Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(10): e2318771121, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38416686

RESUMO

Apical cilia on epithelial cells defend the lung by propelling pathogens and particulates out of the respiratory airways. Ciliated cells produce ATP that powers cilia beating by densely grouping mitochondria just beneath the apical membrane. However, this efficient localization comes at a cost because electrons leaked during oxidative phosphorylation react with molecular oxygen to form superoxide, and thus, the cluster of mitochondria creates a hotspot for oxidant production. The relatively high oxygen concentration overlying airway epithelia further intensifies the risk of generating superoxide. Thus, airway ciliated cells face a unique challenge of producing harmful levels of oxidants. However, surprisingly, highly ciliated epithelia produce less reactive oxygen species (ROS) than epithelia with few ciliated cells. Compared to other airway cell types, ciliated cells express high levels of mitochondrial uncoupling proteins, UCP2 and UCP5. These proteins decrease mitochondrial protonmotive force and thereby reduce production of ROS. As a result, lipid peroxidation, a marker of oxidant injury, decreases. However, mitochondrial uncoupling proteins exact a price for decreasing oxidant production; they decrease the fraction of mitochondrial respiration that generates ATP. These findings indicate that ciliated cells sacrifice mitochondrial efficiency in exchange for safety from damaging oxidation. Employing uncoupling proteins to prevent oxidant production, instead of relying solely on antioxidants to decrease postproduction oxidant levels, may offer an advantage for targeting a local area of intense ROS generation.


Assuntos
Canais Iônicos , Superóxidos , Humanos , Espécies Reativas de Oxigênio/metabolismo , Proteínas de Desacoplamento Mitocondrial/metabolismo , Superóxidos/metabolismo , Canais Iônicos/metabolismo , Estresse Oxidativo , Trifosfato de Adenosina/metabolismo , Células Epiteliais/metabolismo , Oxidantes/farmacologia , Oxigênio/metabolismo , Proteínas Mitocondriais/metabolismo
2.
Dev Cell ; 57(18): 2221-2236.e5, 2022 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-36108628

RESUMO

Pulmonary neuroendocrine cells (PNECs) are rare airway cells with potential sensory capacity linked to vagal neurons and immune cells. How PNECs sense and respond to external stimuli remains poorly understood. We discovered PNECs located within pig and human submucosal glands, a tissue that produces much of the mucus that defends the lung. These PNECs sense succinate, an inflammatory molecule in liquid lining the airway surface. The results indicate that succinate migrates down the submucosal gland duct to the acinus, where it triggers apical succinate receptors, causing PNECs to release ATP. The short-range ATP signal stimulates the contraction of myoepithelial cells wrapped tightly around the submucosal glands. Succinate-triggered gland contraction may complement the action of neurotransmitters that induce mucus release but not gland contraction to promote mucus ejection onto the airway surface. These findings identify a local circuit in which rare PNECs within submucosal glands sense an environmental cue to orchestrate the function of airway glands.


Assuntos
Células Neuroendócrinas , Trifosfato de Adenosina/metabolismo , Animais , Humanos , Pulmão/metabolismo , Muco/metabolismo , Ácido Succínico/metabolismo , Suínos
3.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35046051

RESUMO

Submucosal glands (SMGs) protect lungs but can also contribute to disease. For example, in cystic fibrosis (CF), SMGs produce abnormal mucus that disrupts mucociliary transport. CF is an ion transport disease, yet knowledge of the ion transporters expressed by SMG acini, which produce mucus, and SMG ducts that carry it to the airway lumen is limited. Therefore, we isolated SMGs from newborn pigs and used single-cell messenger RNA sequencing, immunohistochemistry, and in situ hybridization to identify cell types, gene expression, and spatial distribution. Cell types and transcript levels were the same in non-CF and CF SMGs, suggesting that loss of epithelial anion secretion rather than an intrinsic cell defect causes CF mucus abnormalities. Gene signatures of acinar mucous and acinar serous cells revealed specialized functions in producing mucins and antimicrobials, respectively. However, surprisingly, these two cell types expressed the same ion transporters and neurohumoral receptors, suggesting the importance of balancing mucin and liquid secretion to produce optimal mucus properties. SMG duct cell transcripts suggest that they secrete HCO3- and Cl-, and thus have some similarity to pancreatic ducts that are also defective in CF. These and additional findings suggest the functions of the SMG acinus and duct and provide a baseline for understanding how environmental and genetic challenges impact their contribution to lung disease.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/genética , Fibrose Cística/metabolismo , Mutação , Mucosa Respiratória/metabolismo , Células Acinares/metabolismo , Animais , Biomarcadores , Fibrose Cística/etiologia , Fibrose Cística/patologia , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Modelos Animais de Doenças , Imunofluorescência , Expressão Gênica , Técnicas de Silenciamento de Genes , Predisposição Genética para Doença , Mucinas/metabolismo , Depuração Mucociliar , Muco/metabolismo , Mucosa Respiratória/patologia , Suínos
4.
Pharmacol Res Perspect ; 9(6): e00882, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34747573

RESUMO

Zinc protoporphyrin (ZnPP), a naturally occurring metalloprotoporphyrin (MPP), is currently under development as a chemotherapeutic agent although its mechanism is unclear. When tested against other MPPs, ZnPP was the most effective DNA synthesis and cellular proliferation inhibitor while promoting apoptosis in telomerase positive but not telomerase negative cells. Concurrently, ZnPP down-regulated telomerase expression and was the best overall inhibitor of telomerase activity in intact cells and cellular extracts with IC50 and EC50  values of ca 2.5 and 6 µM, respectively. The natural fluorescence properties of ZnPP enabled direct imaging in cellular fractions using non-denaturing agarose gel electrophoresis, western blots, and confocal fluorescence microscopy. ZnPP localized to large cellular complexes (>600 kD) that contained telomerase and dysskerin as confirmed with immunocomplex mobility shift, immunoprecipitation, and immunoblot analyses. Confocal fluorescence studies showed that ZnPP co-localized with telomerase reverse transcriptase (TERT) and telomeres in the nucleus of synchronized S-phase cells. ZnPP also co-localized with TERT in the perinuclear regions of log phase cells but did not co-localize with telomeres on the ends of metaphase chromosomes, a site known to be devoid of telomerase complexes. Overall, these results suggest that ZnPP does not bind to telomeric sequences per se, but alternatively, interacts with other structural components of the telomerase complex to inhibit telomerase activity. In conclusion, ZnPP actively interferes with telomerase activity in neoplastic cells, thus promoting pro-apoptotic and anti-proliferative properties. These data support further development of natural or synthetic protoporphyrins for use as chemotherapeutic agents to augment current treatment protocols for neoplastic disease.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Protoporfirinas/farmacologia , Telomerase/metabolismo , Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Inibidores Enzimáticos/administração & dosagem , Células HEK293 , Humanos , Concentração Inibidora 50 , Microscopia Confocal , Protoporfirinas/administração & dosagem , Telomerase/antagonistas & inibidores , Telômero/metabolismo
5.
Dev Cell ; 54(4): 488-500.e5, 2020 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-32730755

RESUMO

In response to respiratory insults, airway submucosal glands secrete copious mucus strands to increase mucociliary clearance and protect the lung. However, in cystic fibrosis, stimulating submucosal glands has the opposite effect, disrupting mucociliary transport. In cystic fibrosis (CF) pigs, loss of cystic fibrosis transmembrane conductance regulator (CFTR) anion channels produced submucosal gland mucus that was abnormally acidic with an increased protein concentration. To test whether these variables alter mucus, we produced a microfluidic model of submucosal glands using mucus vesicles from banana slugs. Acidic pH and increased protein concentration decreased mucus gel volume and increased mucus strand elasticity and tensile strength. However, once mucus strands were formed, changing pH or protein concentration largely failed to alter the biophysical properties. Likewise, raising pH or apical perfusion did not improve clearance of mucus strands from CF airways. These findings reveal mechanisms responsible for impaired mucociliary transport in CF and have important implications for potential treatments.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/metabolismo , Pulmão/metabolismo , Mucosa Respiratória/metabolismo , Animais , Transporte Biológico , Fibrose Cística/genética , Fibrose Cística/patologia , Humanos , Pulmão/patologia , Muco/metabolismo , Mucosa Respiratória/patologia , Soroalbumina Bovina/farmacologia , Suínos , Traqueia/metabolismo , Traqueia/patologia
6.
J Virol ; 94(16)2020 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-32493818

RESUMO

The literature on the egress of different herpesviruses after secondary envelopment is contradictory. In this report, we investigated varicella-zoster virus (VZV) egress in a cell line from a child with Pompe disease, a glycogen storage disease caused by a defect in the enzyme required for glycogen digestion. In Pompe cells, both the late autophagy pathway and the mannose-6-phosphate receptor (M6PR) pathway are interrupted. We have postulated that intact autophagic flux is required for higher recoveries of VZV infectivity. To test that hypothesis, we infected Pompe cells and then assessed the VZV infectious cycle. We discovered that the infectious cycle in Pompe cells was remarkably different from that of either fibroblasts or melanoma cells. No large late endosomes filled with VZV particles were observed in Pompe cells; only individual viral particles in small vacuoles were seen. The distribution of the M6PR pathway (trans-Golgi network to late endosomes) was constrained in infected Pompe cells. When cells were analyzed with two different anti-M6PR antibodies, extensive colocalization of the major VZV glycoprotein gE (known to contain M6P residues) and the M6P receptor (M6PR) was documented in the viral highways at the surfaces of non-Pompe cells after maximum-intensity projection of confocal z-stacks, but neither gE nor the M6PR was seen in abundance at the surfaces of infected Pompe cells. Taken together, our results suggested that (i) Pompe cells lack a VZV trafficking pathway within M6PR-positive large endosomes and (ii) most infectious VZV particles in conventional cell substrates are transported via large M6PR-positive vacuoles without degradative xenophagy to the plasma membrane.IMPORTANCE The long-term goal of this research has been to determine why VZV, when grown in cultured cells, invariably is more cell associated and has a lower titer than other alphaherpesviruses, such as herpes simplex virus 1 (HSV1) or pseudorabies virus (PRV). Data from both HSV1 and PRV laboratories have identified a Rab6 secretory pathway for the transport of single enveloped viral particles from the trans-Golgi network within small vacuoles to the plasma membrane. In contrast, after secondary envelopment in fibroblasts or melanoma cells, multiple infectious VZV particles accumulated within large M6PR-positive late endosomes that were not degraded en route to the plasma membrane. We propose that this M6PR pathway is most utilized in VZV infection and least utilized in HSV1 infection, with PRV's usage being closer to HSV1's usage. Supportive data from other VZV, PRV, and HSV1 laboratories about evidence for two egress pathways are included.


Assuntos
Doença de Depósito de Glicogênio Tipo II/metabolismo , Herpesvirus Humano 3/metabolismo , Infecção pelo Vírus da Varicela-Zoster/fisiopatologia , Autofagia/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Varicela/virologia , Endossomos , Exocitose/fisiologia , Herpes Zoster/metabolismo , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 1/patogenicidade , Herpesvirus Humano 3/patogenicidade , Humanos , Macroautofagia/fisiologia , Receptor IGF Tipo 2/metabolismo , Vacúolos , Infecção pelo Vírus da Varicela-Zoster/metabolismo , Proteínas do Envelope Viral/metabolismo , Vírion , Rede trans-Golgi/metabolismo
7.
Proc Natl Acad Sci U S A ; 115(6): 1370-1375, 2018 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-29358407

RESUMO

Differentiated airway epithelia produce sonic hedgehog (SHH), which is found in the thin layer of liquid covering the airway surface. Although previous studies showed that vertebrate HH signaling requires primary cilia, as airway epithelia mature, the cells lose primary cilia and produce hundreds of motile cilia. Thus, whether airway epithelia have apical receptors for SHH has remained unknown. We discovered that motile cilia on airway epithelial cells have HH signaling proteins, including patched and smoothened. These cilia also have proteins affecting cAMP-dependent signaling, including Gαi and adenylyl cyclase 5/6. Apical SHH decreases intracellular levels of cAMP, which reduces ciliary beat frequency and pH in airway surface liquid. These results suggest that apical SHH may mediate noncanonical HH signaling through motile cilia to dampen respiratory defenses at the contact point between the environment and the lung, perhaps counterbalancing processes that stimulate airway defenses.


Assuntos
Brônquios/citologia , Células Epiteliais/metabolismo , Proteínas Hedgehog/metabolismo , Traqueia/citologia , Células Cultivadas , Cílios/metabolismo , Cílios/fisiologia , AMP Cíclico/metabolismo , Células Epiteliais/citologia , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais , Receptor Smoothened/genética , Receptor Smoothened/metabolismo , Proteína Gli2 com Dedos de Zinco/genética , Proteína Gli2 com Dedos de Zinco/metabolismo
8.
Proc Natl Acad Sci U S A ; 114(26): 6842-6847, 2017 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-28607090

RESUMO

Gel-forming mucins, the primary macromolecular components of airway mucus, facilitate airway clearance by mucociliary transport. In cystic fibrosis (CF) altered mucus properties impair mucociliary transport. Airways primarily secrete two closely related gel-forming mucins, MUC5B and MUC5AC. However, their morphologic structures and associations in airways that contain abundant submucosal glands and goblet cells are uncertain. Moreover, there is limited knowledge about mucins in airways not affected by inflammation, infection, or remodeling or in CF airways. Therefore, we examined airways freshly excised from newborn non-CF pigs and CF pigs before secondary manifestations develop. We found that porcine submucosal glands produce MUC5B, whereas goblet cells produce predominantly MUC5AC plus some MUC5B. We found that MUC5B emerged from submucosal gland ducts in the form of strands composed of multiple MUC5B filaments. In contrast, MUC5AC emerged from goblet cells as wispy threads and sometimes formed mucin sheets. In addition, MUC5AC often partially coated the MUC5B strands. Compared with non-CF, MUC5B more often filled CF submucosal gland ducts. MUC5AC sheets also accumulated in CF airways overlying MUC5B strands. These results reveal distinct morphology and interactions for MUC5B and MUC5AC and suggest that the two mucins make distinct contributions to mucociliary transport. Thus, they provide a framework for understanding abnormalities in disease.


Assuntos
Remodelação das Vias Aéreas , Fibrose Cística/metabolismo , Células Caliciformes/metabolismo , Mucina-5AC/metabolismo , Mucina-5B/metabolismo , Animais , Fibrose Cística/genética , Fibrose Cística/patologia , Células Caliciformes/patologia , Camundongos , Camundongos Knockout , Mucina-5AC/genética , Mucina-5B/genética
9.
PLoS One ; 12(1): e0166853, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28056029

RESUMO

INTRODUCTION: Telomerase repairs the telomeric ends of chromosomes and is active in nearly all malignant cells. Hepatitis C virus (HCV) is known to be oncogenic and potential interactions with the telomerase system require further study. We determined the effects of HCV infection on human telomerase reverse transcriptase (TERT) expression and enzyme activity in primary human hepatocytes and continuous cell lines. RESULTS: Primary human hepatocytes and Huh-7.5 hepatoma cells showed early de novo TERT protein expression 2-4 days after infection and these events coincided with increased TERT promoter activation, TERT mRNA, and telomerase activity. Immunoprecipitation studies demonstrated that NS3-4A protease-helicase, in contrast to core or NS5A, specifically bound to the C-terminal region of TERT through interactions between helicase domain 2 and protease sequences. Increased telomerase activity was noted when NS3-4A was transfected into cells, when added to reconstituted mixtures of TERT and telomerase RNA, and when incubated with high molecular weight telomerase 'holoenzyme' complexes. The NS3-4A catalytic effect on telomerase was inhibited with primuline or danoprevir, agents that are known to inhibit NS3 helicase and protease activities respectively. In HCV infected cells, NS3-4A could be specifically recovered with telomerase holoenzyme complexes in contrast to NS5A or core protein. HCV infection also activated the effector caspase 7 which is known to target TERT. Activation coincided with the appearance of lower molecular weight carboxy-terminal fragment(s) of TERT, chiefly sized at 45 kD, which could be inhibited with pancaspase or caspase 7 inhibitors. CONCLUSIONS: HCV infection induces TERT expression and stimulates telomerase activity in addition to triggering Caspase activity that leads to increased TERT degradation. These activities suggest multiple points whereby the virus can influence neoplasia. The NS3-4A protease-helicase can directly bind to TERT, increase telomerase activity, and thus potentially influence telomere repair and host cell neoplastic behavior.


Assuntos
Hepacivirus/patogenicidade , Telomerase/metabolismo , Western Blotting , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Imunofluorescência , Hepatócitos/metabolismo , Humanos , Imunoprecipitação , Regiões Promotoras Genéticas/genética , RNA/metabolismo , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo
10.
J Clin Invest ; 126(3): 879-91, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26808501

RESUMO

Cystic fibrosis (CF) disrupts respiratory host defenses, allowing bacterial infection, inflammation, and mucus accumulation to progressively destroy the lungs. Our previous studies revealed that mucus with abnormal behavior impaired mucociliary transport in newborn CF piglets prior to the onset of secondary manifestations. To further investigate mucus abnormalities, here we studied airway surface liquid (ASL) collected from newborn piglets and ASL on cultured airway epithelia. Fluorescence recovery after photobleaching revealed that the viscosity of CF ASL was increased relative to that of non-CF ASL. CF ASL had a reduced pH, which was necessary and sufficient for genotype-dependent viscosity differences. The increased viscosity of CF ASL was not explained by pH-independent changes in HCO3- concentration, altered glycosylation, additional pH-induced disulfide bond formation, increased percentage of nonvolatile material, or increased sulfation. Treating acidic ASL with hypertonic saline or heparin largely reversed the increased viscosity, suggesting that acidic pH influences mucin electrostatic interactions. These findings link loss of cystic fibrosis transmembrane conductance regulator-dependent alkalinization to abnormal CF ASL. In addition, we found that increasing Ca2+ concentrations elevated ASL viscosity, in part, independently of pH. The results suggest that increasing pH, reducing Ca2+ concentration, and/or altering electrostatic interactions in ASL might benefit early CF.


Assuntos
Fibrose Cística/metabolismo , Muco/metabolismo , Mucosa Respiratória/metabolismo , Células Epiteliais Alveolares/metabolismo , Animais , Bicarbonatos/metabolismo , Sequência de Carboidratos , Células Cultivadas , Fibrose Cística/induzido quimicamente , Feminino , Humanos , Concentração de Íons de Hidrogênio , Masculino , Cloreto de Metacolina , Mucina-5AC/genética , Mucina-5AC/metabolismo , Mucina-5B/genética , Mucina-5B/metabolismo , Polissacarídeos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sus scrofa , Viscosidade
11.
Science ; 345(6198): 818-22, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25124441

RESUMO

Lung disease in people with cystic fibrosis (CF) is initiated by defective host defense that predisposes airways to bacterial infection. Advanced CF is characterized by a deficit in mucociliary transport (MCT), a process that traps and propels bacteria out of the lungs, but whether this deficit occurs first or is secondary to airway remodeling has been unclear. To assess MCT, we tracked movement of radiodense microdisks in airways of newborn piglets with CF. Cholinergic stimulation, which elicits mucus secretion, substantially reduced microdisk movement. Impaired MCT was not due to periciliary liquid depletion; rather, CF submucosal glands secreted mucus strands that remained tethered to gland ducts. Inhibiting anion secretion in non-CF airways replicated CF abnormalities. Thus, impaired MCT is a primary defect in CF, suggesting that submucosal glands and tethered mucus may be targets for early CF treatment.


Assuntos
Fibrose Cística/fisiopatologia , Glândulas Exócrinas/metabolismo , Depuração Mucociliar , Muco/metabolismo , Mucosa Respiratória/fisiopatologia , Sistema Respiratório/fisiopatologia , Animais , Animais Recém-Nascidos , Ânions/metabolismo , Cílios/fisiologia , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Modelos Animais de Doenças , Pulmão/fisiopatologia , Cloreto de Metacolina/farmacologia , Suínos , Traqueia/fisiopatologia
12.
Proc Natl Acad Sci U S A ; 111(24): 8961-6, 2014 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-24889629

RESUMO

Stimulating presynaptic terminals can increase the proton concentration in synapses. Potential receptors for protons are acid-sensing ion channels (ASICs), Na(+)- and Ca(2+)-permeable channels that are activated by extracellular acidosis. Those observations suggest that protons might be a neurotransmitter. We found that presynaptic stimulation transiently reduced extracellular pH in the amygdala. The protons activated ASICs in lateral amygdala pyramidal neurons, generating excitatory postsynaptic currents. Moreover, both protons and ASICs were required for synaptic plasticity in lateral amygdala neurons. The results identify protons as a neurotransmitter, and they establish ASICs as the postsynaptic receptor. They also indicate that protons and ASICs are a neurotransmitter/receptor pair critical for amygdala-dependent learning and memory.


Assuntos
Canais Iônicos Sensíveis a Ácido/genética , Tonsila do Cerebelo/fisiologia , Plasticidade Neuronal , Neurotransmissores/metabolismo , Prótons , Sinapses/fisiologia , Bloqueadores do Canal Iônico Sensível a Ácido/química , Acidose , Tonsila do Cerebelo/metabolismo , Animais , Encéfalo/metabolismo , Eletrodos , Potenciais Pós-Sinápticos Excitadores , Concentração de Íons de Hidrogênio , Canais Iônicos/química , Aprendizagem , Potenciação de Longa Duração , Memória , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/metabolismo , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Peptídeos/química , Venenos de Aranha/química
13.
PLoS One ; 8(12): e83624, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24349537

RESUMO

To develop stem/progenitor cell-based therapy for cystic fibrosis (CF) lung disease, it is first necessary to identify markers of human lung epithelial progenitor/stem cells and to better understand the potential for differentiation into distinct lineages. Here we investigated integrin α6ß4 as an epithelial progenitor cell marker in the human distal lung. We identified a subpopulation of α6ß4(+) cells that localized in distal small airways and alveolar walls and were devoid of pro-surfactant protein C expression. The α6ß4(+) epithelial cells demonstrated key properties of stem cells ex vivo as compared to α6ß4(-) epithelial cells, including higher colony forming efficiency, expression of stem cell-specific transcription factor Nanog, and the potential to differentiate into multiple distinct lineages including basal and Clara cells. Co-culture of α6ß4(+) epithelial cells with endothelial cells enhanced proliferation. We identified a subset of adeno-associated virus (AAVs) serotypes, AAV2 and AAV8, capable of transducing α6ß4(+) cells. In addition, reconstitution of bronchi epithelial cells from CF patients with only 5% normal α6ß4(+) epithelial cells significantly rescued defects in Cl(-) transport. Therefore, targeting the α6ß4(+) epithelial population via either gene delivery or progenitor cell-based reconstitution represents a potential new strategy to treat CF lung disease.


Assuntos
Fibrose Cística/metabolismo , Células Epiteliais/metabolismo , Integrina alfa6beta4/metabolismo , Pulmão/metabolismo , Mucosa Respiratória/metabolismo , Células-Tronco/metabolismo , Células Cultivadas , Fibrose Cística/genética , Fibrose Cística/patologia , Fibrose Cística/terapia , Dependovirus , Células Epiteliais/patologia , Feminino , Terapia Genética , Humanos , Integrina alfa6beta4/genética , Pulmão/patologia , Masculino , Mucosa Respiratória/patologia , Células-Tronco/patologia , Transdução Genética
14.
Mol Ther ; 21(5): 947-53, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23511247

RESUMO

Cystic fibrosis (CF) pigs spontaneously develop sinus and lung disease resembling human CF. The CF pig presents a unique opportunity to use gene transfer to test hypotheses to further understand the pathogenesis of CF sinus disease. In this study, we investigated the ion transport defect in the CF sinus and found that CF porcine sinus epithelia lack cyclic AMP (cAMP)-stimulated anion transport. We asked whether we could restore CF transmembrane conductance regulator gene (CFTR) current in the porcine CF sinus epithelia by gene transfer. We quantified CFTR transduction using an adenovirus expressing CFTR and green fluorescent protein (GFP). We found that as little as 7% of transduced cells restored 6% of CFTR current with 17-28% of transduced cells increasing CFTR current to 50% of non-CF levels. We also found that we could overcorrect cAMP-mediated current in non-CF epithelia. Our findings indicate that CF porcine sinus epithelia lack anion transport, and a relatively small number of cells expressing CFTR are required to rescue the ion transport phenotype. These studies support the use of the CF pig as a preclinical model for future gene therapy trials in CF sinusitis.


Assuntos
Adenoviridae/genética , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/genética , Fibrose Cística/metabolismo , Vetores Genéticos/genética , Mucosa Nasal/metabolismo , Animais , Animais Geneticamente Modificados , Transporte Biológico , AMP Cíclico/metabolismo , Fibrose Cística/terapia , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Modelos Animais de Doenças , Expressão Gênica , Técnicas de Transferência de Genes , Terapia Genética , Proteínas de Fluorescência Verde/genética , Humanos , Mucosa Nasal/ultraestrutura , Sódio/metabolismo , Suínos , Técnicas de Cultura de Tecidos , Transdução Genética , Transgenes
15.
Proc Natl Acad Sci U S A ; 110(8): 3083-8, 2013 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-23382208

RESUMO

Peripheral nervous system abnormalities, including neuropathy, have been reported in people with cystic fibrosis. These abnormalities have largely been attributed to secondary manifestations of the disease. We tested the hypothesis that disruption of the cystic fibrosis transmembrane conductance regulator (CFTR) gene directly influences nervous system function by studying newborn CFTR(-/-) pigs. We discovered CFTR expression and activity in Schwann cells, and loss of CFTR caused ultrastructural myelin sheath abnormalities similar to those in known neuropathies. Consistent with neuropathic changes, we found increased transcripts for myelin protein zero, a gene that, when mutated, can cause axonal and/or demyelinating neuropathy. In addition, axon density was reduced and conduction velocities of the trigeminal and sciatic nerves were decreased. Moreover, in vivo auditory brainstem evoked potentials revealed delayed conduction of the vestibulocochlear nerve. Our data suggest that loss of CFTR directly alters Schwann cell function and that some nervous system defects in people with cystic fibrosis are likely primary.


Assuntos
Animais Recém-Nascidos , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Sistema Nervoso Periférico/fisiopatologia , Animais , Axônios , Sequência de Bases , Sistema Nervoso Central/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Primers do DNA , Masculino , Bainha de Mielina/genética , Bainha de Mielina/patologia , Sistema Nervoso Periférico/metabolismo , Reação em Cadeia da Polimerase , Suínos
16.
Nature ; 487(7405): 109-13, 2012 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-22763554

RESUMO

Cystic fibrosis (CF) is a life-shortening disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Although bacterial lung infection and the resulting inflammation cause most of the morbidity and mortality, how the loss of CFTR function first disrupts airway host defence has remained uncertain. To investigate the abnormalities that impair elimination when a bacterium lands on the pristine surface of a newborn CF airway, we interrogated the viability of individual bacteria immobilized on solid grids and placed onto the airway surface. As a model, we studied CF pigs, which spontaneously develop hallmark features of CF lung disease. At birth, their lungs lack infection and inflammation, but have a reduced ability to eradicate bacteria. Here we show that in newborn wild-type pigs, the thin layer of airway surface liquid (ASL) rapidly kills bacteria in vivo, when removed from the lung and in primary epithelial cultures. Lack of CFTR reduces bacterial killing. We found that the ASL pH was more acidic in CF pigs, and reducing pH inhibited the antimicrobial activity of ASL. Reducing ASL pH diminished bacterial killing in wild-type pigs, and, conversely, increasing ASL pH rescued killing in CF pigs. These results directly link the initial host defence defect to the loss of CFTR, an anion channel that facilitates HCO(3)(-) transport. Without CFTR, airway epithelial HCO(3)(-) secretion is defective, the ASL pH falls and inhibits antimicrobial function, and thereby impairs the killing of bacteria that enter the newborn lung. These findings suggest that increasing ASL pH might prevent the initial infection in patients with CF, and that assaying bacterial killing could report on the benefit of therapeutic interventions.


Assuntos
Fibrose Cística/metabolismo , Fibrose Cística/microbiologia , Pulmão/metabolismo , Pulmão/microbiologia , Viabilidade Microbiana , Sistema Respiratório/metabolismo , Animais , Animais Recém-Nascidos , Anti-Infecciosos/farmacologia , Bicarbonatos/metabolismo , Líquidos Corporais/efeitos dos fármacos , Líquidos Corporais/metabolismo , Fibrose Cística/patologia , Fibrose Cística/terapia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Modelos Animais de Doenças , Feminino , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Transporte de Íons , Pulmão/patologia , Masculino , Viabilidade Microbiana/efeitos dos fármacos , Sus scrofa/microbiologia
17.
Laryngoscope ; 122(9): 1898-905, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22711071

RESUMO

OBJECTIVES/HYPOTHESIS: Chronic sinusitis is nearly universal in humans with cystic fibrosis (CF) and is accompanied by sinus hypoplasia (small sinuses). However, whether impaired sinus development is a primary feature of loss of the cystic fibrosis transmembrane conductance regulator (CFTR) or a secondary consequence of chronic infection remains unknown. Our objective was to study the early pathogenesis of sinus disease in CF. STUDY DESIGN: Animal/basic science research. METHODS: Sinus development was studied in a porcine CF model. RESULTS: Porcine sinus epithelia expressed CFTR and exhibited transepithelial anion transport. Disruption of the CFTR gene eliminated both. Sinuses of newborn CF pigs were not infected and showed no evidence of inflammation, yet were hypoplastic at birth. Older CF pigs spontaneously developed sinus disease similar to that seen in humans with CF. CONCLUSIONS: These results define a role for CFTR in sinus development and suggest the potential of the CF pig as a genetic model of CF-sinus disease in which to test therapeutic strategies to minimize sinus-related CF morbidity.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/genética , Regulação da Expressão Gênica no Desenvolvimento , Seios Paranasais/patologia , Sinusite/genética , Envelhecimento , Animais , Animais Recém-Nascidos , Biópsia por Agulha , Doença Crônica , Fibrose Cística/fisiopatologia , Modelos Animais de Doenças , Feminino , Predisposição Genética para Doença/epidemiologia , Imuno-Histoquímica , Incidência , Masculino , Mucosa Nasal/patologia , Organogênese/genética , Seios Paranasais/embriologia , Distribuição Aleatória , Valores de Referência , Medição de Risco , Sinusite/diagnóstico , Sinusite/epidemiologia , Suínos
18.
Am J Physiol Lung Cell Mol Physiol ; 303(2): L152-60, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22637155

RESUMO

A balance between alveolar liquid absorption and secretion is critical for maintaining optimal alveolar subphase liquid height and facilitating gas exchange in the alveolar space. However, the role of cystic fibrosis transmembrane regulator protein (CFTR) in this homeostatic process has remained elusive. Using a newly developed porcine model of cystic fibrosis, in which CFTR is absent, we investigated ion transport properties and alveolar liquid transport in isolated type II alveolar epithelial cells (T2AECs) cultured at the air-liquid interface. CFTR was distributed exclusively to the apical surface of cultured T2AECs. Alveolar epithelia from CFTR(-/-) pigs failed to increase liquid absorption in response to agents that increase cAMP, whereas cAMP-stimulated liquid absorption in CFTR(+/-) epithelia was similar to that in CFTR(+/+) epithelia. Expression of recombinant CFTR restored stimulated liquid absorption in CFTR(-/-) T2AECs but had no effect on CFTR(+/+) epithelia. In ex vivo studies of nonperfused lungs, stimulated liquid absorption was defective in CFTR(-/-) alveolar epithelia but similar between CFTR(+/+) and CFTR(+/-) epithelia. When epithelia were studied at the air-liquid interface, elevating cAMP levels increased subphase liquid height in CFTR(+/+) but not in CFTR(-/-) T2AECs. Our findings demonstrate that CFTR is required for maximal liquid absorption under cAMP stimulation, but it is not the rate-limiting factor. Furthermore, our data define a role for CFTR in liquid secretion by T2AECs. These insights may help to develop new treatment strategies for pulmonary edema and respiratory distress syndrome, diseases in which lung liquid transport is disrupted.


Assuntos
Células Epiteliais Alveolares/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Água Extravascular Pulmonar/metabolismo , Absorção , Células Epiteliais Alveolares/fisiologia , Animais , Transporte Biológico , Polaridade Celular , Células Cultivadas , Cloretos/metabolismo , AMP Cíclico/fisiologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Impedância Elétrica , Feminino , Técnicas de Inativação de Genes , Técnicas In Vitro , Masculino , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/metabolismo , Tensão Superficial , Sus scrofa , Junções Íntimas/metabolismo
19.
Cell ; 143(6): 911-23, 2010 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-21145458

RESUMO

Defective transepithelial electrolyte transport is thought to initiate cystic fibrosis (CF) lung disease. Yet, how loss of CFTR affects electrolyte transport remains uncertain. CFTR⁻(/)⁻ pigs spontaneously develop lung disease resembling human CF. At birth, their airways exhibit a bacterial host defense defect, but are not inflamed. Therefore, we studied ion transport in newborn nasal and tracheal/bronchial epithelia in tissues, cultures, and in vivo. CFTR⁻(/)⁻ epithelia showed markedly reduced Cl⁻ and HCO3⁻ transport. However, in contrast to a widely held view, lack of CFTR did not increase transepithelial Na(+) or liquid absorption or reduce periciliary liquid depth. Like human CF, CFTR⁻(/)⁻ pigs showed increased amiloride-sensitive voltage and current, but lack of apical Cl⁻ conductance caused the change, not increased Na(+) transport. These results indicate that CFTR provides the predominant transcellular pathway for Cl⁻ and HCO3⁻ in porcine airway epithelia, and reduced anion permeability may initiate CF airway disease.


Assuntos
Ânions/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/metabolismo , Transporte de Íons , Sistema Respiratório/patologia , Animais , Animais Recém-Nascidos , Epitélio/metabolismo , Humanos , Sistema Respiratório/metabolismo , Sus scrofa
20.
Science ; 325(5944): 1131-4, 2009 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-19628819

RESUMO

Cilia are microscopic projections that extend from eukaryotic cells. There are two general types of cilia; primary cilia serve as sensory organelles, whereas motile cilia exert mechanical force. The motile cilia emerging from human airway epithelial cells propel harmful inhaled material out of the lung. We found that these cells express sensory bitter taste receptors, which localized on motile cilia. Bitter compounds increased the intracellular calcium ion concentration and stimulated ciliary beat frequency. Thus, airway epithelia contain a cell-autonomous system in which motile cilia both sense noxious substances entering airways and initiate a defensive mechanical mechanism to eliminate the offending compound. Hence, like primary cilia, classical motile cilia also contain sensors to detect the external environment.


Assuntos
Cílios/fisiologia , Células Epiteliais/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Mucosa Respiratória/metabolismo , Transdução de Sinais , Paladar , Monoterpenos Bicíclicos , Brônquios/citologia , Cálcio/metabolismo , Células Cultivadas , Cílios/metabolismo , Humanos , Monoterpenos/metabolismo , Monoterpenos/farmacologia , Movimento , Noxas , Fosfolipase C beta/metabolismo , Compostos de Amônio Quaternário/metabolismo , Compostos de Amônio Quaternário/farmacologia , Mucosa Respiratória/citologia , Traqueia/citologia , Transducina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA