Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Nano Lett ; 14(3): 1202-7, 2014 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-24555574

RESUMO

Experimental data on dynamic interactions between individual nanoparticles and membrane processes at nanoscale, essential for biomedical applications of nanoparticles, remain scarce due to limitations of imaging techniques. We were able to follow single 200 nm carboxyl-modified particles interacting with identified membrane structures at the rate of 15 s/frame using a scanning ion conductance microscope modified for simultaneous high-speed topographical and fluorescence imaging. The imaging approach demonstrated here opens a new window into the complexity of nanoparticle-cell interactions.


Assuntos
Membrana Celular/metabolismo , Nanopartículas/química , Linhagem Celular , Humanos , Microscopia de Fluorescência/instrumentação , Microscopia de Fluorescência/métodos , Microscopia de Vídeo/instrumentação , Microscopia de Vídeo/métodos
2.
Carbon N Y ; 78: 26-37, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25780270

RESUMO

Multiwalled carbon nanotube (MWCNT) length is suggested to critically determine their pulmonary toxicity. This stems from in vitro and in vivo rodent studies and in vitro human studies using cell lines (typically cancerous). There is little data using primary human lung cells. We addressed this knowledge gap, using highly relevant, primary human alveolar cell models exposed to precisely synthesized and thoroughly characterized MWCNTs. In this work, transformed human alveolar type-I-like epithelial cells (TT1), primary human alveolar type-II epithelial cells (ATII) and alveolar macrophages (AM) were treated with increasing concentrations of MWCNTs before measuring cytotoxicity, inflammatory mediator release and MAP kinase signalling. Strikingly, we observed that short MWCNTs (~0.6 µm in length) induced significantly greater responses from the epithelial cells, whilst AM were particularly susceptible to long MWCNTs (~20 µm). These differences in the pattern of mediator release were associated with alternative profiles of JNK, p38 and ERK1/2 MAP kinase signal transduction within each cell type. This study, using highly relevant target human alveolar cells and well defined and characterized MWCNTs, shows marked cellular responses to the MWCNTs that vary according to the target cell type, as well as the aspect ratio of the MWCNT.

3.
Am J Physiol Cell Physiol ; 300(3): C466-76, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21160031

RESUMO

Rab27 small GTPases regulate secretion and movement of lysosome-related organelles such as T cell cytolytic granules and platelet-dense granules. Previous studies indicated that Rab27a and Rab27b are expressed in the murine lung suggesting that they regulate secretory processes in the lung. Consistent with those studies, we found that Rab27a and Rab27b are expressed in cell types that contain secretory granules: alveolar epithelial type II (AEII) and Clara cells. We then used Rab27a/Rab27b double knockout (DKO) mice to examine the functional consequence of loss of Rab27 proteins in the murine lung. Light and electron microscopy revealed a number of morphological changes in lungs from DKO mice when compared with those in control animals. In aged DKO mice we observed atrophy of the bronchiolar and alveolar epithelium with reduction of cells numbers, thinning of the bronchiolar epithelium and alveolar walls, and enlargement of alveolar airspaces. In these samples we also observed increased numbers of activated foamy alveolar macrophages and granulocyte containing infiltrates together with reduction in the numbers of Clara cells and AEII cells compared with control. At the ultrastructural level we observed accumulation of cytoplasmic membranes and vesicles in Clara cells. Meanwhile, AEII cells in DKO accumulated large mature lamellar bodies and lacked immature/precursor lamellar bodies. We hypothesize that the morphological changes observed at the ultrastructural level in DKO samples result from secretory defects in AEII and Clara cells and that over time these defects lead to atrophy of the epithelium.


Assuntos
Pulmão/anormalidades , Pulmão/metabolismo , Mucosa Respiratória/anormalidades , Mucosa Respiratória/metabolismo , Proteínas rab de Ligação ao GTP/deficiência , Animais , Atrofia , Pulmão/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Alvéolos Pulmonares/anormalidades , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/ultraestrutura , Mucosa Respiratória/ultraestrutura , Vesículas Secretórias/patologia , Vesículas Secretórias/ultraestrutura , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/ultraestrutura , Proteínas rab27 de Ligação ao GTP
4.
Am J Respir Crit Care Med ; 182(1): 73-82, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20203246

RESUMO

RATIONALE: Patients with idiopathic pulmonary fibrosis (IPF), a progressive disease with a dismal prognosis, exhibit an unexplained disparity of increased alveolar epithelial cell (AEC) apoptosis but reduced fibroblast apoptosis. OBJECTIVES: To examine whether the failure of patients with IPF to up-regulate cyclooxygenase (COX)-2, and thus the antifibrotic mediator prostaglandin (PG)E(2), accounts for this imbalance. METHODS: Fibroblasts and primary type II AECs were isolated from control and fibrotic human lung tissue. The effects of COX-2 inhibition and exogenous PGE(2) on fibroblast and AEC sensitivity to Fas ligand (FasL)-induced apoptosis were assessed. MEASUREMENTS AND MAIN RESULTS: IPF lung fibroblasts are resistant to FasL-induced apoptosis compared with control lung fibroblasts. Inhibition of COX-2 in control lung fibroblasts resulted in an apoptosis-resistant phenotype. Administration of PGE(2) almost doubled the rate of FasL-induced apoptosis in fibrotic lung fibroblasts compared with FasL alone. Conversely, in primary fibrotic lung type II AECs, PGE(2) protected against FasL-induced apoptosis. In human control and, to a greater extent, fibrotic lung fibroblasts, PGE(2) inhibits the phosphorylation of Akt, suggesting that regulation of this prosurvival protein kinase is an important mechanism by which PGE(2) modulates cellular apoptotic responses. CONCLUSIONS: The observation that PGE(2) deficiency results in increased AEC but reduced fibroblast sensitivity to apoptosis provides a novel pathogenic insight into the mechanisms driving persistent fibroproliferation in IPF.


Assuntos
Apoptose/fisiologia , Ciclo-Oxigenase 2/fisiologia , Dinoprostona/fisiologia , Fibroblastos/fisiologia , Fibrose Pulmonar Idiopática/fisiopatologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Células Cultivadas , Células Epiteliais/fisiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Alvéolos Pulmonares/fisiologia , Cicatrização/fisiologia
5.
Am J Respir Crit Care Med ; 179(5): 414-25, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19060230

RESUMO

RATIONALE: Studies in patients and experimental animals provide compelling evidence of the involvement of the major thrombin receptor, proteinase-activated receptor-1 (PAR(1)), and the potent chemokine, chemokine (CC motif) ligand-2 (CCL2)/monocyte chemotactic protein-1, in the pathogenesis of idiopathic pulmonary fibrosis (IPF). PAR(1) knockout mice are protected from bleomycin-induced lung inflammation and fibrosis and this protection is associated with marked attenuation in CCL2 induction. OBJECTIVES: The aim of this study was to determine which cell types represent the major source of PAR(1)-inducible CCL2 in the fibrotic lung. METHODS: Using immunohistochemistry and dual immunofluorescence, we examined PAR(1) and CCL2 expression in the bleomycin model and human IPF lung. PAR(1) and CCL2 gene expression was also assessed in laser-captured alveolar septae from patients with IPF. The ability of PAR(1) to induce CCL2 production by lung epithelial cells was also examined in vitro. MEASUREMENTS AND MAIN RESULTS: We report for the first time that PAR(1) and CCL2 are coexpressed and co-up-regulated on the activated epithelium in fibrotic areas in IPF. Similar observations were found in bleomycin-induced lung injury. Furthermore, we show that thrombin is a potent inducer of CCL2 gene expression and protein release by cultured lung epithelial cells via a PAR(1)-dependent mechanism. CONCLUSIONS: These data support the notion that PAR(1) activation on lung epithelial cells may represent an important mechanism leading to increased local CCL2 release in pulmonary fibrosis. Targeting PAR(1) on the pulmonary epithelium may offer a unique opportunity for therapeutic intervention in pulmonary fibrosis and other inflammatory and fibroproliferative conditions associated with excessive local generation of thrombin and CCL2 release.


Assuntos
Quimiocina CCL2/metabolismo , Fibrose Pulmonar/metabolismo , Receptor PAR-1/metabolismo , Sequência de Aminoácidos , Animais , Bleomicina , Estudos de Casos e Controles , Quimiocina CCL2/biossíntese , Quimiocina CCL2/genética , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Feminino , Expressão Gênica , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Pessoa de Meia-Idade , Dados de Sequência Molecular , Fibrose Pulmonar/genética , Fibrose Pulmonar/patologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptor PAR-1/biossíntese , Receptor PAR-1/genética , Receptores CCR2/metabolismo , Trombina/farmacologia
6.
Sci Rep ; 10(1): 20486, 2020 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-33235275

RESUMO

To overcome the scarcity of primary human alveolar epithelial cells for lung research, and the limitations of current cell lines to recapitulate the phenotype, functional and molecular characteristics of the healthy human alveolar epithelium, we have developed a new method to immortalise primary human alveolar epithelial lung cells using a non-viral vector to transfect the telomerase catalytic subunit (hTERT) and the simian virus 40 large-tumour antigen (SV40). Twelve strains of immortalised cells (ICs) were generated and characterised using molecular, immunochemical and morphological techniques. Cell proliferation and sensitivity to polystyrene nanoparticles (PS) were evaluated. ICs expressed caveolin-1, podoplanin and receptor for advanced glycation end-products (RAGE), and most cells were negative for alkaline phosphatase staining, indicating characteristics of AT1-like cells. However, most strains also contained some cells that expressed pro-surfactant protein C, classically described to be expressed only by AT2 cells. Thus, the ICs mimic the cellular heterogeneity in the human alveolar epithelium. These ICs can be passaged, replicate rapidly and remain confluent beyond 15 days. ICs showed differential sensitivity to positive and negatively charged PS nanoparticles, illustrating their potential value as an in vitro model to study respiratory bioreactivity. These novel ICs offer a unique resource to study human alveolar epithelial biology.


Assuntos
Células Epiteliais Alveolares/metabolismo , Vetores Genéticos/metabolismo , Fosfatase Alcalina/metabolismo , Células Epiteliais Alveolares/ultraestrutura , Linhagem Celular Transformada , Proliferação de Células , Respiração Celular , Sobrevivência Celular , Células Cultivadas , Humanos , Hidrodinâmica , Lipídeos/química , Nanopartículas/ultraestrutura , Tamanho da Partícula , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Eletricidade Estática , Transcrição Gênica , Transfecção
7.
Pharm Res ; 26(5): 1172-80, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19199008

RESUMO

PURPOSE: To evaluate the inflammatory response and barrier formation of a new alveolar type 1-like (transformed type I; TT1) cell line to establish its suitability for toxicity and drug transport studies. METHODS: TT1 and A549 cells were challenged with lipopolysaccharide (LPS). Secretion of inflammatory mediators was quantified by ELISA. The barrier properties of TT1 cells were evaluated by transepithelial electrical resistance (TEER), fluorescein sodium (flu-Na) apparent permeability (P(app)) and staining of zona occludens-1 (ZO-1). RESULTS: LPS stimulated similar levels of secretion of IL-6 and IL-8 in TT1 and A549 cells. TNF-alpha was not produced by either cell line. In contrast to A549 cells, TT1 cells did not secrete SLPI or elafin. TT1 cells produced maximal TEER of approximately 55 ohms cm(2) and flu-Na P(app) of approximately 6.0 x 10(-6) cm/s. ZO-1 staining was weak and discontinuous. Attempts to optimise culture conditions did not increase the barrier properties of the TT1 cell layers. CONCLUSIONS: The TT1 cell line models the alveolar inflammatory response to LPS challenge and provides a valuable complement to cell lines currently used in toxicity assays. However, under the experimental conditions used the TT1 cell line did not form the highly restrictive tight junctions which exist in vivo.


Assuntos
Linhagem Celular Transformada/imunologia , Linhagem Celular Transformada/metabolismo , Escherichia coli/imunologia , Lipopolissacarídeos/imunologia , Pulmão/citologia , Preparações Farmacêuticas/metabolismo , Linhagem Celular Transformada/efeitos dos fármacos , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular , Células Cultivadas , Citocinas/imunologia , Impedância Elétrica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Lipopolissacarídeos/farmacologia , Proteínas/metabolismo , Junções Íntimas/metabolismo , Junções Íntimas/ultraestrutura
8.
Pharmacol Ther ; 198: 189-205, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30796927

RESUMO

Lung cancer is an umbrella term for a subset of heterogeneous diseases that are collectively responsible for the most cancer-related deaths worldwide. Despite the tremendous progress made in understanding lung tumour biology, advances in early diagnosis, multimodal therapy and deciphering molecular mechanisms of drug resistance, overall curative outcomes remain low, especially in metastatic disease. Nanotechnology, in particular nanoparticles (NPs), continue to progressively impact the way by which tumours are diagnosed and treated. The unique physicochemical properties of materials at the nanoscale grant access to a diverse molecular toolkit that can be manipulated for use in respiratory oncology. This realisation has resulted in several clinically approved NP formulations and many more in clinical trials. However, NPs are not a panacea and have yet to be utilised to maximal effect in lung cancer, and medicine in a wider context. This review serves to: describe the complexity of lung cancer, the current diagnostic and therapeutic environment, and highlight the recent advancements of nanotechnology based approaches in diagnosis and treatment of respiratory malignancies. Finally, a brief outlook on the future directions of nanomedicine is provided; presently the full potential of the field is yet to be realised. By gleaning lessons and integrating advancements from neighbouring disciplines, nanomedicine can be elevated to a position where the current barriers that stymie full clinical impact are lifted.


Assuntos
Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/tratamento farmacológico , Nanotecnologia , Animais , Humanos
9.
ACS Appl Mater Interfaces ; 11(18): 16336-16346, 2019 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-30986026

RESUMO

Gold nanoparticles (AuNPs) have emerged as promising drug delivery candidates that can be leveraged for cancer therapy. Lung cancer (LC) is a heterogeneous disease that imposes a significant burden on society, with an unmet need for new therapies. Chemotherapeutic drugs such as afatinib (Afb), which is clinically approved for the treatment of epidermal growth factor receptor positive LC, is hydrophobic and has low bioavailability leading to spread around the body, causing severe side effects. Herein, we present a novel afatinib-AuNP formulation termed Afb-AuNPs, with the aim of improving drug efficacy and biocompatibility. This was achieved by synthesis of an alkyne-bearing Afb derivative and reaction with azide-functionalized lipoic acid using copper-catalyzed click chemistry, then conjugation to AuNPs via alkylthiol-gold bond formation. The Afb-AuNPs were found to possess up to 3.7-fold increased potency when administered to LC cells in vitro and were capable of significantly inhibiting cancer cell proliferation, as assessed by MTT assay and electric cell-substrate impedance sensing, respectively. Furthermore, when exposed to Afb-AuNPs, human alveolar epithelial type I-like cells, a model of the healthy lung epithelium, maintained viability and were found to release less proinflammatory cytokines when compared to free drug, demonstrating the biocompatibility of our formulation. This study provides a new platform for the development of nontraditional AuNP conjugates which can be applied to other molecules of therapeutic or diagnostic utility, with potential to be combined with photothermal therapy in other cancers.


Assuntos
Afatinib/química , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Nanoconjugados/administração & dosagem , Inibidores de Proteínas Quinases/administração & dosagem , Afatinib/administração & dosagem , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Humanos , Teste de Materiais , Nanopartículas Metálicas/administração & dosagem , Nanopartículas Metálicas/química , Nanoconjugados/química , Polietilenoglicóis/química , Inibidores de Proteínas Quinases/química , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/química
10.
Lab Chip ; 19(19): 3152-3161, 2019 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-31469131

RESUMO

The lung is a complex organ; it is both the initial barrier for inhaled agents and the site of metabolism and therapeutic effect for a subset of systemically administered drugs. Comprised of more than 40 cell types that are responsible for various important functions, the lung's complexity contributes to the subsequent challenges in developing complex in vitro co-culture models (also called microphysiological systems (MPS), complex in vitro models or organs-on-a-chip). Although there are multiple considerations and limitations in the development and qualification of such in vitro systems, MPS exhibit great promise in the fields of pharmacology and toxicology. Successful development and implementation of MPS models may enable mechanistic bridging between non-clinical species and humans, and increase clinical relevance of safety endpoints, while decreasing overall animal use. This article summarizes, from a biopharmaceutical industry perspective, essential elements for the development and qualification of lung MPS models. Its purpose is to guide MPS developers and manufacturers to expedite MPS utilization for safety assessment in the biopharmaceutical industry.


Assuntos
Técnicas de Cocultura , Dispositivos Lab-On-A-Chip , Pulmão/metabolismo , Técnicas Analíticas Microfluídicas , Modelos Biológicos , Técnicas de Cocultura/instrumentação , Humanos , Pulmão/efeitos dos fármacos , Pulmão/patologia , Técnicas Analíticas Microfluídicas/instrumentação
11.
Am J Respir Cell Mol Biol ; 39(5): 591-7, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18539954

RESUMO

Primary human alveolar type 2 (AT2) cells were immortalized by transduction with the catalytic subunit of telomerase and simian virus 40 large-tumor antigen. Characterization by immunochemical and morphologic methods demonstrated an AT1-like cell phenotype. Unlike primary AT2 cells, immortalized cells no longer expressed alkaline phosphatase, pro-surfactant protein C, and thyroid transcription factor-1, but expressed increased caveolin-1 and receptor for advanced glycation end products (RAGE). Live cell imaging using scanning ion conductance microscopy showed that the cuboidal primary AT2 cells were approximately 15 microm and enriched with surface microvilli, while the immortal AT1 cells were attenuated more than 40 microm, resembling these cells in situ. Transmission electron microscopy highlighted the attenuated morphology and showed endosomal vesicles in some immortal AT1 cells (but not primary AT2 cells) as found in situ. Particulate air pollution exacerbates cardiopulmonary disease. Interaction of ultrafine, nano-sized particles with the alveolar epithelium and/or translocation into the cardiovasculature may be a contributory factor. We hypothesized differential uptake of nanoparticles by AT1 and AT2 cells, depending on particle size and surface charge. Uptake of 50-nm and 1-microm fluorescent latex particles was investigated using confocal microscopy and scanning surface confocal microscopy of live cells. Fewer than 10% of primary AT2 cells internalized particles. In contrast, 75% immortal AT1 cells internalized negatively charged particles, while less than 55% of these cells internalized positively charged particles; charge, rather than size, mattered. The process was rapid: one-third of the total cell-associated negatively charged 50-nm particle fluorescence measured at 24 hours was internalized during the first hour. AT1 cells could be important in translocation of particles from the lung into the circulation.


Assuntos
Separação Celular/métodos , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Nanopartículas , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/metabolismo , Biomarcadores , Caveolina 1/metabolismo , Sobrevivência Celular , Células Cultivadas , Humanos , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Proteínas Nucleares/metabolismo , Fator Nuclear 1 de Tireoide , Fatores de Transcrição/metabolismo
12.
Toxicol In Vitro ; 48: 146-158, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29408664

RESUMO

There is a need to assess human and ecosystem health effects of copper oxide nanoparticles (CuO NPs), extensively used in many industrial products. Here, we aimed to determine the cytotoxicity and cellular mechanisms involved in the toxicity of CuO NPs in mussel cells (hemocytes and gill cells) in parallel with exposures to ionic Cu and bulk CuO, and to compare the sensitivity of mussel primary cells with a well-established human cell line (pulmonary TT1 cells). At similar doses, CuO NPs promoted dose-dependent cytotoxicity and increased reactive oxygen species (ROS) production in mussel and human cells. In mussel cells, ionic Cu was more toxic than CuO NPs and the latter more than bulk CuO. Ionic Cu and CuO NPs increased catalase and acid phosphatase activities in both mussel cells and decreased gill cells Na-K-ATPase activity. All Cu forms produced DNA damage in hemocytes, whereas in gill cells only ionic Cu and CuO NPs were genotoxic. Induction of the MXR transport activity was found in gill cells exposed to all forms of Cu and in hemocytes exposed to ionic Cu and CuO NPs. Phagocytosis increased only in hemocytes exposed to CuO NPs, indicating a nanoparticle-specific immunostimulatory effect. In conclusion, toxicity of CuO NPs is driven by ROS in human and mussel cells. Mussel cells respond to CuO NP exposure by triggering an array of defensive mechanisms.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Cobre/toxicidade , Nanopartículas Metálicas/toxicidade , Mytilus/citologia , Fosfatase Ácida/metabolismo , Animais , Catalase/metabolismo , Linhagem Celular , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/ultraestrutura , Dano ao DNA , Brânquias/citologia , Brânquias/efeitos dos fármacos , Hemócitos/efeitos dos fármacos , Humanos , Mytilus/efeitos dos fármacos , Tamanho da Partícula , Fagocitose/efeitos dos fármacos , Cultura Primária de Células , Espécies Reativas de Oxigênio/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Especificidade da Espécie
13.
Oncotarget ; 8(30): 48737-48754, 2017 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-28467787

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a progressive disease that usually affects elderly people. It has a poor prognosis and there are limited therapies. Since epigenetic alterations are associated with IPF, histone deacetylase (HDAC) inhibitors offer a novel therapeutic strategy to address the unmet medical need. This study investigated the potential of romidepsin, an FDA-approved HDAC inhibitor, as an anti-fibrotic treatment and evaluated biomarkers of target engagement that may have utility in future clinical trials. The anti-fibrotic effects of romidepsin were evaluated both in vitro and in vivo together with any harmful effect on alveolar type II cells (ATII). Bronchoalveolar lavage fluid (BALF) from IPF or control donors was analyzed for the presence of lysyl oxidase (LOX). In parallel with an increase in histone acetylation, romidepsin potently inhibited fibroblast proliferation, myofibroblast differentiation and LOX expression. ATII cell numbers and their lamellar bodies were unaffected. In vivo, romidepsin inhibited bleomycin-induced pulmonary fibrosis in association with suppression of LOX expression. LOX was significantly elevated in BALF of IPF patients compared to controls. These data show the anti-fibrotic effects of romidepsin, supporting its potential use as novel treatment for IPF with LOX as a companion biomarker for evaluation of early on-target effects.


Assuntos
Depsipeptídeos/farmacologia , Inibidores de Histona Desacetilases/farmacologia , Fibrose Pulmonar Idiopática/genética , Fibrose Pulmonar Idiopática/metabolismo , Acetilação , Biomarcadores , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Depsipeptídeos/uso terapêutico , Epigênese Genética , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Inibidores de Histona Desacetilases/uso terapêutico , Histona Desacetilases/metabolismo , Histonas/metabolismo , Humanos , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/patologia , Masculino
14.
ACS Nano ; 10(5): 5070-85, 2016 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-27035850

RESUMO

Uptake and translocation of short functionalized multi-walled carbon nanotubes (short-fMWCNTs) through the pulmonary respiratory epithelial barrier depend on physicochemical property and cell type. Two monoculture models, immortalized human alveolar epithelial type 1 (TT1) cells and primary human alveolar epithelial type 2 cells (AT2), which constitute the alveolar epithelial barrier, were employed to investigate the uptake and transport of 300 and 700 nm in length, poly(4-vinylpyridine)-functionalized, multi-walled carbon nanotubes (p(4VP)-MWCNTs) using quantitative imaging and spectroscopy techniques. The p(4VP)-MWCNT exhibited no toxicity on TT1 and AT2 cells, but significantly decreased barrier integrity (*p < 0.01). Uptake of p(4VP)-MWCNTs was observed in 70% of TT1 cells, correlating with compromised barrier integrity and basolateral p(4VP)-MWCNT translocation. There was a small but significantly greater uptake of 300 nm p(4VP)-MWCNTs than 700 nm p(4VP)-MWCNTs by TT1 cells. Up to 3% of both the 300 and 700 nm p(4VP)-MWCNTs reach the basal chamber; this relatively low amount arose because the supporting transwell membrane minimized the amount of p(4VP)-MWCNT translocating to the basal chamber, seen trapped between the basolateral cell membrane and the membrane. Only 8% of AT2 cells internalized p(4VP)-MWCNT, accounting for 17% of applied p(4VP)-MWCNT), with transient effects on barrier function, which initially fell then returned to normal; there was no MWCNT basolateral translocation. The transport rate was MWCNT length modulated. The comparatively lower p(4VP)-MWCNT uptake by AT2 cells is proposed to reflect a primary barrier effect of type 2 cell secretions and the functional differences between the type 1 and type 2 alveolar epithelial cells.


Assuntos
Células Epiteliais , Pulmão/citologia , Nanotubos de Carbono , Alvéolos Pulmonares/citologia , Técnicas de Cultura de Células , Humanos , Mucosa Respiratória
15.
Colloids Surf B Biointerfaces ; 145: 167-175, 2016 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-27182651

RESUMO

Accompanying increased commercial applications and production of silver nanomaterials is an increased probability of human exposure, with inhalation a key route. Nanomaterials that deposit in the pulmonary alveolar region following inhalation will interact firstly with pulmonary surfactant before they interact with the alveolar epithelium. It is therefore critical to understand the effects of human pulmonary surfactant when evaluating the inhalation toxicity of silver nanoparticles. In this study, we evaluated the toxicity of AgNPs on human alveolar type-I-like epithelial (TT1) cells in the absence and presence of Curosurf(®) (a natural pulmonary surfactant substitute), hypothesising that the pulmonary surfactant would act to modify toxicity. We demonstrated that 20nm citrate-capped AgNPs induce toxicity in human alveolar type I-like epithelial cells and, in agreement with our hypothesis, that pulmonary surfactant acts to mitigate this toxicity, possibly through reducing AgNP dissolution into cytotoxic Ag(+) ions. For example, IL-6 and IL-8 release by TT1 cells significantly increased 10.7- and 35-fold, respectively (P<0.01), 24h after treatment with 25µg/ml AgNPs. In contrast, following pre-incubation of AgNPs with Curosurf(®), this effect was almost completely abolished. We further determined that the mechanism of this toxicity is likely associated with Ag(+) ion release and lysosomal disruption, but not with increased reactive oxygen species generation. This study provides a critical understanding of the toxicity of AgNPs in target human alveolar type-I-like epithelial cells and the role of pulmonary surfactant in mitigating this toxicity. The observations reported have important implications for the manufacture and application of AgNPs, in particular for applications involving use of aerosolised AgNPs.


Assuntos
Células Epiteliais/patologia , Nanopartículas Metálicas/toxicidade , Alvéolos Pulmonares/patologia , Surfactantes Pulmonares/farmacologia , Prata/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Humanos , Mediadores da Inflamação/metabolismo , Íons , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Nanopartículas Metálicas/ultraestrutura , Espécies Reativas de Oxigênio/metabolismo
16.
Nanotoxicology ; 9(4): 482-92, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25137294

RESUMO

There can be significant variability between bioreactivity studies of nanomaterials that are apparently the same, possibly reflecting differences in the models used and differing sources of experimental material. In this study, we have generated two crystal forms of titanium dioxide nanoparticles (nano-TiO2), pure anatase and pure rutile to address the hypothesis that the bioreactivity of these nanoparticles with human alveolar epithelium will depend on their crystal phase. We used a human alveolar type-I-like epithelial cell model (TT1; generated in-house from primary human alveolar epithelial type II cells); these cells cover 95% of the alveolar epithelial surface area and are an important target cell for inhaled nanomaterials. Using literature as a guide, we hypothesised that pure anatase nano-TiO2 would display greater bioreactivity with TT1 cells in comparison to pure rutile nano-TiO2. However, we found the profile and pattern of inflammatory mediator release was similar between these two nano-TiO2 formats, although pure rutile treatment caused a small, but consistently greater, response for IL-6, IL-8 and MCP-1. Interestingly, the temporal induction of oxidative stress (increased reactive oxygen species levels and depleted glutathione) varied markedly between the different nano-TiO2 formats. We have shown that a combination of using nanomaterials synthesised specifically for toxicological study and the use of a highly relevant, reproducible human lung cell model, offers a useful approach to delineating the physicochemical properties of nanomaterials that may be important in their cellular reactivity.


Assuntos
Nanopartículas Metálicas/química , Alvéolos Pulmonares/efeitos dos fármacos , Titânio/química , Quimiocinas/metabolismo , Cristalização , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/efeitos dos fármacos , Humanos , Microscopia Eletrônica de Transmissão , Alvéolos Pulmonares/citologia , Espécies Reativas de Oxigênio/metabolismo
17.
ACS Nano ; 8(11): 11778-89, 2014 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-25360809

RESUMO

The ability to manipulate the size and surface properties of nanomaterials makes them a promising vector for improving drug delivery and efficacy. Inhalation is a desirable route of administration as nanomaterials preferentially deposit in the alveolar region, a large surface area for drug absorption. However, as yet, the mechanisms by which particles translocate across the alveolar epithelial layer are poorly understood. Here we show that human alveolar type I epithelial cells internalize nanoparticles, whereas alveolar type II epithelial cells do not, and that nanoparticles translocate across the epithelial monolayer but are unable to penetrate the tight junctions between cells, ruling out paracellular translocation. Furthermore, using siRNA, we demonstrate that 50 nm nanoparticles enter largely by passive diffusion and are found in the cytoplasm, whereas 100 nm nanoparticles enter primarily via clathrin- and also caveolin-mediated endocytosis and are found in endosomes. Functionalization of nanoparticles increases their uptake and enhances binding of surfactant which further promotes uptake. Thus, we demonstrate that uptake and translocation across the pulmonary epithelium is controlled by alveolar type I epithelial cells, and furthermore, we highlight a number of factors that should be considered when designing new nanomedicines in order to improve drug delivery to the lung.


Assuntos
Nanopartículas , Alvéolos Pulmonares/metabolismo , Transporte Biológico , Linhagem Celular Transformada , Humanos
18.
Biomaterials ; 35(17): 4729-38, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24631251

RESUMO

The use of a thermochemical grafting approach provides a versatile means to functionalise as-synthesised, bulk multi-walled carbon nanotubes (MWNTs) without altering their inherent structure. The associated retention of properties is desirable for a wide range of commercial applications, including for drug delivery and medical purposes; it is also pertinent to studies of intrinsic toxicology. A systematic series of water-compatible MWNTs, with diameter around 12 nm have been prepared, to provide structurally-equivalent samples predominantly stabilised by anionic, cationic, or non-ionic groups. The surface charge of MWNTs was controlled by varying the grafting reagents and subsequent post-functionalisation modifications. The degree of grafting was established by thermal analysis (TGA). High resolution transmission electron microscope (HRTEM) and Raman measurements confirmed that the structural framework of the MWNTs was unaffected by the thermochemical treatment, in contrast to a conventional acid-oxidised control which was severely damaged. The effectiveness of the surface modification was demonstrated by significantly improved solubility and stability in both water and cell culture medium, and further quantified by zeta-potential analysis. The grafted MWNTs exhibited relatively low bioreactivity on transformed human alveolar epithelial type 1-like cells (TT1) following 24 h exposure as demonstrated by 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) and lactate dehydrogenase release (LDH) assays. The exposure of TT1 cells to MWNTs suppressed the release of the inflammatory mediators, interleukin 6 (IL-6) and interleukin 8 (IL-8). TEM cell uptake studies indicated efficient cellular entry of MWNTs into TT1 cells, via a range of mechanisms. Cationic MWNTs showed a more substantial interaction with TT1 cell membranes than anionic MWNTs, demonstrating a surface charge effect on cell uptake.


Assuntos
Materiais Biocompatíveis/química , Nanotubos de Carbono/química , Materiais Biocompatíveis/metabolismo , Materiais Biocompatíveis/toxicidade , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Endocitose , Humanos , Íons/química , Nanotubos de Carbono/análise , Nanotubos de Carbono/toxicidade , Nanotubos de Carbono/ultraestrutura , Solubilidade , Eletricidade Estática , Propriedades de Superfície , Água/química
19.
Pharmacol Ther ; 140(2): 176-85, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23811125

RESUMO

Recent advances in nanotechnology have revolutionised all aspects of life, from engineering to cosmetics. One of the most exciting areas of development is that of nanomedicine. Due to their size (less than 100nm in one aspect), nanoparticles exhibit properties that are unlike that of the same material in bulk size. These unique properties are being exploited to create new diagnostics and therapeutics for application in a broad spectrum of organ systems. Indeed, nanoparticles are already being developed as effective carriers of drugs to target regions of the body that were previously hard to access using traditional drug formulation methods. However, in addition to their role as a vehicle for drug delivery, nanoparticles themselves have the potential to have therapeutic benefit. Through manipulation of their elemental composition, size, shape, charge and surface modification or functionalisation it may be possible to target particles to specific organs where they may elicit their therapeutic effect. In this review we will focus on the recent advances in nanotechnology for therapeutic applications with a particular focus on the respiratory system, cancer and vaccinations. In addition we will also address developments in the field of nanotoxicology and the need for concomitant studies in to the toxicity of emerging nanotechnologies. It is possible that the very properties that make nanoparticles a desirable technology for therapeutic intervention may also lead to adverse health effects. It is thus important to determine, and appreciate, the fine balance between the efficacy and toxicity of nanomedicine.


Assuntos
Sistemas de Liberação de Medicamentos , Nanomedicina , Animais , Humanos , Pulmão/metabolismo , Pneumopatias/tratamento farmacológico , Neoplasias/tratamento farmacológico , Vacinas
20.
Nanoscale ; 5(20): 9839-47, 2013 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-23970174

RESUMO

Silver nanowires (AgNWs) are being developed for use in optoelectronics. However before widespread usage, it is crucial to determine their potential effects on human health. It is accepted that Ag nanoparticles (AgNPs) exert toxic effects by releasing Ag(+) ions, but much less is known about whether Ag(+) reacts with compounds, or any downstream bioactive effects of transformed AgNPs. Analytical high-resolution transmission electron microscopy has been employed to elucidate cellular uptake and reactivity of AgNWs inside human alveolar epithelial type 1-like cells. AgNWs were observed in the cytoplasm and membrane-bound vesicles, and precipitation of Ag2S within the cell occurred after 1 h exposure. Cell viability studies showed no evidence of cytotoxicity and reactive oxygen species were not observed on exposure of cells to AgNWs. We suggest that Ag2S formation acts as a 'trap' for free Ag(+), significantly limiting short-term toxicological effects - with important consequences for the safety of Ag-nanomaterials to human health.


Assuntos
Nanofios/química , Prata/química , Sulfetos/química , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Eletrônica , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Humanos , Inativação Metabólica , Microscopia Eletrônica de Transmissão , Nanofios/toxicidade , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA