (ca. 100 m2) in a relatively small tissue volume (ca. 6 L). This is possible due to a complex tissue architecture that has resulted in one of the most challenging organs to be recreated in the lab. The need for realistic and robust in vitro lung models becomes even more evident as causal therapies, especially for chronic respiratory diseases, are lacking. Here, we describe the Cyclic I n VI tro Cell-stretch (CIVIC) "breathing" lung bioreactor for pulmonary epithelial cells at the air-liquid interface (ALI) experiencing cyclic stretch while monitoring stretch-related parameters (amplitude, frequency, and membrane elastic modulus) under real-time conditions. The previously described biomimetic copolymeric BETA membrane (5 µm thick, bioactive, porous, and elastic) was attempted to be improved for even more biomimetic permeability, elasticity (elastic modulus and stretchability), and bioactivity by changing its chemical composition. This biphasic membrane supports both the initial formation of a tight monolayer of pulmonary epithelial cells (A549 and 16HBE14o-) under submerged conditions and the subsequent cell-stretch experiments at the ALI without preconditioning of the membrane. The newly manufactured versions of the BETA membrane did not improve the characteristics of the previously determined optimum BETA membrane (9.35% PCL and 6.34% gelatin [w/v solvent]). Hence, the optimum BETA membrane was used to investigate quantitatively the role of physiologic cyclic mechanical stretch (10% linear stretch; 0.33 Hz: light exercise conditions) on size-dependent cellular uptake and transepithelial transport of nanoparticles (100 nm) and microparticles (1,000 nm) for alveolar epithelial cells (A549) under ALI conditions. Our results show that physiologic stretch enhances cellular uptake of 100 nm nanoparticles across the epithelial cell barrier, but the barrier becomes permeable for both nano- and micron-sized particles (100 and 1,000 nm). This suggests that currently used static in vitro assays may underestimate cellular uptake and transbarrier transport of nanoparticles in the lung.

3.
Part Fibre Toxicol ; 17(1): 16, 2020 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-32450889

RESUMEN

Toxicity testing and regulation of advanced materials at the nanoscale, i.e. nanosafety, is challenged by the growing number of nanomaterials and their property variants requiring assessment for potential human health impacts. The existing animal-reliant toxicity testing tools are onerous in terms of time and resources and are less and less in line with the international effort to reduce animal experiments. Thus, there is a need for faster, cheaper, sensitive and effective animal alternatives that are supported by mechanistic evidence. More importantly, there is an urgency for developing alternative testing strategies that help justify the strategic prioritization of testing or targeting the most apparent adverse outcomes, selection of specific endpoints and assays and identifying nanomaterials of high concern. The Adverse Outcome Pathway (AOP) framework is a systematic process that uses the available mechanistic information concerning a toxicological response and describes causal or mechanistic linkages between a molecular initiating event, a series of intermediate key events and the adverse outcome. The AOP framework provides pragmatic insights to promote the development of alternative testing strategies. This review will detail a brief overview of the AOP framework and its application to nanotoxicology, tools for developing AOPs and the role of toxicogenomics, and summarize various AOPs of relevance to inhalation toxicity of nanomaterials that are currently under various stages of development. The review also presents a network of AOPs derived from connecting all AOPs, which shows that several adverse outcomes induced by nanomaterials originate from a molecular initiating event that describes the interaction of nanomaterials with lung cells and involve similar intermediate key events. Finally, using the example of an established AOP for lung fibrosis, the review will discuss various in vitro tests available for assessing lung fibrosis and how the information can be used to support a tiered testing strategy for lung fibrosis. The AOPs and AOP network enable deeper understanding of mechanisms involved in inhalation toxicity of nanomaterials and provide a strategy for the development of alternative test methods for hazard and risk assessment of nanomaterials.


Asunto(s)
Rutas de Resultados Adversos , Alternativas a las Pruebas en Animales , Nanoestructuras/toxicidad , Proyectos de Investigación , Pruebas de Toxicidad/métodos , Animales , Humanos
4.
Behav Brain Res ; 352: 187-196, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28966146

RESUMEN

Since decades, model organisms have provided an important approach for understanding the mechanistic basis of human diseases. The German Mouse Clinic (GMC) was the first phenotyping facility that established a collaboration-based platform for phenotype characterization of mouse lines. In order to address individual projects by a tailor-made phenotyping strategy, the GMC advanced in developing a series of pipelines with tests for the analysis of specific disease areas. For a general broad analysis, there is a screening pipeline that covers the key parameters for the most relevant disease areas. For hypothesis-driven phenotypic analyses, there are thirteen additional pipelines with focus on neurological and behavioral disorders, metabolic dysfunction, respiratory system malfunctions, immune-system disorders and imaging techniques. In this article, we give an overview of the pipelines and describe the scientific rationale behind the different test combinations.


Asunto(s)
Modelos Animales de Enfermedad , Ratones Transgénicos , Fenotipo , Animales , Humanos
5.
PLoS One ; 10(9): e0136188, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26340365

RESUMEN

Proteasome inhibition has been shown to prevent development of fibrosis in several organs including the lung. However, effects of proteasome inhibitors on lung fibrosis are controversial and cytotoxic side effects of the overall inhibition of proteasomal protein degradation cannot be excluded. Therefore, we hypothesized that local lung-specific application of a novel, selective proteasome inhibitor, oprozomib (OZ), provides antifibrotic effects without systemic toxicity in a mouse model of lung fibrosis. Oprozomib was first tested on the human alveolar epithelial cancer cell line A549 and in primary mouse alveolar epithelial type II cells regarding its cytotoxic effects on alveolar epithelial cells and compared to the FDA approved proteasome inhibitor bortezomib (BZ). OZ was less toxic than BZ and provided high selectivity for the chymotrypsin-like active site of the proteasome. In primary mouse lung fibroblasts, OZ showed significant anti-fibrotic effects, i.e. reduction of collagen I and α smooth muscle actin expression, in the absence of cytotoxicity. When applied locally into the lungs of healthy mice via instillation, OZ was well tolerated and effectively reduced proteasome activity in the lungs. In bleomycin challenged mice, however, locally applied OZ resulted in accelerated weight loss and increased mortality of treated mice. Further, OZ failed to reduce fibrosis in these mice. While upon systemic application OZ was well tolerated in healthy mice, it rather augmented instead of attenuated fibrotic remodelling of the lung in bleomycin challenged mice. To conclude, low toxicity and antifibrotic effects of OZ in pulmonary fibroblasts could not be confirmed for pulmonary fibrosis of bleomycin-treated mice. In light of these data, the use of proteasome inhibitors as therapeutic agents for the treatment of fibrotic lung diseases should thus be considered with caution.


Asunto(s)
Colágeno/antagonistas & inhibidores , Oligopéptidos/efectos adversos , Inhibidores de Proteasoma/efectos adversos , Fibrosis Pulmonar/tratamiento farmacológico , Administración por Inhalación , Animales , Bleomicina , Línea Celular Tumoral , Colágeno/biosíntesis , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Intubación Intratraqueal , Pulmón , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Cultivo Primario de Células , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/mortalidad , Análisis de Supervivencia , Pérdida de Peso/efectos de los fármacos
6.
Int J Toxicol ; 33(1): 3-13, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24434722

RESUMEN

The 13th International Congress on Combustion By-Products and their Health Effects was held in New Orleans, Louisiana from May 15 to 18, 2013. The congress, sponsored by the Superfund Research Program, National Institute of Environmental Health Sciences, and National Science Foundation, brought together international academic and government researchers, engineers, scientists, and policymakers. With industrial growth, increased power needs and generation and coal consumption and their concomitant emissions, pernicious health effects associated with exposures to these emissions are on the rise. This congress provides a unique platform for interdisciplinary exchange and discussion of these topics. The formation, conversion, control, and health effects of combustion by-products, including particulate matter and associated heavy metals, persistent organic pollutants, and environmentally persistent free radicals, were discussed during the congress. This review will summarize and discuss the implications of the data presented.


Asunto(s)
Contaminación del Aire/prevención & control , Ingeniería , Salud Global , Política de Salud , Contaminación del Aire/efectos adversos , Ingeniería/tendencias , Incendios/prevención & control , Salud Global/tendencias , Política de Salud/tendencias , Humanos , Humo/efectos adversos , Humo/prevención & control , Emisiones de Vehículos/prevención & control , Emisiones de Vehículos/toxicidad
7.
J Biol Chem ; 288(23): 16690-16703, 2013 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-23620591

RESUMEN

The nuclei of most vertebrate cells contain members of the high mobility group N (HMGN) protein family, which bind specifically to nucleosome core particles and affect chromatin structure and function, including transcription. Here, we study the biological role of this protein family by systematic analysis of phenotypes and tissue transcription profiles in mice lacking functional HMGN variants. Phenotypic analysis of Hmgn1(tm1/tm1), Hmgn3(tm1/tm1), and Hmgn5(tm1/tm1) mice and their wild type littermates with a battery of standardized tests uncovered variant-specific abnormalities. Gene expression analysis of four different tissues in each of the Hmgn(tm1/tm1) lines reveals very little overlap between genes affected by specific variants in different tissues. Pathway analysis reveals that loss of an HMGN variant subtly affects expression of numerous genes in specific biological processes. We conclude that within the biological framework of an entire organism, HMGNs modulate the fidelity of the cellular transcriptional profile in a tissue- and HMGN variant-specific manner.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Proteínas HMGN/metabolismo , Transcripción Genética/fisiología , Animales , Proteínas HMGN/genética , Ratones , Ratones Mutantes , Especificidad de Órganos/fisiología
8.
Biomaterials ; 32(9): 2388-98, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21183213

RESUMEN

Since off-target effects in non-viral siRNA delivery are quite common but not well understood, in this study various polymer-related effects observed in transfection studies were described and their mechanisms of toxicity were investigated. A variety of stably luciferase-expressing cell lines was compared concerning polymer-mediated effects after transfection with polyplexes of siRNA and poly(ethylene imine) (PEI) or poly(ethylene glycol)-grafted PEI (PEG-PEI). Cell viability, LDH release, gene expression profiles of apoptosis-related genes and promoter activation were investigated. Interestingly, PEG-PEI, which is generally better tolerated than PEI, was found to activate apoptosis in a cell line- and concentration-dependent manner. While both polymers showed sigmoidal dose-response of cell viability in L929 cells (IC(50)(PEI) = 6 µg/ml, IC(50)(PEG-PEI) = 11 µg/ml), H1299/Luc cells exhibited biphasic dose-response for PEG-PEI and stronger apoptosis at 2 µg/ml than at 20 µg/ml PEG-PEI, as shown in TUNEL assays. Gene expression profiling confirmed that H1299/Luc cells underwent apoptosis via thousand-fold activation of TNF receptor-associated factors. Additionally, it was demonstrated that NFkB-mediated CMV promoter activation in stably transfected cells can lead to increased target gene levels after transfection instead of siRNA-mediated knockdown. With these results, polymeric vectors were shown not to be inert substances. Therefore, alterations in gene expression caused by the delivery agent must be known to correctly interpret gene-silencing experiments, to understand the mechanisms of off-target effects, and most of all to further develop vectors with reduced side effects. Taking these observations into account, one established cell line was eventually identified to be suitable for RNAi experiments. As shown by these experiments, materials that have been used for many years can elicit unexpected off-target effects. Therefore, non-viral vectors must be screened for several levels of toxicity to make them promising candidates.


Asunto(s)
Técnicas de Transferencia de Gen , Polietilenglicoles/farmacología , Polietileneimina/análogos & derivados , ARN Interferente Pequeño/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Forma de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citomegalovirus/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Genes Reporteros , Humanos , L-Lactato Deshidrogenasa/metabolismo , Luciferasas/metabolismo , Ratones , Polietileneimina/farmacología , Regiones Promotoras Genéticas/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Virus/metabolismo
9.
Inhal Toxicol ; 20(3): 319-37, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18300050

RESUMEN

In this article, we review and analyze different modes of exposure to ultrafine particles in order to assess particle-induced inflammatory responses and the underlying mechanisms in vitro and in vivo. Based on results from monocytic cells cultured under submerged conditions, we discuss (1) the impact of particle properties such as surface area and oxidative potential on lipid metabolism as a highly sensitive regulatory pathway and (2) the interference of diesel exhaust particles with toll-like receptor-mediated inflammatory responses. Furthermore, new developments of air-liquid interface exposure used as an alternative approach to simulate cell particle interactions are presented. In addition to the in vitro approaches, animal exposure studies are described that apply selected mouse models to elucidate potential allergic and inflammatory pulmonary responses and mast-cell-related mechanisms after particle exposure. Long-term inhalation of ultrafine particles might lead to irreversible changes in lung structure and function. Clinical studies addressing the characteristics of inflammatory airway cells are a promising approach to understand underlying pathophysiological mechanisms in chronic obstructive pulmonary disease. Finally, a potential outcome of human particle exposure is chronic cough in children. Here, discrimination between asthmatic and nonasthmatic cough by means of immunological parameters appears to be an important step toward improving diagnosis and therapy.


Asunto(s)
Inflamación/inducido químicamente , Macrófagos Alveolares/efectos de los fármacos , Monocitos/efectos de los fármacos , Material Particulado/efectos adversos , Animales , Biomarcadores/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Exposición por Inhalación , Metabolismo de los Lípidos , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Oxidación-Reducción , Tamaño de la Partícula , Receptores Toll-Like/metabolismo
10.
Proc Natl Acad Sci U S A ; 101(51): 17699-704, 2004 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-15591342

RESUMEN

The metabolism of poly(ADP-ribose) (PAR) is critical for genomic stability in multicellular eukaryotes. Here, we show that the failure to degrade PAR by means of disruption of the murine poly(ADP-ribose) glycohydrolase (PARG) gene unexpectedly causes early embryonic lethality and enhanced sensitivity to genotoxic stress. This lethality results from the failure to hydrolyze PAR, because PARG null embryonic day (E) 3.5 blastocysts accumulate PAR and concurrently undergo apoptosis. Moreover, embryonic trophoblast stem cell lines established from early PARG null embryos are viable only when cultured in medium containing the poly(ADP-ribose) polymerase inhibitor benzamide. Cells lacking PARG also show reduced growth, accumulation of PAR, and increased sensitivity to cytotoxicity induced by N-methyl-N'-nitro-N-nitrosoguanidine and menadione after benzamide withdrawal. These results provide compelling evidence that the failure to degrade PAR has deleterious consequences. Further, they define a role for PARG in embryonic development and a protective role in the response to genotoxic stress.


Asunto(s)
Pérdida del Embrión/inducido químicamente , Pérdida del Embrión/metabolismo , Glicósido Hidrolasas/metabolismo , Poli Adenosina Difosfato Ribosa/metabolismo , Animales , Apoptosis , Blastocisto/citología , Blastocisto/metabolismo , Proliferación Celular , Pérdida del Embrión/embriología , Pérdida del Embrión/enzimología , Femenino , Glicósido Hidrolasas/deficiencia , Glicósido Hidrolasas/genética , Metilnitronitrosoguanidina/farmacología , Metilnitronitrosoguanidina/toxicidad , Ratones , Ratones Noqueados , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Embarazo , Trofoblastos/citología , Trofoblastos/metabolismo , Vitamina K 3/farmacología , Vitamina K 3/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA