Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Physiol Genomics ; 54(7): 273-282, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35658672

RESUMEN

Ion channels are potentially exploitable as pharmacological targets to treat asthma. This study evaluated the role of KCa3.1 channels, encoded by Kcnn4, in regulating the gene expression of mouse airway epithelium and the development of asthma traits. We used the ovalbumin (OVA) challenge as an asthma model in wild-type and Kcnn4-/- mice, performed histological analysis, and measured serum IgE to evaluate asthma traits. We analyzed gene expression of isolated epithelial cells of trachea or bronchi using mRNA sequencing and gene ontology and performed Ussing chamber experiments in mouse trachea to evaluate anion secretion. Gene expression of epithelial cells from mouse airways differed between trachea and bronchi, indicating regional differences in the inflammatory and transepithelial transport properties of proximal and distal airways. We found that Kcnn4 silencing reduced mast cell numbers, mucus, and collagen in the airways, and reduced the amount of epithelial anion secretion in the OVA-challenged animals. In addition, gene expression was differentially modified in the trachea and bronchi, with Kcnn4 genetic silencing significantly altering the expression of genes involved in the TNF pathway, supporting the potential of KCa3.1 as a therapeutic target for asthma.


Asunto(s)
Asma , Tráquea , Animales , Asma/genética , Asma/metabolismo , Asma/patología , Bronquios/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/metabolismo , Tráquea/metabolismo , Tráquea/patología
2.
Am J Physiol Lung Cell Mol Physiol ; 321(2): L466-L476, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34231389

RESUMEN

Allergic asthma is a chronic airway inflammatory response to different triggers like inhaled allergens. Excessive ATP in fluids from patients with asthma is considered an inflammatory signal and an important autocrine/paracrine modulator of airway physiology. Here, we investigated the deleterious effect of increased extracellular ATP (eATP) concentration on the mucociliary clearance (MCC) effectiveness and determined the role of ATP releasing channels during airway inflammation in an ovalbumin (OVA)-sensitized mouse model. Our allergic mouse model exhibited high levels of eATP measured in the tracheal fluid with a luciferin-luciferase assay and reduced MCC velocity determined by microspheres tracking in the trachea ex vivo. Addition of ATP had a dual effect on MCC, where lower ATP concentration (µM) increased microspheres velocity, whereas higher concentration (mM) transiently stopped microspheres movement. Also, an augmented ethidium bromide uptake by the allergic tracheal airway epithelium suggests an increase in ATP release channel functionality during inflammatory conditions. The use of carbenoxolone, a nonspecific inhibitor of connexin and pannexin1 channels reduced the eATP concentration in the allergic mouse tracheal fluid and dye uptake by the airway epithelium, providing evidence that these ATP release channels are facilitating the net flux of ATP to the lumen during airway inflammation. However, only the specific inhibition of pannexin1 with 10Panx peptide significantly reduced eATP in bronchoalveolar lavage and decreased airway hyperresponsiveness in OVA-allergic mouse model. These data provide evidence that blocking eATP may be a pharmacological alternative to be explored in rescue therapy during episodes of airflow restriction in patients with asthma.


Asunto(s)
Adenosina Trifosfato/inmunología , Asma/inmunología , Carbenoxolona/farmacología , Conexinas/inmunología , Proteínas del Tejido Nervioso/inmunología , Mucosa Respiratoria/inmunología , Tráquea/inmunología , Animales , Asma/inducido químicamente , Asma/tratamiento farmacológico , Asma/patología , Conexinas/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos BALB C , Microesferas , Péptidos/inmunología , Péptidos/farmacología , Mucosa Respiratoria/patología , Tráquea/patología
3.
JCI Insight ; 5(16)2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32814712

RESUMEN

Airway mucociliary clearance (MCC) is the main mechanism of lung defense keeping airways free of infection and mucus obstruction. Airway surface liquid volume, ciliary beating, and mucus are central for proper MCC and critically regulated by sodium absorption and anion secretion. Impaired MCC is a key feature of muco-obstructive diseases. The calcium-activated potassium channel KCa.3.1, encoded by Kcnn4, participates in ion secretion, and studies showed that its activation increases Na+ absorption in airway epithelia, suggesting that KCa3.1-induced hyperpolarization was sufficient to drive Na+ absorption. However, its role in airway epithelium is not fully understood. We aimed to elucidate the role of KCa3.1 in MCC using a genetically engineered mouse. KCa3.1 inhibition reduced Na+ absorption in mouse and human airway epithelium. Furthermore, the genetic deletion of Kcnn4 enhanced cilia beating frequency and MCC ex vivo and in vivo. Kcnn4 silencing in the Scnn1b-transgenic mouse (Scnn1btg/+), a model of muco-obstructive lung disease triggered by increased epithelial Na+ absorption, improved MCC, reduced Na+ absorption, and did not change the amount of mucus but did reduce mucus adhesion, neutrophil infiltration, and emphysema. Our data support that KCa3.1 inhibition attenuated muco-obstructive disease in the Scnn1btg/+ mice. K+ channel modulation may be a therapeutic strategy to treat muco-obstructive lung diseases.


Asunto(s)
Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Enfermedades Pulmonares Obstructivas/etiología , Depuración Mucociliar/fisiología , Animales , Calcio/metabolismo , Células Cultivadas , Cilios/efectos de los fármacos , Cilios/metabolismo , Modelos Animales de Enfermedad , Epitelio/metabolismo , Femenino , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/antagonistas & inhibidores , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Pulmón/fisiopatología , Enfermedades Pulmonares Obstructivas/genética , Masculino , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Depuración Mucociliar/efectos de los fármacos , Sodio/metabolismo
6.
Front Physiol ; 10: 694, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31263421

RESUMEN

Calcium-activated anion secretion is expected to ameliorate cystic fibrosis, a genetic disease that carries an anion secretory defect in exocrine tissues. Human patients and animal models of the disease that present a mild intestinal phenotype have been postulated to bear a compensatory calcium-activated anion secretion in the intestine. TMEM16A is calcium-activated anion channel whose presence in the intestinal epithelium is contradictory. We aim to test the functional expression of TMEM16A using animal models with Cftr and/or Tmem16a intestinal silencing. Expression of TMEM16A was studied in a wild type and intestinal Tmem16a knockout mice by mRNA-seq, mass-spectrometry, q-PCR, Western blotting and immunolocalization. Calcium-activated anion secretion was recorded in the ileum and proximal colon of these animals including intestinal Cftr knockout and double mutants with dual Tmem16a and Cftr intestinal ablation. Mucus homeostasis was studied by immune-analysis of Mucin-2 (Muc2) and survival curves were recorded. Tmem16a transcript was found in intestine. Nevertheless, protein was barely detected in colon samples. Electrophysiological measurements demonstrated that the intestinal deletion of Tmem16a did not change calcium-activated anion secretion induced by carbachol or ATP in ileum and proximal colon. Muc2 architecture was not altered by Tmem16a silencing as was observed when Cftr was deleted from mouse intestine. Tmem16a silencing neither affected animal survival nor modified the lethality observed in the intestinal Cftr-null mouse. Our results demonstrate that TMEM16A function in the murine intestine is not related to electrogenic calcium-activated anion transport and does not affect mucus homeostasis and survival of animals.

7.
Sci Rep ; 7(1): 9439, 2017 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-28842605

RESUMEN

Deficient insulin signaling is a key event mediating diabetic glomerulopathy. Additionally, diabetic kidney disease has been related to increased levels of adenosine. Therefore, we tested a link between insulin deficiency and dysregulated activity of the equilibrative nucleoside transporters (ENTs) responsible for controlling extracellular levels of adenosine. In ex vivo glomeruli, high D-glucose decreased nucleoside uptake mediated by ENT1 and ENT2 transporters, resulting in augmented extracellular levels of adenosine. This condition was reversed by exposure to insulin. Particularly, insulin through insulin receptor/PI3K pathway markedly upregulated ENT2 uptake activity to restores the extracellular basal level of adenosine. Using primary cultured rat podocytes as a cellular model, we found insulin was able to increase ENT2 maximal velocity of transport. Also, PI3K activity was necessary to maintain ENT2 protein levels in the long term. In glomeruli of streptozotocin-induced diabetic rats, insulin deficiency leads to decreased activity of ENT2 and chronically increased extracellular levels of adenosine. Treatment of diabetic rats with adenosine deaminase attenuated both the glomerular loss of nephrin and proteinuria. In conclusion, we evidenced ENT2 as a target of insulin signaling and sensitive to dysregulation in diabetes, leading to chronically increased extracellular adenosine levels and thereby setting conditions conducive to kidney injury.


Asunto(s)
Adenosina/metabolismo , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Transportador Equilibrativo 2 de Nucleósido/genética , Insulina/metabolismo , Animales , Biopsia , Nefropatías Diabéticas/patología , Transportador Equilibrativo 2 de Nucleósido/metabolismo , Espacio Extracelular/metabolismo , Regulación de la Expresión Génica , Cinética , Masculino , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas , Transducción de Señal
8.
Cytokine ; 88: 115-125, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27599257

RESUMEN

Renal fibrosis is a common irreversible process of chronic kidney disease (CKD) characterized by uncontrolled deposits of extracellular matrix, replacement of cellular parenchyma and progressive loss of renal function. Recent evidence suggests that a series of phenotypic transformations of resident renal cells are responsible for the formation of interstitial myofibroblasts, cells that play a key role in the fibrotic process. In the renal glomerulus transformation of mesangial cells to myofibroblasts is an event that orchestrates glomerulosclerosis and the participation of other cells types has also been suggested. Recent findings clarify the role of tubular epithelium in mediating the generation of ECM producing cells in the tubule interstitium. Also, crosstalk between injured cells and myofibroblasts for amplification of the fibrogenic cascade in CKD occurs. The crucial conductor of these changes in the kidney is the transforming growth factor-ß (TGF-ß). Thus, this review focuses on the control of this cytokines signaling mechanisms and their dysregulation in CKD. Further, some of the promising interventional alternatives targeting TGF-ß are also discussed.


Asunto(s)
Túbulos Renales/metabolismo , Miofibroblastos/metabolismo , Insuficiencia Renal Crónica/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Fibrosis , Humanos , Túbulos Renales/patología , Miofibroblastos/patología , Insuficiencia Renal Crónica/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA