Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 3271, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38627373

RESUMO

Selective binding of TCR-like antibodies that target a single tumour-specific peptide antigen presented by human leukocyte antigens (HLA) is the absolute prerequisite for their therapeutic suitability and patient safety. To date, selectivity assessment has been limited to peptide library screening and predictive modeling. We developed an experimental platform to de novo identify interactomes of TCR-like antibodies directly in human tissues using mass spectrometry. As proof of concept, we confirm the target epitope of a MAGE-A4-specific TCR-like antibody. We further determine cross-reactive peptide sequences for ESK1, a TCR-like antibody with known off-target activity, in human liver tissue. We confirm off-target-induced T cell activation and ESK1-mediated liver spheroid killing. Off-target sequences feature an amino acid motif that allows a structural groove-coordination mimicking that of the target peptide, therefore allowing the interaction with the engager molecule. We conclude that our strategy offers an accurate, scalable route for evaluating the non-clinical safety profile of TCR-like antibody therapeutics prior to first-in-human clinical application.


Assuntos
Anticorpos , Peptídeos , Humanos , Linhagem Celular Tumoral , Peptídeos/química , Antígenos de Neoplasias , Receptores de Antígenos de Linfócitos T/metabolismo
2.
Front Pharmacol ; 14: 1114739, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36959848

RESUMO

Prolonged exposure to environmental respirable toxicants can lead to the development and worsening of severe respiratory diseases such as asthma, chronic obstructive pulmonary disease (COPD) and fibrosis. The limited number of FDA-approved inhaled drugs for these serious lung conditions has led to a shift from in vivo towards the use of alternative in vitro human-relevant models to better predict the toxicity of inhaled particles in preclinical research. While there are several inhalation exposure models for the upper airways, the fragile and dynamic nature of the alveolar microenvironment has limited the development of reproducible exposure models for the distal lung. Here, we present a mechanistic approach using a new generation of exposure systems, the Cloud α AX12. This novel in vitro inhalation tool consists of a cloud-based exposure chamber (VITROCELL) that integrates the breathing AXLung-on-chip system (AlveoliX). The ultrathin and porous membrane of the AX12 plate was used to create a complex multicellular model that enables key physiological culture conditions: the air-liquid interface (ALI) and the three-dimensional cyclic stretch (CS). Human-relevant cellular models were established for a) the distal alveolar-capillary interface using primary cell-derived immortalized alveolar epithelial cells (AXiAECs), macrophages (THP-1) and endothelial (HLMVEC) cells, and b) the upper-airways using Calu3 cells. Primary human alveolar epithelial cells (AXhAEpCs) were used to validate the toxicity results obtained from the immortalized cell lines. To mimic in vivo relevant aerosol exposures with the Cloud α AX12, three different models were established using: a) titanium dioxide (TiO2) and zinc oxide nanoparticles b) polyhexamethylene guanidine a toxic chemical and c) an anti-inflammatory inhaled corticosteroid, fluticasone propionate (FL). Our results suggest an important synergistic effect on the air-blood barrier sensitivity, cytotoxicity and inflammation, when air-liquid interface and cyclic stretch culture conditions are combined. To the best of our knowledge, this is the first time that an in vitro inhalation exposure system for the distal lung has been described with a breathing lung-on-chip technology. The Cloud α AX12 model thus represents a state-of-the-art pre-clinical tool to study inhalation toxicity risks, drug safety and efficacy.

3.
Front Toxicol ; 4: 840606, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35832493

RESUMO

The evaluation of inhalation toxicity, drug safety and efficacy assessment, as well as the investigation of complex disease pathomechanisms, are increasingly relying on in vitro lung models. This is due to the progressive shift towards human-based systems for more predictive and translational research. While several cellular models are currently available for the upper airways, modelling the distal alveolar region poses several constraints that make the standardization of reliable alveolar in vitro models relatively difficult. In this work, we present a new and reproducible alveolar in vitro model, that combines a human derived immortalized alveolar epithelial cell line (AXiAEC) and organ-on-chip technology mimicking the lung alveolar biophysical environment (AXlung-on-chip). The latter mimics key features of the in vivo alveolar milieu: breathing-like 3D cyclic stretch (10% linear strain, 0.2 Hz frequency) and an ultrathin, porous and elastic membrane. AXiAECs cultured on-chip were characterized for their alveolar epithelial cell markers by gene and protein expression. Cell barrier properties were examined by TER (Transbarrier Electrical Resistance) measurement and tight junction formation. To establish a physiological model for the distal lung, AXiAECs were cultured for long-term at air-liquid interface (ALI) on-chip. To this end, different stages of alveolar damage including inflammation (via exposure to bacterial lipopolysaccharide) and the response to a profibrotic mediator (via exposure to Transforming growth factor ß1) were analyzed. In addition, the expression of relevant host cell factors involved in SARS-CoV-2 infection was investigated to evaluate its potential application for COVID-19 studies. This study shows that AXiAECs cultured on the AXlung-on-chip exhibit an enhanced in vivo-like alveolar character which is reflected into: 1) Alveolar type 1 (AT1) and 2 (AT2) cell specific phenotypes, 2) tight barrier formation (with TER above 1,000 Ω cm2) and 3) reproducible long-term preservation of alveolar characteristics in nearly physiological conditions (co-culture, breathing, ALI). To the best of our knowledge, this is the first time that a primary derived alveolar epithelial cell line on-chip representing both AT1 and AT2 characteristics is reported. This distal lung model thereby represents a valuable in vitro tool to study inhalation toxicity, test safety and efficacy of drug compounds and characterization of xenobiotics.

4.
Front Med (Lausanne) ; 8: 644678, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34026781

RESUMO

The coronavirus disease 2019 (COVID-19) pandemic has caused considerable socio-economic burden, which fueled the development of treatment strategies and vaccines at an unprecedented speed. However, our knowledge on disease recovery is sparse and concerns about long-term pulmonary impairments are increasing. Causing a broad spectrum of symptoms, COVID-19 can manifest as acute respiratory distress syndrome (ARDS) in the most severely affected patients. Notably, pulmonary infection with Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2), the causing agent of COVID-19, induces diffuse alveolar damage (DAD) followed by fibrotic remodeling and persistent reduced oxygenation in some patients. It is currently not known whether tissue scaring fully resolves or progresses to interstitial pulmonary fibrosis. The most aggressive form of pulmonary fibrosis is idiopathic pulmonary fibrosis (IPF). IPF is a fatal disease that progressively destroys alveolar architecture by uncontrolled fibroblast proliferation and the deposition of collagen and extracellular matrix (ECM) proteins. It is assumed that micro-injuries to the alveolar epithelium may be induced by inhalation of micro-particles, pathophysiological mechanical stress or viral infections, which can result in abnormal wound healing response. However, the exact underlying causes and molecular mechanisms of lung fibrosis are poorly understood due to the limited availability of clinically relevant models. Recently, the emergence of SARS-CoV-2 with the urgent need to investigate its pathogenesis and address drug options, has led to the broad application of in vivo and in vitro models to study lung diseases. In particular, advanced in vitro models including precision-cut lung slices (PCLS), lung organoids, 3D in vitro tissues and lung-on-chip (LOC) models have been successfully employed for drug screens. In order to gain a deeper understanding of SARS-CoV-2 infection and ultimately alveolar tissue regeneration, it will be crucial to optimize the available models for SARS-CoV-2 infection in multicellular systems that recapitulate tissue regeneration and fibrotic remodeling. Current evidence for SARS-CoV-2 mediated pulmonary fibrosis and a selection of classical and novel lung models will be discussed in this review.

5.
Commun Biol ; 4(1): 168, 2021 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-33547387

RESUMO

The air-blood barrier with its complex architecture and dynamic environment is difficult to mimic in vitro. Lung-on-a-chips enable mimicking the breathing movements using a thin, stretchable PDMS membrane. However, they fail to reproduce the characteristic alveoli network as well as the biochemical and physical properties of the alveolar basal membrane. Here, we present a lung-on-a-chip, based on a biological, stretchable and biodegradable membrane made of collagen and elastin, that emulates an array of tiny alveoli with in vivo-like dimensions. This membrane outperforms PDMS in many ways: it does not absorb rhodamine-B, is biodegradable, is created by a simple method, and can easily be tuned to modify its thickness, composition and stiffness. The air-blood barrier is reconstituted using primary lung alveolar epithelial cells from patients and primary lung endothelial cells. Typical alveolar epithelial cell markers are expressed, while the barrier properties are preserved for up to 3 weeks.


Assuntos
Elasticidade/fisiologia , Dispositivos Lab-On-A-Chip , Pulmão/citologia , Membranas Artificiais , Alvéolos Pulmonares/fisiologia , Células Epiteliais Alveolares/citologia , Células Epiteliais Alveolares/fisiologia , Barreira Alveolocapilar/citologia , Barreira Alveolocapilar/fisiologia , Comunicação Celular/fisiologia , Permeabilidade da Membrana Celular/fisiologia , Técnicas de Cocultura/instrumentação , Técnicas de Cocultura/métodos , Humanos , Pulmão/fisiologia , Microtecnologia , Cultura Primária de Células/instrumentação , Cultura Primária de Células/métodos , Alvéolos Pulmonares/citologia , Estresse Mecânico , Engenharia Tecidual/instrumentação , Engenharia Tecidual/métodos , Alicerces Teciduais/química
6.
Respir Res ; 21(1): 29, 2020 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-31992294

RESUMO

After publication of our article [1], we have been notified that an extra alpha symbol (α) was mistakenly added at the beginning of the title.

7.
Respir Res ; 21(1): 25, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31941499

RESUMO

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a chronic fatal lung disease without a cure and new drug strategies are urgently needed. Differences in behavior between diseased and healthy cells are well known and drug response can be different between cells isolated from IPF patients and controls. The macrolide Azithromycin (AZT) has anti-inflammatory and immunomodulatory properties. Recently anti-fibrotic effects have been described. However, the anti-fibrotic effects on primary IPF-fibroblasts (FB) directly compared to control-FB are unknown. We hypothesized that IPF-FB react differently to AZT in terms of anti-fibrotic effects. METHODS: Primary normal human lung and IPF-FB were exposed to TGF-ß (5 ng/ml), Azithromycin (50 µM) alone or in combination prior to gene expression analysis. Pro-collagen Iα1 secretion was assessed by ELISA and protein expression by western blot (αSMA, Fibronectin, ATP6V1B2, LC3 AB (II/I), p62, Bcl-xL). Microarray analysis was performed to screen involved genes and pathways after Azithromycin treatment in control-FB. Apoptosis and intraluminal lysosomal pH were analyzed by flow cytometry. RESULTS: AZT significantly reduced collagen secretion in TGF-ß treated IPF-FB compared to TGF-ß treatment alone, but not in control-FB. Pro-fibrotic gene expression was similarly reduced after AZT treatment in IPF and control-FB. P62 and LC3II/I western blot revealed impaired autophagic flux after AZT in both control and IPF-FB with significant increase of LC3II/I after AZT in control and IPF-FB, indicating enhanced autophagy inhibition. Early apoptosis was significantly higher in TGF-ß treated IPF-FB compared to controls after AZT. Microarray analysis of control-FB treated with AZT revealed impaired lysosomal pathways. The ATPase and lysosomal pH regulator ATP6V0D2 was significantly less increased after additional AZT in IPF-FB compared to controls. Lysosomal function was impaired in both IPF and control FB, but pH was significantly more increased in TGF-ß treated IPF-FB. CONCLUSION: We report different treatment responses after AZT with enhanced anti-fibrotic and pro-apoptotic effects in IPF compared to control-FB. Possibly impaired lysosomal function contributes towards these effects. In summary, different baseline cell phenotype and behavior of IPF and control cells contribute to enhanced anti-fibrotic and pro-apoptotic effects in IPF-FB after AZT treatment and strengthen its role as a new potential anti-fibrotic compound, that should further be evaluated in clinical studies.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Azitromicina/farmacologia , Fibroblastos/efeitos dos fármacos , Fibrose Pulmonar Idiopática , Pulmão/efeitos dos fármacos , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Apoptose/fisiologia , Azitromicina/uso terapêutico , Células Cultivadas , Fibroblastos/patologia , Humanos , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/patologia , Pulmão/patologia , Fator de Crescimento Transformador beta/farmacologia
8.
Eur J Pharm Biopharm ; 144: 11-17, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31499161

RESUMO

With rapid advances in micro-fabrication processes and the availability of biologically-relevant lung cells, the development of lung-on-chip platforms is offering novel avenues for more realistic inhalation assays in pharmaceutical research, and thereby an opportunity to depart from traditional in vitro lung assays. As advanced models capturing the cellular pulmonary make-up at an air-liquid interface (ALI), lung-on-chips emulate both morphological features and biological functionality of the airway barrier with the ability to integrate respiratory breathing motions and ensuing tissue strains. Such in vitro systems allow importantly to mimic more realistic physiological respiratory flow conditions, with the opportunity to integrate physically-relevant transport determinants of aerosol inhalation therapy, i.e. recapitulating the pathway from airborne flight to deposition on the airway lumen. In this short opinion, we discuss such points and describe how these attributes are paving new avenues for exploring improved drug carrier designs (e.g. shape, size, etc.) and targeting strategies (e.g. conductive vs. respiratory regions) amongst other. We argue that while technical challenges still lie along the way in rendering in vitro lung-on-chip platforms more widespread across the general pharmaceutical research community, significant momentum is steadily underway in accelerating the prospect of establishing these as in vitro "gold standards".


Assuntos
Aerossóis/metabolismo , Bioensaio/métodos , Pulmão/metabolismo , Administração por Inalação , Portadores de Fármacos/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Humanos , Modelos Biológicos , Tamanho da Partícula , Respiração/efeitos dos fármacos , Terapia Respiratória/métodos
9.
Neoplasia ; 21(2): 185-196, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30591423

RESUMO

Cell lines are essential tools to standardize and compare experimental findings in basic and translational cancer research. The current dogma states that cancer stem cells feature an increased tumor initiation capacity and are also chemoresistant. Here, we identified and comprehensively characterized three morphologically distinct cellular subtypes in the non-small cell lung cancer cell line A549 and challenge the current cancer stem cell dogma. Subtype-specific cellular morphology is maintained during short-term culturing, resulting in the formation of holoclonal, meroclonal, and paraclonal colonies. A549 holoclone cells were characterized by an epithelial and stem-like phenotype, paraclone cells featured a mesenchymal phenotype, whereas meroclone cells were phenotypically intermediate. Cell-surface marker expression of subpopulations changed over time, indicating an active epithelial-to-mesenchymal transition (EMT), in vitro and in vivo. EMT has been associated with the overexpression of the immunomodulators PD-L1 and PD-L2, which were 37- and 235-fold overexpressed in para- versus holoclone cells, respectively. We found that DNA methylation is involved in epigenetic regulation of marker expression. Holoclone cells were extremely sensitive to cisplatin and radiotherapy in vitro, whereas paraclone cells were highly resistant. However, inhibition of the receptor tyrosine kinase AXL, whose expression is associated with an EMT, specifically targeted the otherwise highly resistant paraclone cells. Xenograft tumor formation capacity was 24- and 269-fold higher in holo- than mero- and paraclone cells, respectively. Our results show that A549 subpopulations might serve as a unique system to explore the network of stemness, cellular plasticity, tumor initiation capacity, invasive and metastatic potential, and chemo/radiotherapy resistance.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Transformação Celular Neoplásica , Transição Epitelial-Mesenquimal , Neoplasias Pulmonares/patologia , Células-Tronco Neoplásicas/metabolismo , Células A549 , Animais , Biomarcadores , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/genética , Dano ao DNA , Metilação de DNA , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal/genética , Perfilação da Expressão Gênica , Humanos , Imunofenotipagem , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Camundongos , Células-Tronco Neoplásicas/patologia , Transcriptoma
10.
Sci Rep ; 8(1): 14359, 2018 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-30254327

RESUMO

Organs-on-chips have the potential to improve drug development efficiency and decrease the need for animal testing. For the successful integration of these devices in research and industry, they must reproduce in vivo contexts as closely as possible and be easy to use. Here, we describe a 'breathing' lung-on-chip array equipped with a passive medium exchange mechanism that provide an in vivo-like environment to primary human lung alveolar cells (hAEpCs) and primary lung endothelial cells. This configuration allows the preservation of the phenotype and the function of hAEpCs for several days, the conservation of the epithelial barrier functionality, while enabling simple sampling of the supernatant from the basal chamber. In addition, the chip design increases experimental throughput and enables trans-epithelial electrical resistance measurements using standard equipment. Biological validation revealed that human primary alveolar type I (ATI) and type II-like (ATII) epithelial cells could be successfully cultured on the chip over multiple days. Moreover, the effect of the physiological cyclic strain showed that the epithelial barrier permeability was significantly affected. Long-term co-culture of primary human lung epithelial and endothelial cells demonstrated the potential of the lung-on-chip array for reproducible cell culture under physiological conditions. Thus, this breathing lung-on-chip array, in combination with patients' primary ATI, ATII, and lung endothelial cells, has the potential to become a valuable tool for lung research, drug discovery and precision medicine.


Assuntos
Alvéolos Pulmonares/citologia , Respiração , Análise Serial de Tecidos/métodos , Células Epiteliais/citologia , Desenho de Equipamento , Humanos , Alvéolos Pulmonares/fisiologia , Reprodutibilidade dos Testes , Análise Serial de Tecidos/instrumentação
11.
Angiogenesis ; 21(4): 861-871, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29967964

RESUMO

Idiopathic pulmonary fibrosis is characterized by a progressive scarring and stiffening of the peripheral lung tissue that decreases lung function. Over the course of the disease, the lung microvasculature undergoes extensive remodeling. There is increased angiogenesis around fibrotic foci and an absence of microvessels within the foci. To elucidate how the anti-fibrotic drug nintedanib acts on vascular remodeling, we used an in vitro model of perfusable microvessels made with primary endothelial cells and primary lung fibroblasts in a microfluidic chip. The microvasculature model allowed us to study the impact of nintedanib on permeability, vascularized area, and cell-cell interactions. The anti-vasculogenic impact of nintedanib was visible at the minimal concentrations of 10 nM, showing a significant increase in vessel permeability. Furthermore, nintedanib decreased microvessel density, diameter, and influenced fibroblast organization around endothelial microvessels. These results show that nintedanib acts on the endothelial network formation and endothelial-perivascular interactions. Advanced in vitro microvasculature models may thus serve to pinpoint the mechanistic effect of anti-fibrotic drugs on the microvascular remodeling in 3D and refine findings from animal studies.


Assuntos
Fibroblastos , Fibrose Pulmonar Idiopática , Indóis/farmacologia , Pulmão , Microvasos , Remodelação Vascular/efeitos dos fármacos , Técnicas de Cultura de Células , Técnicas de Cocultura , Relação Dose-Resposta a Droga , Fibroblastos/metabolismo , Fibroblastos/patologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Dispositivos Lab-On-A-Chip , Pulmão/irrigação sanguínea , Pulmão/metabolismo , Pulmão/patologia , Microvasos/metabolismo , Microvasos/patologia
12.
Biochim Biophys Acta ; 1863(8): 2124-34, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27155084

RESUMO

The molecular basis involving adsorption of pulmonary surfactant at the respiratory air-liquid interface and the specific roles of the surfactant proteins SP-B and SP-C in this process have not been completely resolved. The reasons might be found in the largely unknown structural assembly in which surfactant lipids and proteins are released from alveolar type II cells, and the difficulties to sample, manipulate and visualize the adsorption of these micron-sized particles at an air-liquid interface under appropriate physiological conditions. Here, we introduce several approaches to overcome these problems. First, by immunofluorescence we could demonstrate the presence of SP-B and SP-C on the surface of exocytosed surfactant particles. Second, by sampling the released particles and probing their adsorptive capacity we could demonstrate a remarkably high rate of interfacial adsorption, whose rate and extent was dramatically affected by treatment with antibodies against SP-B and SP-C. The effect of both antibodies was additive and specific. Third, direct microscopy of an inverted air-liquid interface revealed that the blocking effect is due to a stabilization of the released particles when contacting the air-liquid interface, precluding their transformation and the formation of surface films. We conclude that SP-B and SP-C are acting as essential, preformed molecular keys in the initial stages of surfactant unpacking and surface film formation. We further propose that surfactant activation might be transduced by a conformational change of the surfactant proteins upon contact with surface forces acting on the air-liquid interface.


Assuntos
Células Epiteliais Alveolares/metabolismo , Proteína B Associada a Surfactante Pulmonar/fisiologia , Proteína C Associada a Surfactante Pulmonar/fisiologia , Adsorção , Células Epiteliais Alveolares/efeitos dos fármacos , Células Epiteliais Alveolares/ultraestrutura , Animais , Compostos de Boro , Células Cultivadas , Exocitose , Corantes Fluorescentes , Compostos Heterocíclicos com 3 Anéis , Interações Hidrofóbicas e Hidrofílicas , Microscopia Confocal , Organelas/efeitos dos fármacos , Organelas/metabolismo , Proteína B Associada a Surfactante Pulmonar/antagonistas & inibidores , Proteína B Associada a Surfactante Pulmonar/farmacologia , Proteína C Associada a Surfactante Pulmonar/antagonistas & inibidores , Proteína C Associada a Surfactante Pulmonar/farmacologia , Surfactantes Pulmonares/química , Ratos , Ratos Sprague-Dawley , Propriedades de Superfície , Tensão Superficial
13.
Cell Calcium ; 58(6): 628-37, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26560688

RESUMO

Ambroxol (Ax) is a frequently prescribed drug used to facilitate mucociliary clearance, but its mode of action is yet poorly understood. Here we show by X-ray spectroscopy that Ax accumulates in lamellar bodies (LBs), the surfactant storing, secretory lysosomes of type II pneumocytes. Using lyso- and acidotropic substances in combination with fluorescence imaging we confirm that these vesicles belong to the class of acidic Ca(2+) stores. Ax lead to a significant neutralization of LB pH, followed by intracellular Ca(2+) release, and to a dose-dependent surfactant exocytosis. Ax-induced Ca(2+) release was significantly reduced and slowed down by pretreatment of the cells with bafilomycin A1 (Baf A1), an inhibitor of the vesicular H(+) ATPase. These results could be nearly reproduced with NH3/NH4(+). The findings suggest that Ax accumulates within LBs and severely affects their H(+) and Ca(2+) homeostasis. This is further supported by an Ax-induced change of nanostructural assembly of surfactant layers. We conclude that Ax profoundly affects LBs presumably by disordering lipid bilayers and by acting as a weak base. The pH change triggers - at least in part - Ca(2+) release from stores and secretion of surfactant from type II cells. This novel mechanism of Ax as a lysosomal secretagogue may also play a role for its recently discussed use for lysosomal storage and other degenerative diseases.


Assuntos
Ambroxol/farmacologia , Cálcio/metabolismo , Exocitose/efeitos dos fármacos , Lisossomos/efeitos dos fármacos , Vesículas Secretórias/efeitos dos fármacos , Células Epiteliais Alveolares , Animais , Separação Celular/métodos , Exocitose/fisiologia , Concentração de Íons de Hidrogênio , Ratos Sprague-Dawley
14.
Biochim Biophys Acta ; 1838(7): 1842-50, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24582711

RESUMO

Pulmonary surfactant (PS) is a surface active complex of lipids and proteins that prevents the alveolar structures from collapsing and reduces the work of breathing by lowering the surface tension at the alveolar air-liquid interface (ALI). Surfactant is synthesized by the alveolar type II (AT II) cells, and it is stored in specialized organelles, the lamellar bodies (LBs), as tightly packed lipid bilayers. Upon secretion into the alveolar lining fluid, a large fraction of these particles retain most of their packed lamellar structure, giving rise to the term lamellar body like-particles (LBPs). Due to their stability in aqueous media, freshly secreted LBPs can be harvested from AT II cell preparations. However, when LBPs get in contact with an ALI, they quickly and spontaneously adsorb into a highly organized surface film. In the present study we investigated the adsorptive capacity of LBPs at an ALI under relevant physiological parameters that characterize the alveolar environment in homeostatic or in pathological conditions. Adsorption of LBPs at an ALI is highly sensitive to pH, temperature and albumin concentration and to a relatively lesser extent to changes in osmolarity or Ca(2+) concentrations in the physiological range. Furthermore, proteolysis of LBPs significantly decreases their adsorptive capacity confirming the important role of surfactant proteins in the formation of surface active films.


Assuntos
Alvéolos Pulmonares/metabolismo , Surfactantes Pulmonares/metabolismo , Adsorção , Animais , Cálcio/metabolismo , Concentração de Íons de Hidrogênio , Bicamadas Lipídicas/metabolismo , Pressão Osmótica , Proteínas/metabolismo , Ratos
15.
PLoS One ; 9(1): e84926, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24465451

RESUMO

Leucine-rich repeat kinase 2 (LRRK2) is known to play a role in the pathogenesis of various diseases including Parkinson disease, morbus Crohn, leprosy and cancer. LRRK2 is suggested to be involved in a number of cell biological processes such as vesicular trafficking, transcription, autophagy and lysosomal pathways. Recent histological studies of lungs of LRRK2 knock-out (LRRK2 -/-) mice revealed significantly enlarged lamellar bodies (LBs) in alveolar type II (ATII) epithelial cells. LBs are large, lysosome-related storage organelles for pulmonary surfactant, which is released into the alveolar lumen upon LB exocytosis. In this study we used high-resolution, subcellular live-cell imaging assays to investigate whether similar morphological changes can be observed in primary ATII cells from LRRK2 -/- rats and whether such changes result in altered LB exocytosis. Similarly to the report in mice, ATII cells from LRRK2 -/- rats contained significantly enlarged LBs resulting in a >50% increase in LB volume. Stimulation of ATII cells with ATP elicited LB exocytosis in a significantly increased proportion of cells from LRRK2 -/- animals. LRRK2 -/- cells also displayed increased intracellular Ca(2+) release upon ATP treatment and significant triggering of LB exocytosis. These findings are in line with the strong Ca(2+)-dependence of LB fusion activity and suggest that LRRK2 -/- affects exocytic response in ATII cells via modulating intracellular Ca(2+) signaling. Post-fusion regulation of surfactant secretion was unaltered. Actin coating of fused vesicles and subsequent vesicle compression to promote surfactant expulsion were comparable in cells from LRRK2 -/- and wt animals. Surprisingly, surfactant (phospholipid) release from LRRK2 -/- cells was reduced following stimulation of LB exocytosis possibly due to impaired LB maturation and surfactant loading of LBs. In summary our results suggest that LRRK2 -/- affects LB size, modulates intracellular Ca(2+) signaling and promotes LB exocytosis upon stimulation of ATII cells with ATP.


Assuntos
Sinalização do Cálcio , Exocitose/genética , Organelas/metabolismo , Proteínas Serina-Treonina Quinases/deficiência , Alvéolos Pulmonares/metabolismo , Surfactantes Pulmonares/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Biomarcadores/metabolismo , Cálcio/metabolismo , Exocitose/efeitos dos fármacos , Deleção de Genes , Expressão Gênica , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Organelas/efeitos dos fármacos , Organelas/ultraestrutura , Proteínas Serina-Treonina Quinases/genética , Alvéolos Pulmonares/patologia , Ratos , Vesículas Secretórias/efeitos dos fármacos , Vesículas Secretórias/metabolismo , Vesículas Secretórias/ultraestrutura
16.
Nanotoxicology ; 8(8): 813-23, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23914786

RESUMO

The outstanding properties of gold nanoparticles (NPs) make them very attractive for biomedical applications. In particular, the inhalation route has gained considerable interest as an innovative strategy for diagnosis and treatment of pulmonary diseases. It is, therefore, important to scrutinise the potentially deleterious or side effects of NPs on lung epithelium. The present study investigates, for the first time, the impact of polyethylene glycol (PEG)-coated NPs on freshly purified primary cultures of rat alveolar type II (ATII) cells. These cells play a central role in the respiratory function of the lungs. They are responsible for synthesizing and secreting pulmonary surfactant (PS), which is required to stabilise the respiratory surface during breathing dynamics. Cytotoxicity and cellular uptake of NPs was evaluated by analysing morphology, viability and exocytotic activity of ATII cells (PS secretion). The impact of ATII cells' exposure to NPs was studied in a wide range of gold concentration with particles sizes of 15 and 100 nm. The results show that PEG-coated NPs are very modestly internalised by ATII cells and it neither leads to detectable morphological changes nor to decreased cell viability nor to alterations in basic functional parameters such as PS secretion, even on exposure to high gold concentration (~0.2 mM) during relatively long periods of time (24-48 h).


Assuntos
Exocitose/efeitos dos fármacos , Ouro/toxicidade , Nanopartículas Metálicas/toxicidade , Polietilenoglicóis/toxicidade , Alvéolos Pulmonares/citologia , Animais , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Corantes Fluorescentes/análise , Corantes Fluorescentes/metabolismo , Ouro/química , Ouro/farmacocinética , Espaço Intracelular/química , Espaço Intracelular/metabolismo , Macrófagos Alveolares/efeitos dos fármacos , Masculino , Nanopartículas Metálicas/química , Polietilenoglicóis/química , Surfactantes Pulmonares , Ratos , Ratos Sprague-Dawley
17.
FASEB J ; 27(4): 1772-83, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23307836

RESUMO

Two fundamental mechanisms within alveoli are essential for lung function: regulated fluid transport and secretion of surfactant. Surfactant is secreted via exocytosis of lamellar bodies (LBs) in alveolar type II (ATII) cells. We recently reported that LB exocytosis results in fusion-activated cation entry (FACE) via P2X4 receptors on LBs. We propose that FACE, in addition to facilitating surfactant secretion, modulates alveolar fluid transport. Correlative fluorescence and atomic force microscopy revealed that FACE-dependent water influx correlated with individual fusion events in rat primary ATII cells. Moreover, ATII cell monolayers grown at air-liquid interface exhibited increases in short-circuit current (Isc) on stimulation with ATP or UTP. Both are potent agonists for LB exocytosis, but only ATP activates FACE. ATP, not UTP, elicited additional fusion-dependent increases in Isc. Overexpressing dominant-negative P2X4 abrogated this effect by ∼50%, whereas potentiating P2X4 lead to ∼80% increase in Isc. Finally, we monitored changes in alveolar surface liquid (ASL) on ATII monolayers by confocal microscopy. Only stimulation with ATP, not UTP, led to a significant, fusion-dependent, 20% decrease in ASL, indicating apical-to-basolateral fluid transport across ATII monolayers. Our data support the first direct link between LB exocytosis, regulation of surfactant secretion, and transalveolar fluid resorption via FACE.


Assuntos
Trifosfato de Adenosina/farmacologia , Fusão de Membrana/efeitos dos fármacos , Alvéolos Pulmonares/metabolismo , Surfactantes Pulmonares/metabolismo , Receptores Purinérgicos P2X4/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Cátions/metabolismo , Exocitose/fisiologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Microscopia de Força Atômica/métodos , Alvéolos Pulmonares/citologia , Ratos , Ratos Sprague-Dawley , Uridina Trifosfato/farmacologia
18.
Am J Physiol Lung Cell Mol Physiol ; 303(2): L117-29, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22610352

RESUMO

Previous work from our group (Ravasio A, Hobi N, Bertocchi C, Jesacher A, Dietl P, Haller T. Am J Physiol Cell Physiol 300: C1456-C1465, 2011.) showed that contact of alveolar epithelial type II cells with an air-liquid interface (I(AL)) leads to a paradoxical situation. It is a potential threat that can cause cell injury, but also a Ca(2+)-dependent stimulus for surfactant secretion. Both events can be explained by the impact of interfacial tensile forces on cellular structures. Here, the strength of this mechanical stimulus became also apparent in microarray studies by a rapid and significant change on the transcriptional level. Cells challenged with an I(AL) in two different ways showed activation/inactivation of cellular pathways involved in stress response and defense, and a detailed Pubmatrix search identified genes associated with several lung diseases and injuries. Altogether, they suggest a close relationship of interfacial stress sensation with current models in alveolar micromechanics. Further similarities between I(AL) and cell stretch were found with respect to the underlying signaling events. The source of Ca(2+) was extracellular, and the transmembrane Ca(2+) entry pathway suggests the involvement of a mechanosensitive channel. We conclude that alveolar type II cells, due to their location and morphology, are specific sensors of the I(AL), but largely protected from interfacial stress by surfactant release.


Assuntos
Células Epiteliais Alveolares/metabolismo , Sinalização do Cálcio , Expressão Gênica , Estresse Fisiológico , Células Epiteliais Alveolares/enzimologia , Animais , Produtos Biológicos/farmacologia , Células Cultivadas , Análise por Conglomerados , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , L-Lactato Desidrogenase/metabolismo , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Fosfolipídeos/farmacologia , Alvéolos Pulmonares/citologia , Surfactantes Pulmonares/metabolismo , Surfactantes Pulmonares/farmacologia , Ratos , Ratos Sprague-Dawley , Análise de Célula Única , Tensão Superficial
19.
J Cell Sci ; 125(Pt 11): 2765-74, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22427691

RESUMO

Secretion of vesicular contents by exocytosis is a fundamental cellular process. Increasing evidence suggests that post-fusion events play an important role in determining the composition and quantity of the secretory output. In particular, regulation of fusion pore dilation and closure is considered a key regulator of the post-fusion phase. However, depending on the nature of the cargo, additional mechanisms might be essential to facilitate effective release. We have recently described that in alveolar type II (ATII) cells, lamellar bodies (LBs), which are secretory vesicles that store lung surfactant, are coated with actin following fusion with the plasma membrane. Surfactant, a lipoprotein complex, does not readily diffuse out of fused LBs following opening and dilation of the fusion pore. Using fluorescence microscopy, atomic force microscopy and biochemical assays, we present evidence that actin coating and subsequent contraction of the actin coat is essential to facilitate surfactant secretion. Latrunculin B prevents actin coating of fused LBs and inhibits surfactant secretion almost completely. Simultaneous imaging of the vesicle membrane and the actin coat revealed that contraction of the actin coat compresses the vesicle following fusion. This leads to active extrusion of vesicle contents. Initial actin coating of fused vesicles is dependent on activation of Rho and formin-dependent actin nucleation. Actin coat contraction is facilitated by myosin II. In summary, our data suggest that fusion pore opening and dilation itself is not sufficient for release of bulky vesicle cargos and that active extrusion mechanisms are required.


Assuntos
Actinas/metabolismo , Proteínas Fetais/metabolismo , Fusão de Membrana , Proteínas dos Microfilamentos/metabolismo , Miosina Tipo II/metabolismo , Proteínas Nucleares/metabolismo , Surfactantes Pulmonares/metabolismo , Vesículas Secretórias/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Depsipeptídeos/farmacologia , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Recuperação de Fluorescência Após Fotodegradação , Forminas , Fusão de Membrana/efeitos dos fármacos , Microscopia de Força Atômica , Ratos , Ratos Sprague-Dawley , Vesículas Secretórias/efeitos dos fármacos , Vesículas Secretórias/ultraestrutura , Tiazolidinas/farmacologia
20.
Am J Physiol Cell Physiol ; 300(6): C1456-65, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21270294

RESUMO

Alveolar type II (AT II) cells are in close contact with an air-liquid interface (I(AL)). This contact may be of considerable physiological relevance; however, no data exist to provide a satisfying description of this specific microenvironment. This is mainly due to the experimental difficulty to manipulate and analyze cell-air contacts in a specific way. Therefore, we designed assays to quantify cell viability, Ca(2+) changes, and exocytosis in the course of interface contact and miniaturized I(AL) devices for direct, subcellular, and real-time analyses of cell-interface interactions by fluorescence microscopy or interferometry. The studies demonstrated that the sole presence of an I(AL) is not sensed by the cells. However, when AT II cells are forced into closer contact with it, they respond promptly with sustained Ca(2+) signals and surfactant exocytosis before the occurrence of irreversible cell damage. This points to a paradoxical situation: a potential threat and potent stimulus for the cells. Furthermore, we found that the signalling mechanism underlying sensation of an I(AL) can be sufficiently explained by mechanical forces. These results demonstrate that the I(AL) itself can play a major, although so-far neglected, role in lung physiology, particularly in the regulatory mechanisms related with surfactant homeostasis. Moreover, they also support a general new concept of mechanosensation in the lung.


Assuntos
Células Epiteliais Alveolares/metabolismo , Pulmão/anatomia & histologia , Pulmão/fisiologia , Alvéolos Pulmonares/citologia , Ar , Células Epiteliais Alveolares/citologia , Animais , Células Cultivadas , Masculino , Microscopia/instrumentação , Microscopia/métodos , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA