Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Cell ; 186(23): 5135-5150.e28, 2023 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-37865090

RESUMEN

Mycobacterium tuberculosis (Mtb) cultured axenically without detergent forms biofilm-like cords, a clinical identifier of virulence. In lung-on-chip (LoC) and mouse models, cords in alveolar cells contribute to suppression of innate immune signaling via nuclear compression. Thereafter, extracellular cords cause contact-dependent phagocyte death but grow intercellularly between epithelial cells. The absence of these mechanopathological mechanisms explains the greater proportion of alveolar lesions with increased immune infiltration and dissemination defects in cording-deficient Mtb infections. Compression of Mtb lipid monolayers induces a phase transition that enables mechanical energy storage. Agent-based simulations demonstrate that the increased energy storage capacity is sufficient for the formation of cords that maintain structural integrity despite mechanical perturbation. Bacteria in cords remain translationally active despite antibiotic exposure and regrow rapidly upon cessation of treatment. This study provides a conceptual framework for the biophysics and function in tuberculosis infection and therapy of cord architectures independent of mechanisms ascribed to single bacteria.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Animales , Ratones , Biopelículas , Pulmón/microbiología , Pulmón/patología , Mycobacterium tuberculosis/fisiología , Tuberculosis/microbiología , Tuberculosis/patología , Virulencia , Fenómenos Biomecánicos
2.
Biomed Microdevices ; 25(4): 40, 2023 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-37851124

RESUMEN

During respiratory infection, barrier dysfunction in alveolar tissue can result from "cytokine storm" caused by overly reactive immune response. Particularly, interleukin 6 (IL-6) is implicated as a key biomarker of cytokine storm responsible for and further progression to pulmonary edema. In this study, alveolar-like tissue was reconstructed in a microfluidic device with: (1) human microvascular lung endothelial cells (HULEC-5a) cultured under flow-induced shear stress and (2) human epithelial cells (Calu-3) cultured at air-liquid interface. The effects of IL-6 and the soluble form of its receptor (sIL-6R) on the permeability, electrical resistance, and morphology of the endothelial and epithelial layers were evaluated. The diffusion barrier properties of both the endothelial and epithelial layers were significantly degraded only when IL-6 treatment was combined with sIL-6R. As suggested by recent review and clinical studies, our results provide unequivocal evidence that the barrier dysfunction occurs through trans-signaling in which IL-6 and sIL-6R form a complex and then bind to the surface of endothelial and epithelial cells, but not by classical signaling in which IL-6 binds to membrane-expressed IL-6 receptor. This finding suggests that the role of both IL-6 and sIL-6R should be considered as important biomarkers in developing strategies for treating cytokine storm.


Asunto(s)
Células Endoteliales , Interleucina-6 , Humanos , Receptor gp130 de Citocinas/metabolismo , Síndrome de Liberación de Citoquinas , Células Endoteliales/metabolismo , Células Epiteliales , Interleucina-6/metabolismo
3.
Environ Sci Technol ; 57(48): 19223-19235, 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-37933439

RESUMEN

Insights into how biological systems respond to high- and low-dose acute environmental stressors are a fundamental aspect of exposome research. However, studying the impact of low-level environmental exposure in conventional in vitro settings is challenging. This study employed a three-dimensional (3D) biomimetic microfluidic lung-on-chip (µLOC) platform and RNA-sequencing to examine the effects of two model anthropogenic engineered nanoparticles (NPs): zinc oxide nanoparticles (Nano-ZnO) and copier center nanoparticles (Nano-CCP). The airway epithelium exposed to these NPs exhibited dose-dependent increases in cytotoxicity and barrier dysregulation (dominance of the external exposome). Interestingly, even nontoxic and low-level exposure (10 µg/mL) of the epithelium compartment to Nano-ZnO triggered chemotaxis of lung fibroblasts toward the epithelium. An increase in α smooth muscle actin (α-SMA) expression and contractile activity was also observed in these cells, indicating a bystander-like adaptive response (dominance of internal exposome). Further bioinformatics and network analysis showed that a low-dose Nano-ZnO significantly induced a robust transcriptomic response and upregulated several hub genes associated with the development of lung fibrosis. We propose that Nano-ZnO, even at a no observable effect level (NOEL) dose according to conventional standards, can function as a potent nanostressor to disrupt airway epithelium homeostasis. This leads to a cascade of profibrotic events in a cross-tissue compartment fashion. Our findings offer new insights into the early acute events of respiratory harm associated with environmental NPs exposure, paving the way for better exposomic understanding of this emerging class of anthropogenic nanopollutants.


Asunto(s)
Exposoma , Nanopartículas , Óxido de Zinc , Biomimética , Microfluídica , Nanopartículas/toxicidad , Fibroblastos , Óxido de Zinc/toxicidad
4.
J Appl Toxicol ; 39(11): 1484-1491, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31025406

RESUMEN

Exposure to atmospheric particulate matter (PM) can affect human health, causing asthma, atherosclerosis, renal disease and cancer. In the last few years, outdoor air pollution has increased globally, leading to a public health emergency. Epidemiological studies have reported a correlation between the development of severe respiratory and systemic diseases and exposure to PM. To evaluate the toxic effect of PM of different origins, conventional experimental toxicological investigations have been conducted in animals; however, animal experimentation poses major ethical issues and usually differs from human conditions. As an alternative, human cell cultures are increasingly being used to investigate cellular and molecular mechanisms of PM toxicity. Although 2D cell cultures have been proven helpful, they are far from being a valid alternative to animal tests. Recently, 3D cell culture and organ-on-chip technology have provided systems that are more complex and that can be more informative for toxicity studies. In this review, the results of the 2D systems that are most frequently used for PM toxicity evaluations are summarized with a special focus on their limitations. We also examined to which extent 3D cell culture and particularly the organ-on-chip technology may overcome these limitations and represent effective tools to improve airborne PM toxicity evaluations.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Alternativas a las Pruebas en Animales/métodos , Material Particulado/toxicidad , Pruebas de Toxicidad/métodos , Técnicas de Cultivo de Célula/métodos , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/ultraestructura , Humanos , Sistema Respiratorio/citología , Sistema Respiratorio/efectos de los fármacos
5.
Tissue Eng Part B Rev ; 30(1): 82-96, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37597193

RESUMEN

Respiratory infections caused by coronaviruses (CoVs) have become a major public health concern in the past two decades as revealed by the emergence of SARS-CoV in 2002, MERS-CoV in 2012, and SARS-CoV-2 in 2019. The most severe clinical phenotypes commonly arise from exacerbation of immune response following the infection of alveolar epithelial cells localized at the pulmonary blood-air barrier. Preclinical rodent models do not adequately represent the essential genetic properties of the barrier, thus necessitating the use of humanized transgenic models. However, existing monolayer cell culture models have so far been unable to mimic the complex lung microenvironment. In this respect, air-liquid interface models, tissue engineered models, and organ-on-a-chip systems, which aim to better imitate the infection site microenvironment and microphysiology, are being developed to replace the commonly used monolayer cell culture models, and their use is becoming more widespread every day. On the contrary, studies on the development of nanoparticles (NPs) that mimic respiratory viruses, and those NPs used in therapy are progressing rapidly. The first part of this review describes in vitro models that mimic the blood-air barrier, the tissue interface that plays a central role in COVID-19 progression. In the second part of the review, NPs mimicking the virus and/or designed to carry therapeutic agents are explained and exemplified.


Asunto(s)
COVID-19 , Coronavirus del Síndrome Respiratorio de Oriente Medio , Nanopartículas , Humanos , SARS-CoV-2 , Barrera Alveolocapilar
6.
Adv Healthc Mater ; 13(21): e2400357, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38695274

RESUMEN

Multicellular organisms exhibit synergistic effects among their components, giving rise to emergent properties crucial for their genesis and overall functionality and survival. Morphogenesis involves and relies upon intricate and biunivocal interactions among cells and their environment, that is, the extracellular matrix (ECM). Cells secrete their own ECM, which in turn, regulates their morphogenetic program by controlling time and space presentation of matricellular signals. The ECM, once considered passive, is now recognized as an informative space where both biochemical and biophysical signals are tightly orchestrated. Replicating this sophisticated and highly interconnected informative media in a synthetic scaffold for tissue engineering is unattainable with current technology and this limits the capability to engineer functional human organs in vitro and in vivo. This review explores current limitations to in vitro organ morphogenesis, emphasizing the interplay of gene regulatory networks, mechanical factors, and tissue microenvironment cues. In vitro efforts to replicate biological processes for barrier organs such as the lung and intestine, are examined. The importance of maintaining cells within their native microenvironmental context is highlighted to accurately replicate organ-specific properties. The review underscores the necessity for microphysiological systems that faithfully reproduce cell-native interactions, for advancing the understanding of developmental disorders and disease progression.


Asunto(s)
Matriz Extracelular , Ingeniería de Tejidos , Humanos , Ingeniería de Tejidos/métodos , Matriz Extracelular/metabolismo , Animales , Microambiente Celular/fisiología , Andamios del Tejido/química , Pulmón/citología , Pulmón/metabolismo , Pulmón/fisiología
7.
Discov Med ; 35(177): 539-552, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37553308

RESUMEN

BACKGROUND: Influenza is an important respiratory tract pathogen that causes substantial seasonal and pandemic morbidity and mortality. The aim of this study was to systematically analyze the transcriptome characteristics of peripheral blood mononuclear cells (PBMCs) after influenza A virus infection by constructing a human lung microarray model composed of PBMCs to simulate the influenza A virus infection process. METHODS: A human lung microarray model was constructed using alveolar epithelial cells, vascular endothelial cells, alveolar macrophages and PBMCs, for simulation of the process of influenza A virus infection. The transcriptome characteristics of PBMCs after influenza A virus infection were analyzed by a single-cell RNA sequencing system. RESULTS: The study could realistically mimic the structure and physiological functions of the alveoli in vitro using immunofluorescence staining and expression of the specific marker. After the influenza A virus infected the upper lung chip channels, the epithelial cells underwent a high inflammatory response and spread to endothelial cells. Under experimental conditions, the Influenza A virus infection did not compromise the integrity of epithelial cells, but caused damage to endothelial cells and barrier dysfunction. Single-cell RNA sequencing of PBMCs showed that B and cluster of differentiation 4 (CD4) T cells played important immunomodulatory roles in response to influenza A virus infection, including significantly activating type I interferon signaling pathway, regulating cytokine and chemokine signaling pathway. Especially genes involved in cellular communication were significantly highly expressed post-infection. CONCLUSIONS: All these results suggested that the interactions among immune cells played a crucial role in endothelial cell injury and immune cell recruitment after influenza virus infection. This lung-on-chip infection model combined with single-cell RNA sequencing provided a unique platform that can closely investigate the lung immune response to influenza A virus infection and new therapeutic strategies for influenza.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Lesión Pulmonar , Gripe Humana/complicaciones , Gripe Humana/inmunología , Lesión Pulmonar/etiología , Lesión Pulmonar/inmunología , Técnicas Biosensibles , Humanos , Células Endoteliales , Citocinas/inmunología , Análisis de la Célula Individual , Leucocitos Mononucleares/inmunología
8.
Adv Mater ; 35(13): e2210519, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36750972

RESUMEN

As post-COVID complications, chronic respiratory diseases are one of the foremost causes of mortality. The quest for a cure for this recent global challenge underlines that the lack of predictive in vitro lung models is one of the main bottlenecks in pulmonary preclinical drug development. Despite rigorous efforts to develop biomimetic in vitro lung models, the current cutting-edge models represent a compromise in numerous technological and biological aspects. Most advanced in vitro models are still in the "proof-of-concept" phase with a low clinical translation of the findings. On the other hand, advances in cellular and molecular studies are mainly based on relatively simple and unrealistic in vitro models. Herein, the current challenges and potential strategies toward not only bioinspired but truly biomimetic lung models are discussed.


Asunto(s)
Biomimética , COVID-19 , Humanos , Pulmón
9.
Front Pharmacol ; 14: 1229313, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37342594

RESUMEN

[This corrects the article DOI: 10.3389/fphar.2023.1114739.].

10.
Front Pharmacol ; 14: 1114739, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36959848

RESUMEN

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.

11.
Eur J Pharm Sci ; 187: 106485, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37270149

RESUMEN

Acute respiratory distress syndrome (ARDS) is a severe lung condition with high mortality and various causes, including lung infection. No specific treatment is currently available and more research aimed at better understanding the pathophysiology of ARDS is needed. Most lung-on-chip models that aim at mimicking the air-blood barrier are designed with a horizontal barrier through which immune cells can migrate vertically, making it challenging to visualize and investigate their migration. In addition, these models often lack a barrier of natural protein-derived extracellular matrix (ECM) suitable for live cell imaging to investigate ECM-dependent migration of immune cells as seen in ARDS. This study reports a novel inflammation-on-chip model with live cell imaging of immune cell extravasation and migration during lung inflammation. The three-channel perfusable inflammation-on-chip system mimics the lung endothelial barrier, the ECM environment and the (inflamed) lung epithelial barrier. A chemotactic gradient was established across the ECM hydrogel, leading to the migration of immune cells through the endothelial barrier. We found that immune cell extravasation depends on the presence of an endothelial barrier, on the ECM density and stiffness, and on the flow profile. In particular, bidirectional flow, broadly used in association with rocking platforms, was found to significantly delay extravasation of immune cells in contrast to unidirectional flow. Extravasation was increased in the presence of lung epithelial tissue. This model is currently used to study inflammation-induced immune cell migration but can be used to study infection-induced immune cell migration under different conditions, such as ECM composition, density and stiffness, type of infectious agents used, and the presence of organ-specific cell types.


Asunto(s)
Neumonía , Síndrome de Dificultad Respiratoria , Humanos , Pulmón/metabolismo , Inflamación/metabolismo , Movimiento Celular
12.
Adv Biol (Weinh) ; 6(2): e2101139, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34962104

RESUMEN

The substantial socioeconomic burden of lung diseases, recently highlighted by the disastrous impact of the coronavirus disease 2019 (COVID-19) pandemic, accentuates the need for interventive treatments capable of decelerating disease progression, limiting organ damage, and contributing to a functional tissue recovery. However, this is hampered by the lack of accurate human lung research models, which currently fail to reproduce the human pulmonary architecture and biochemical environment. Induced pluripotent stem cells (iPSCs) and organ-on-chip (OOC) technologies possess suitable characteristics for the generation of physiologically relevant in vitro lung models, allowing for developmental studies, disease modeling, and toxicological screening. Importantly, these platforms represent potential alternatives for animal testing, according to the 3Rs (replace, reduce, refine) principle, and hold promise for the identification and approval of new chemicals under the European REACH (registration, evaluation, authorization and restriction of chemicals) framework. As such, this review aims to summarize recent progress made in human iPSC- and OOC-based in vitro lung models. A general overview of the present applications of in vitro lung models is presented, followed by a summary of currently used protocols to generate different lung cell types from iPSCs. Lastly, recently developed iPSC-based lung models are discussed.


Asunto(s)
COVID-19 , Células Madre Pluripotentes Inducidas , Preparaciones Farmacéuticas , Animales , Humanos , Pulmón , SARS-CoV-2
13.
Micromachines (Basel) ; 12(10)2021 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-34683301

RESUMEN

For centuries, animal experiments have contributed much to our understanding of mechanisms of human disease, but their value in predicting the effectiveness of drug treatments in the clinic has remained controversial. Animal models, including genetically modified ones and experimentally induced pathologies, often do not accurately reflect disease in humans, and therefore do not predict with sufficient certainty what will happen in humans. Organ-on-chip (OOC) technology and bioengineered tissues have emerged as promising alternatives to traditional animal testing for a wide range of applications in biological defence, drug discovery and development, and precision medicine, offering a potential alternative. Recent technological breakthroughs in stem cell and organoid biology, OOC technology, and 3D bioprinting have all contributed to a tremendous progress in our ability to design, assemble and manufacture living organ biomimetic systems that more accurately reflect the structural and functional characteristics of human tissue in vitro, and enable improved predictions of human responses to drugs and environmental stimuli. Here, we provide a historical perspective on the evolution of the field of bioengineering, focusing on the most salient milestones that enabled control of internal and external cell microenvironment. We introduce the concepts of OOCs and Microphysiological systems (MPSs), review various chip designs and microfabrication methods used to construct OOCs, focusing on blood-brain barrier as an example, and discuss existing challenges and limitations. Finally, we provide an overview on emerging strategies for 3D bioprinting of MPSs and comment on the potential role of these devices in precision medicine.

14.
Front Med (Lausanne) ; 8: 644678, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34026781

RESUMEN

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.

15.
Front Bioeng Biotechnol ; 8: 581163, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33304889

RESUMEN

Microphysiological systems, also known as organs-on-chips, are microfluidic devices designed to model human physiology in vitro. Polydimethylsiloxane (PDMS) is the most widely used material for organs-on-chips due to established microfabrication methods, and properties that make it suitable for biological applications such as low cytotoxicity, optical transparency, gas permeability. However, absorption of small molecules and leaching of uncrosslinked oligomers might hinder the adoption of PDMS-based organs-on-chips for drug discovery assays. Here, we have engineered a modular, PDMS-free microphysiological system that is capable of recapitulating biologic barrier functions commonly demonstrated in PDMS-based devices. Our microphysiological system is comprised of a microfluidic chip to house cell cultures and pneumatic microfluidic pumps to drive flow with programmable pressure and shear stress. The modular architecture and programmable pumps enabled us to model multiple in vivo microenvironments. First, we demonstrate the ability to generate cyclic strain on the culture membrane and establish a model of the alveolar air-liquid interface. Next, we utilized three-dimensional finite element analysis modeling to characterize the fluid dynamics within the device and develop a model of the pressure-driven filtration that occurs at the glomerular filtration barrier. Finally, we demonstrate that our model can be used to recapitulate sphingolipid induced kidney injury. Together, our results demonstrate that a multifunctional and modular microphysiological system can be deployed without the use of PDMS. Further, the bio-inert plastic used in our microfluidic device is amenable to various established, high-throughput manufacturing techniques, such as injection molding. As a result, the development plastic organs-on-chips provides an avenue to meet the increasing demand for organ-on-chip technology.

16.
Front Bioeng Biotechnol ; 8: 581995, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33195144

RESUMEN

For the past 50 years, the route of inhalation has been utilized to administer therapies to treat a variety of respiratory and pulmonary diseases. When compared with other drug administration routes, inhalation offers a targeted, non-invasive approach to deliver rapid onset of drug action to the lung, minimizing systemic drug exposure and subsequent side effects. However, despite advances in inhaled therapies, there is still a need to improve the preclinical screening and the efficacy of inhaled therapeutics. Innovative in vitro models of respiratory physiology to determine therapeutic efficacy of inhaled compounds have included the use of organoids, micro-engineered lung-on-chip systems and sophisticated bench-top platforms to enable a better understanding of pulmonary mechanisms at the molecular level, rapidly progressing inhaled therapeutic candidates to the clinic. Furthermore, the integration of complementary ex vivo models, such as precision-cut lung slices (PCLS) and isolated perfused lung platforms have further advanced preclinical drug screening approaches by providing in vivo relevance. In this review, we address the challenges and advances of in vitro models and discuss the implementation of ex vivo inhaled drug screening models. Specifically, we address the importance of understanding human in vivo pulmonary mechanisms in assessing strategies of the preclinical screening of drug efficacy, toxicity and delivery of inhaled therapeutics.

17.
Trans Indian Natl Acad Eng ; 5(2): 241-250, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-38624434

RESUMEN

Originating in China during December 2019, the novel corona-virus, SARS-CoV-2, has created mayhem worldwide in a very short time. The outbreak has been so rapid and widespread that the only option to treat the patients was administering drugs already available in the market like chloroquine/hydroxychloroquine (an antimalarial drug) and remedesivir. A large number of patients have been cured but the attribution to survival by these drugs has been controversial. Till date, we do not have any specific drug or vaccine available for COVID-19 and the pandemic seems to be far from over. To handle the current challenges posed by the outbreak effectively, we need to employ innovative interdisciplinary approaches. Organ-on-chip (OOC), particularly lung-on-chip, is one such approach which combines the potential of microfluidics, cell culture and molecular biology into a single miniaturised platform. The device is realized to be capable of simulating in-vivo physiological responses of an organ. In the current study, an OOC, which is a multichannel 3D cell culture microfluidic device, is made via soft lithography technique, using polydimethylsiloxane-polymer and diverse polymeric porous/semipermeable membranes. Several polymer membranes i.e. PDMS, polyvinylidene fluoride (PVDF), nitrocellulose, polyester etc., integrated into the microdevices, were efficiently explored to realize their better cell-adhesion and viability property. We also propose for the application of a simple, smart and cost-effective lung-on-chip platform to study the SARS-CoV-2 pathogenesis in humans, drug toxicity testing and provide insights into antigen-antibody interactions. This platform will enable us to study multiple phenomena at a micro-level generating more reliable data and a better understanding of the underlying mechanisms of SARS-CoV-2 infection and pathogenesis.

18.
Adv Drug Deliv Rev ; 129: 78-94, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29269274

RESUMEN

Pulmonary fibrosis includes several lung disorders characterized by scar formation and Idiopathic Pulmonary Fibrosis (IPF) is a particularly severe form of pulmonary fibrosis of unknown etiology with a mean life expectancy of 3years' post-diagnosis. Treatments for IPF are limited to two FDA approved drugs, pirfenidone and nintedanib. Most lead candidate drugs that are identified in pre-clinical animal studies fail in human clinical trials. Thus, there is a need for advanced humanized in vitro models of the lung to improve candidate treatments prior to moving to human clinical trials. The development of 3D tissue models has created systems capable of emulating human lung structure, function, and cell and matrix interactions. The specific models accomplish these features and preliminary studies conducted using some of these systems have shown potential for in vitro anti-fibrotic drug testing. Further characterization and improvements will enable these tissue models to extend their utility for in vitro drug testing, to help identify signaling pathways and mechanisms for new drug targets, and potentially reduce animal models as standard pre-clinical models of study. In the current review, we contrast different in vitro models based on increasing dimensionality (2D, 2.5D and 3D), with added focus on contemporary 3D pulmonary models of fibrosis.


Asunto(s)
Ingeniería Celular , Modelos Biológicos , Fibrosis Pulmonar/patología , Animales , Humanos , Fibrosis Pulmonar/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA