Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Adv Drug Deliv Rev ; 140: 12-32, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30009883

RESUMO

Pathologies of the respiratory system such as lung infections, chronic inflammatory lung diseases, and lung cancer are among the leading causes of morbidity and mortality, killing one in six people worldwide. Development of more effective treatments is hindered by the lack of preclinical models of the human lung that can capture the disease complexity, highly heterogeneous disease phenotypes, and pharmacokinetics and pharmacodynamics observed in patients. The merger of two novel technologies, Organs-on-Chips and human stem cell engineering, has the potential to deliver such urgently needed models. Organs-on-Chips, which are microengineered bioinspired tissue systems, recapitulate the mechanochemical environment and physiological functions of human organs while concurrent advances in generating and differentiating human stem cells promise a renewable supply of patient-specific cells for personalized and precision medicine. Here, we discuss the challenges of modeling human lung pathophysiology in vitro, evaluate past and current models including Organs-on-Chips, review the current status of lung tissue modeling using human pluripotent stem cells, explore in depth how stem-cell based Lung-on-Chips may advance disease modeling and drug testing, and summarize practical consideration for the design of Lung-on-Chips for academic and industry applications.


Assuntos
Células-Tronco Embrionárias , Células-Tronco Pluripotentes Induzidas , Pulmão , Engenharia Tecidual/métodos , Animais , Humanos , Pulmão/fisiopatologia , Pneumopatias , Modelos Biológicos
2.
Vaccines (Basel) ; 6(3)2018 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-30060554

RESUMO

The respiratory tract harbours a variety of microorganisms, collectively called the respiratory microbiota. Over the past few years, alterations in respiratory and gut microbiota composition have been associated with chronic inflammatory diseases of the lungs. How these changes influence disease development and progression is an active field of investigation. Identifying and understanding host-microbiota interactions and factors contributing to these interactions could promote the development of novel therapeutic strategies aimed at restoring host-microbiota homeostasis. In this review, we discuss recent literature on host-microbiota interactions in the respiratory tract, with a specific focus on the influence of endogenous host defence peptides and proteins (HDPs) on the composition of microbiota populations in vivo and explore possible HDPs-related therapeutic approaches targeting microbiota dysbiosis in chronic inflammatory lung diseases.

3.
Lab Chip ; 17(13): 2264-2271, 2017 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-28598479

RESUMO

Trans-epithelial electrical resistance (TEER) is broadly used as an experimental readout and a quality control assay for measuring the integrity of epithelial monolayers cultured under static conditions in vitro, however, there is no standard methodology for its application to microfluidic organ-on-a-chip (organ chip) cultures. Here, we describe a new microfluidic organ chip design that contains embedded electrodes, and we demonstrate its utility for assessing formation and disruption of barrier function both within a human lung airway chip lined by a fully differentiated mucociliary human airway epithelium and in a human gut chip lined by intestinal epithelial cells. These chips with integrated electrodes enable real-time, non-invasive monitoring of TEER and can be applied to measure barrier function in virtually any type of cultured cell.


Assuntos
Impedância Elétrica , Células Epiteliais , Dispositivos Lab-On-A-Chip , Modelos Biológicos , Técnicas de Cultura de Órgãos/instrumentação , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Epitélio/fisiologia , Desenho de Equipamento , Humanos
4.
Methods Mol Biol ; 1442: 119-39, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27464691

RESUMO

The choice of model used to study human respiratory syncytial virus (RSV) infection is extremely important. RSV is a human pathogen that is exquisitely adapted to infection of human hosts. Rodent models, such as mice and cotton rats, are semi-permissive to RSV infection and do not faithfully reproduce hallmarks of RSV disease in humans. Furthermore, immortalized airway-derived cell lines, such as HEp-2, BEAS-2B, and A549 cells, are poorly representative of the complexity of the respiratory epithelium. The development of a well-differentiated primary pediatric airway epithelial cell models (WD-PAECs) allows us to simulate several hallmarks of RSV infection of infant airways. They therefore represent important additions to RSV pathogenesis modeling in human-relevant tissues. The following protocols describe how to culture and differentiate both bronchial and nasal primary pediatric airway epithelial cells and how to use these cultures to study RSV cytopathogenesis.


Assuntos
Brônquios/citologia , Nariz/citologia , Vírus Sincicial Respiratório Humano/patogenicidade , Células A549 , Brônquios/virologia , Técnicas de Cultura de Células , Diferenciação Celular , Linhagem Celular , Células Epiteliais/citologia , Células Epiteliais/virologia , Humanos , Técnicas In Vitro , Lactente , Modelos Biológicos , Nariz/virologia
5.
PLoS Pathog ; 12(5): e1005622, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27152417

RESUMO

Respiratory syncytial virus (RSV) is the major cause of viral lower respiratory tract illness in children. In contrast to the RSV prototypic strain A2, clinical isolate RSV 2-20 induces airway mucin expression in mice, a clinically relevant phenotype dependent on the fusion (F) protein of the RSV strain. Epidermal growth factor receptor (EGFR) plays a role in airway mucin expression in other systems; therefore, we hypothesized that the RSV 2-20 F protein stimulates EGFR signaling. Infection of cells with chimeric strains RSV A2-2-20F and A2-2-20GF or over-expression of 2-20 F protein resulted in greater phosphorylation of EGFR than infection with RSV A2 or over-expression of A2 F, respectively. Chemical inhibition of EGFR signaling or knockdown of EGFR resulted in diminished infectivity of RSV A2-2-20F but not RSV A2. Over-expression of EGFR enhanced the fusion activity of 2-20 F protein in trans. EGFR co-immunoprecipitated most efficiently with RSV F proteins derived from "mucogenic" strains. RSV 2-20 F and EGFR co-localized in H292 cells, and A2-2-20GF-induced MUC5AC expression was ablated by EGFR inhibitors in these cells. Treatment of BALB/c mice with the EGFR inhibitor erlotinib significantly reduced the amount of RSV A2-2-20F-induced airway mucin expression. Our results demonstrate that RSV F interacts with EGFR in a strain-specific manner, EGFR is a co-factor for infection, and EGFR plays a role in RSV-induced mucin expression, suggesting EGFR is a potential target for RSV disease.


Assuntos
Receptores ErbB/metabolismo , Mucinas/biossíntese , Infecções por Vírus Respiratório Sincicial/metabolismo , Proteínas Virais de Fusão/metabolismo , Animais , Western Blotting , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Imunofluorescência , Técnicas de Silenciamento de Genes , Imunoprecipitação , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase em Tempo Real , Vírus Sincicial Respiratório Humano
6.
Annu Rev Pathol ; 10: 195-262, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25621660

RESUMO

The ultimate goal of most biomedical research is to gain greater insight into mechanisms of human disease or to develop new and improved therapies or diagnostics. Although great advances have been made in terms of developing disease models in animals, such as transgenic mice, many of these models fail to faithfully recapitulate the human condition. In addition, it is difficult to identify critical cellular and molecular contributors to disease or to vary them independently in whole-animal models. This challenge has attracted the interest of engineers, who have begun to collaborate with biologists to leverage recent advances in tissue engineering and microfabrication to develop novel in vitro models of disease. As these models are synthetic systems, specific molecular factors and individual cell types, including parenchymal cells, vascular cells, and immune cells, can be varied independently while simultaneously measuring system-level responses in real time. In this article, we provide some examples of these efforts, including engineered models of diseases of the heart, lung, intestine, liver, kidney, cartilage, skin and vascular, endocrine, musculoskeletal, and nervous systems, as well as models of infectious diseases and cancer. We also describe how engineered in vitro models can be combined with human inducible pluripotent stem cells to enable new insights into a broad variety of disease mechanisms, as well as provide a test bed for screening new therapies.


Assuntos
Modelos Biológicos , Patologia/métodos , Animais , Doença , Humanos , Técnicas In Vitro
7.
Am J Respir Crit Care Med ; 188(7): 842-51, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23952745

RESUMO

RATIONALE: Respiratory syncytial virus (RSV) is a major pathogen that primarily infects airway epithelium. Most infants suffer mild upper respiratory tract (URT) symptoms, whereas approximately one-third progress to lower respiratory tract (LRT) involvement. Despite the ubiquity of URT infection, little is known about the relative cytopathogenesis of RSV infection in infant URT and LRT. OBJECTIVES: This study aimed to compare RSV cytopathogenesis in nasal- and bronchial-derived epithelium from the same individuals using novel models derived from well-differentiated primary pediatric nasal (WD-PNECs) and bronchial epithelial cells (WD-PBECs). METHODS: WD-PNECs and WD-PBECs were generated from nasal and bronchial brushes, respectively, and mock-infected or infected with RSV BT2a. RSV tropism, infectivity, cytopathology, growth kinetics, cell sloughing, apoptosis, and cytokine and chemokine responses were determined. MEASUREMENTS AND MAIN RESULTS: RSV infection in both cultures was restricted to apical ciliated cells and occasional nonciliated cells but not goblet cells. It did not cause gross cytopathology. Infection resulted in apical release of progeny virus, increased apical cell sloughing, apoptosis, and occasional syncytia. RSV growth kinetics and peak titers were higher in WD-PBECs, coincident with higher ciliated cell contents, cell sloughing, and slightly compromised tight junctions. However, proinflammatory chemokine responses were similar for both cultures. Also, lambda IFNs, especially IL-29, were induced by RSV infection. CONCLUSIONS: RSV induced remarkably similar, albeit quantitatively lower, cytopathogenesis and proinflammatory responses in WD-PNECs compared with WD-PBECs that reproduce many hallmarks of RSV pathogenesis in infants. WD-PNECs may provide an authentic surrogate model with which to study RSV cytopathogenesis in infant airway epithelium.


Assuntos
Brônquios/virologia , Mucosa Nasal/virologia , Infecções por Vírus Respiratório Sincicial/patologia , Infecções Respiratórias/virologia , Brônquios/imunologia , Brônquios/patologia , Diferenciação Celular , Quimiocinas/imunologia , Pré-Escolar , Citocinas/imunologia , Efeito Citopatogênico Viral/imunologia , Células Epiteliais/imunologia , Células Epiteliais/patologia , Células Epiteliais/virologia , Humanos , Irlanda , Mucosa Nasal/imunologia , Mucosa Nasal/patologia , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios , Infecções Respiratórias/imunologia , Infecções Respiratórias/patologia , Replicação Viral
8.
Trends Microbiol ; 21(5): 238-44, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23523320

RESUMO

Although respiratory syncytial virus (RSV) is a major human respiratory pathogen, our knowledge of how it causes disease in humans is limited. Airway epithelial cells are the primary targets of RSV infection in vivo, so the generation and exploitation of RSV infection models based on morphologically and physiologically authentic well-differentiated primary human airway epithelial cells cultured at an air-liquid interface (WD-PAECs) provide timely developments that will help to bridge this gap. Here we review the interaction of RSV with WD-PAEC cultures, the authenticity of the RSV-WD-PAEC models relative to RSV infection of human airway epithelium in vivo, and future directions for their exploitation in our quest to understand RSV pathogenesis in humans.


Assuntos
Células Epiteliais/virologia , Interações Hospedeiro-Patógeno , Mucosa Respiratória/virologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/fisiologia , Animais , Humanos , Mucosa Respiratória/citologia , Vírus Sinciciais Respiratórios/genética
9.
Proc Natl Acad Sci U S A ; 109(13): 5040-5, 2012 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-22411804

RESUMO

Respiratory syncytial virus (RSV) is the major viral cause of severe pulmonary disease in young infants worldwide. However, the mechanisms by which RSV causes disease in humans remain poorly understood. To help bridge this gap, we developed an ex vivo/in vitro model of RSV infection based on well-differentiated primary pediatric bronchial epithelial cells (WD-PBECs), the primary targets of RSV infection in vivo. Our RSV/WD-PBEC model demonstrated remarkable similarities to hallmarks of RSV infection in infant lungs. These hallmarks included restriction of infection to noncontiguous or small clumps of apical ciliated and occasional nonciliated epithelial cells, apoptosis and sloughing of apical epithelial cells, occasional syncytium formation, goblet cell hyperplasia/metaplasia, and mucus hypersecretion. RSV was shed exclusively from the apical surface at titers consistent with those in airway aspirates from hospitalized infants. Furthermore, secretion of proinflammatory chemokines such as CXCL10, CCL5, IL-6, and CXCL8 reflected those chemokines present in airway aspirates. Interestingly, a recent RSV clinical isolate induced more cytopathogenesis than the prototypic A2 strain. Our findings indicate that this RSV/WD-PBEC model provides an authentic surrogate for RSV infection of airway epithelium in vivo. As such, this model may provide insights into RSV pathogenesis in humans that ultimately lead to successful RSV vaccines or therapeutics.


Assuntos
Brônquios/patologia , Modelos Biológicos , Mucosa Respiratória/patologia , Mucosa Respiratória/virologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/fisiologia , Apoptose , Diferenciação Celular , Quimiocinas/metabolismo , Criança , Cílios/patologia , Efeito Citopatogênico Viral , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Células Epiteliais/virologia , Células Gigantes/patologia , Células Gigantes/virologia , Células Caliciformes/patologia , Humanos , Hiperplasia , Muco/metabolismo , Infecções por Vírus Respiratório Sincicial/patologia , Vírus Sinciciais Respiratórios/patogenicidade
10.
Virol J ; 8: 43, 2011 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-21272337

RESUMO

BACKGROUND: Human respiratory syncytial virus (RSV) causes severe respiratory disease in infants. Airway epithelial cells are the principle targets of RSV infection. However, the mechanisms by which it causes disease are poorly understood. Most RSV pathogenesis data are derived using laboratory-adapted prototypic strains. We hypothesized that such strains may be poorly representative of recent clinical isolates in terms of virus/host interactions in primary human bronchial epithelial cells (PBECs). METHODS: To address this hypothesis, we isolated three RSV strains from infants hospitalized with bronchiolitis and compared them with the prototypic RSV A2 in terms of cytopathology, virus growth kinetics and chemokine secretion in infected PBEC monolayers. RESULTS: RSV A2 rapidly obliterated the PBECs, whereas the clinical isolates caused much less cytopathology. Concomitantly, RSV A2 also grew faster and to higher titers in PBECs. Furthermore, dramatically increased secretion of IP-10 and RANTES was evident following A2 infection compared with the clinical isolates. CONCLUSIONS: The prototypic RSV strain A2 is poorly representative of recent clinical isolates in terms of cytopathogenicity, viral growth kinetics and pro-inflammatory responses induced following infection of PBEC monolayers. Thus, the choice of RSV strain may have important implications for future RSV pathogenesis studies.


Assuntos
Células Epiteliais/virologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/isolamento & purificação , Vírus Sincicial Respiratório Humano/patogenicidade , Bronquiolite Viral/virologia , Quimiocinas/metabolismo , Criança , Pré-Escolar , Efeito Citopatogênico Viral , Humanos , Lactente , Mucosa Respiratória/virologia , Vírus Sincicial Respiratório Humano/crescimento & desenvolvimento , Virulência , Replicação Viral
11.
J Virol ; 84(22): 11718-28, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20810726

RESUMO

Sendai virus (SeV) is a murine respiratory virus of considerable interest as a gene therapy or vaccine vector, as it is considered nonpathogenic in humans. However, little is known about its interaction with the human respiratory tract. To address this, we developed a model of respiratory virus infection based on well-differentiated primary pediatric bronchial epithelial cells (WD-PBECs). These physiologically authentic cultures are comprised of polarized pseudostratified multilayered epithelium containing ciliated, goblet, and basal cells and intact tight junctions. To facilitate our studies, we rescued a replication-competent recombinant SeV expressing enhanced green fluorescent protein (rSeV/eGFP). rSeV/eGFP infected WD-PBECs efficiently and progressively and was restricted to ciliated and nonciliated cells, not goblet cells, on the apical surface. Considerable cytopathology was evident in the rSeV/eGFP-infected cultures postinfection. This manifested itself by ciliostasis, cell sloughing, apoptosis, and extensive degeneration of WD-PBEC cultures. Syncytia were also evident, along with significant basolateral secretion of proinflammatory chemokines, including IP-10, RANTES, tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), interleukin 6 (IL-6), and IL-8. Such deleterious responses are difficult to reconcile with a lack of pathogenesis in humans and suggest that caution may be required in exploiting replication-competent SeV as a vaccine vector. Alternatively, such robust responses might constitute appropriate normal host responses to viral infection and be a prerequisite for the induction of efficient immune responses.


Assuntos
Brônquios/citologia , Diferenciação Celular , Células Epiteliais/virologia , Infecções por Respirovirus/virologia , Vírus Sendai/fisiologia , Brônquios/imunologia , Brônquios/patologia , Brônquios/virologia , Células Cultivadas , Criança , Citocinas/imunologia , Efeito Citopatogênico Viral , Células Epiteliais/citologia , Células Epiteliais/imunologia , Células Epiteliais/patologia , Feminino , Humanos , Masculino , Infecções por Respirovirus/imunologia , Infecções por Respirovirus/patologia , Replicação Viral
12.
Pediatr Res ; 67(1): 17-22, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19755931

RESUMO

There is a need for reproducible and effective models of pediatric bronchial epithelium to study disease states such as asthma. We aimed to develop, characterize, and differentiate an effective, an efficient, and a reliable three-dimensional model of pediatric bronchial epithelium to test the hypothesis that children with asthma differ in their epithelial morphologic phenotype when compared with nonasthmatic children. Primary cell cultures from both asthmatic and nonasthmatic children were grown and differentiated at the air-liquid interface for 28 d. Tight junction formation, MUC5AC secretion, IL-8, IL-6, prostaglandin E2 production, and the percentage of goblet and ciliated cells in culture were assessed. Well-differentiated, multilayered, columnar epithelium containing both ciliated and goblet cells from asthmatic and nonasthmatic subjects were generated. All cultures demonstrated tight junction formation at the apical surface and exhibited mucus production and secretion. Asthmatic and nonasthmatic cultures secreted similar quantities of IL-8, IL-6, and prostaglandin E2. Cultures developed from asthmatic children contained considerably more goblet cells and fewer ciliated cells compared with those from nonasthmatic children. A well-differentiated model of pediatric epithelium has been developed that will be useful for more in vivo like study of the mechanisms at play during asthma.


Assuntos
Asma/patologia , Brônquios/anatomia & histologia , Modelos Biológicos , Asma/metabolismo , Brônquios/metabolismo , Brônquios/patologia , Criança , Dinoprostona/biossíntese , Epitélio/anatomia & histologia , Epitélio/metabolismo , Epitélio/patologia , Humanos , Interleucina-6/biossíntese , Interleucina-8/biossíntese , Mucina-5AC/metabolismo , Junções Íntimas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA