Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Cell Physiol ; : e31369, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39014912

RESUMEN

Previously we showed hyperosmotic solution caused TRPV1-dependent NKCC1 activation in the lens by a mechanism that involved ERK1/2 signaling. In various tissues, integrins and the cytoskeletal network play a role in responses to osmotic stress. Here, we examined the association between integrins and TRPV1-dependent activation of NKCC1 in mouse lens epithelium. Wild-type (WT) lenses exposed to the integrin agonist leukadherin-1 (LA-1) for 10 min displayed a ~33% increase in the bumetanide-sensitive rate of Rb uptake indicating NKCC activation. Paclitaxel, a microtubule stabilizing agent, abolished the Rb uptake response. In primary cultured lens epithelium LA-1 caused a robust ERK1/2 activation response that was almost fully suppressed by paclitaxel. The TRPV1 agonist capsaicin caused a similar ERK1/2 activation response. Consistent with an association between integrins and TRPV1, the TRPV1 antagonist A889425 prevented the Rb uptake response to LA-1 as did the ERK inhibitor U0126. LA-1 did not increase Rb uptake by lenses from TRPV1 knockout mice. In cells exposed to a hyperosmotic stimulus, both the ERK1/2 activation and Rb uptake responses were prevented by paclitaxel. Taken together, the findings suggest TRPV1 activation is associated with integrins and the tubulin cytoskeleton. This aligned with the observation that LA-1 elicited a robust cytoplasmic calcium rise in cells from WT lenses but failed to increase calcium in cells from TRPV1 knockout lenses. The results are consistent with the notion that integrin activation by LA-1, or a hyperosmotic stimulus, causes TRPV1 channel opening and the consequent downstream activation of the ERK1/2 and NKCC1 responses.

2.
Cell Biochem Biophys ; 79(1): 57-71, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33095400

RESUMEN

Endothelin-1 (ET-1) is a potent endogenously derived vasoconstrictor, which increases pulmonary hypertension via stimulation of [Ca2+]i level in pulmonary artery smooth muscle cells (PASMCs). In this communication, we sought to investigate the mechanism by which ET-1 causes stimulation of Ca2+ concentration in caveolae vesicles of bovine PASMCs (BPASMCs). ET-1 activates PKC-α in the caveolae vesicles by O2.- derived from PKCζ-NADPH oxidase dependent pathway. PKC-α phosphorylates Kv1.5 channels leading to a marked stimulation of Na+ and Ca2+ concentration in the caveolae vesicles. The stimulation of Ca2+ concentration in the caveolae vesicles by ET-1 occurs predominantly via Cav1.2 channels. Additionally, an increase in Na+ concentration by ET-1 due to stimulation of Nav1.5 channels marginally increases Ca2+ level in the caveolae vesicles via reverse-mode Na+/Ca2+ exchanger (NCX-1) and also through "slip-mode conductance" Nav1.5 channels. 4-AP, a well-known inhibitor of Kv channels, also increases Ca2+ concentration in the caveolae vesicles via Cav1.2 channels, reverse-mode NCX-1 and Nav1.5 channels by phosphorylation independent modulation of Kv1.5 channels without the involvement of PKCζ-NADPH oxidase-PKCα signaling axis. Overall, PKCζ-NADPH oxidase-PKCα dependent phosphorylation of Kv1.5 by ET-1 modulates Nav1.5-NCX1-Cav1.2 axis for stimulation of Ca2+ concentration in caveolae vesicles of BPASMCs, which provides a crucial mechanism for better understanding of ET-1-mediated modulation of pulmonary vascular tone.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Endotelina-1/metabolismo , Músculo Liso Vascular/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Proteína Quinasa C/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , 4-Aminopiridina/farmacología , Animales , Calcio/metabolismo , Bovinos , Caveolas/metabolismo , Membrana Celular/metabolismo , Miocitos del Músculo Liso/metabolismo , NADPH Oxidasas/metabolismo , Fosforilación , Isoformas de Proteínas , Proteína Quinasa C-alfa/metabolismo , Arteria Pulmonar/metabolismo , Transducción de Señal , Sodio/metabolismo
3.
Am J Physiol Cell Physiol ; 318(5): C969-C980, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32293931

RESUMEN

The porcine lens response to a hyperosmotic stimulus involves an increase in the activity of an ion cotransporter sodium-potassium/two-chloride cotransporter 1 (NKCC1). Recent studies with agonists and antagonists pointed to a mechanism that appears to depend on activation of transient receptor potential vanilloid 1 (TRPV1) ion channels. Here, we compare responses in lenses and cultured lens epithelium obtained from TRPV1-/- and wild type (WT) mice. Hydrostatic pressure (HP) in lens surface cells was determined using a manometer-coupled microelectrode approach. The TRPV1 agonist capsaicin (100 nM) caused a transient HP increase in WT lenses that peaked after ∼30 min and then returned toward baseline. Capsaicin did not cause a detectable change of HP in TRPV1-/- lenses. The NKCC inhibitor bumetanide prevented the HP response to capsaicin in WT lenses. Potassium transport was examined by measuring Rb+ uptake. Capsaicin increased Rb+ uptake in cultured WT lens epithelial cells but not in TRPV1-/- cells. Bumetanide, A889425, and the Akt inhibitor Akti prevented the Rb+ uptake response to capsaicin. The bumetanide-sensitive (NKCC-dependent) component of Rb+ uptake more than doubled in response to capsaicin. Capsaicin also elicited rapid (<2 min) NKCC1 phosphorylation in WT but not TRPV1-/- cells. HP recovery was shown to be absent in TRPV1-/- lenses exposed to hyperosmotic solution. Bumetanide and Akti prevented HP recovery in WT lenses exposed to hyperosmotic solution. Taken together, responses to capsaicin and hyperosmotic solution point to a functional role for TRPV1 channels in mouse lens. Lack of NKCC1 phosphorylation and Rb+ uptake responses in TRPV1-/- mouse epithelium reinforces the notion that a hyperosmotic challenge causes TRPV1-dependent NKCC1 activation. The results are consistent with a role for the TRPV1-activated signaling pathway leading to NKCC1 stimulation in lens osmotic homeostasis.


Asunto(s)
Cristalino/metabolismo , Miembro 2 de la Familia de Transportadores de Soluto 12/genética , Canales Catiónicos TRPV/genética , Animales , Bumetanida/farmacología , Capsaicina/farmacología , Línea Celular , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Humanos , Presión Hidrostática/efectos adversos , Cristalino/efectos de los fármacos , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Porcinos
4.
Am J Physiol Cell Physiol ; 315(6): C793-C802, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30207782

RESUMEN

Lens ion homeostasis is crucial in maintaining water content and, in turn, refractive index and transparency of the multicellular syncytium-like structure. New information is emerging on the regulation of ion transport in the lens by mechanisms that rely on transient receptor potential vanilloid (TRPV) ion channels. We found recently that TRPV1 activation leads to Ca2+/PKC-dependent ERK1/2 signaling. Here, we show that the TRPV1 agonist capsaicin (100 nM) and hyperosmotic solution (350 vs. 300 mosM) each caused an increase of bumetanide-inhibitable Rb uptake by intact porcine lenses and Na-K-2Cl cotransporter 1 (NKCC1) phosphorylation in the lens epithelium. The TRPV1 antagonist A889425 (1 µM) abolished the increases of Rb uptake and NKCC1 phosphorylation in response to hyperosmotic solution. Exposing lenses to hyperosmotic solution in the presence of MEK/ERK inhibitor U0126 (10 µM) or the with-no-lysine kinase (WNK) inhibitor WNK463 (1 µM) also prevented NKCC1 phosphorylation and the Rb uptake responses to hyperosmotic solution. WNK463 did not prevent the increase in ERK1/2 phosphorylation that occurs in response to capsaicin or hyperosmotic solution, suggesting that ERK1/2 activation occurs before WNK activation in the sequence of signaling events. Taken together, the evidence indicates that activation of TRPV1 is a critical early step in a signaling mechanism that responds to a hyperosmotic stimulus, possibly lens shrinkage. By activating ERK1/2 and WNK, TRPV1 activation leads to NKCC1 phosphorylation and stimulation of NKCC1-mediated ion transport.


Asunto(s)
Epitelio/efectos de los fármacos , Cristalino/efectos de los fármacos , Miembro 2 de la Familia de Transportadores de Soluto 12/genética , Canales Catiónicos TRPV/genética , Animales , Bumetanida/antagonistas & inhibidores , Bumetanida/farmacología , Butadienos/farmacología , Capsaicina/farmacología , Epitelio/metabolismo , Células Gigantes/efectos de los fármacos , Células Gigantes/metabolismo , Humanos , Imidazoles/farmacología , Cristalino/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Nitrilos/farmacología , Presión Osmótica , Fosforilación/efectos de los fármacos , Pirrolidinas/farmacología , Porcinos
5.
Exp Eye Res ; 172: 128-136, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29654770

RESUMEN

Recently we determined that the Transient Receptor Potential Vanilloid 4 ion channel (TRPV4) has a crucial signaling role in a pathway that regulates various aspects of lens epithelium function. Here, we report on a different TRPV channel, TRPV1, in porcine lens. The presence of TRPV1 in the lens was evident from RT-PCR studies and Western blot analysis of MAPK signaling pathway activation caused by the TRPV1 agonist capsaicin. TRPV1 mRNA was detected in the epithelium of porcine as well as human lens. Transient ERK1/2 and p38 MAPK phosphorylation was detected within 1 min in the epithelium isolated from intact porcine lenses exposed to capsaicin (100 nM), a selective TRPV1 agonist, and the response was significantly inhibited by A889245 (1.0 µM), a TRPV1 antagonist. A similar ERK 1/2 and p38 response in the epithelium, also inhibitable by A889245, was evident in lenses treated with hyperosmotic solution (350 vs 300 mOsm). Lenses pre-treated with either the cytosolic Ca2+ chelator BAPTA-AM or the PKC inhibitor sotrastaurin (1.0 µM) had a diminished ERK1/2 activation response to capsaicin and hyperosmotic solution. Taken together the findings support the notion that TRPV1 functions as a plasma membrane ion channel that, when activated, permits the entry of extracellular calcium into the lens epithelium, leading to activation of PKC, ERK1/2 and p38 MAPK. It is significant that the findings confirm earlier proposals that hyperosmotic stress is linked to TRPV1 channel activation in the mouse lens. Further studies are ongoing to determine what functional changes are triggered by the TRPV1-linked signaling pathways and how they might relate to lens volume homeostasis.


Asunto(s)
Células Epiteliales/metabolismo , Regulación de la Expresión Génica/fisiología , Cristalino/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Canales Catiónicos TRPV/genética , Animales , Western Blotting , Capsaicina/farmacología , Células Cultivadas , Humanos , Fosforilación , Piridinas/farmacología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Fármacos del Sistema Sensorial/farmacología , Porcinos , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
Invest Ophthalmol Vis Sci ; 58(11): 4447-4456, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28863406

RESUMEN

Purpose: Na,K-ATPase activity in lens epithelium is subject to control by Src family tyrosine kinases (SFKs). Previously we showed hyposmotic solution causes an SFK-dependent increase in Na,K-ATPase activity in the epithelium. Here we explored the role of cAMP in the signaling mechanism responsible for the SFK and Na,K-ATPase response. Methods: Intact porcine lenses were exposed to hyposmotic Krebs solution (200 mOsm) then the epithelium was assayed for cAMP, SFK phosphorylation (activation) or Na,K-ATPase activity. Results: An increase of cAMP was observed in the epithelium of lenses exposed to hyposmotic solution. In lenses exposed to hyposmotic solution SFK phosphorylation in the epithelium approximately doubled as did Na,K-ATPase activity and both responses were prevented by H89, a protein kinase A inhibitor. The magnitude of the SFK response to hyposmotic solution was reduced by a TRPV4 antagonist HC067047 added to prevent TRPV4-mediated calcium entry, and by a cytoplasmic Ca2+ chelator BAPTA-AM. The Na,K-ATPase activity response in the epithelium of lenses exposed to hyposmotic solution was abolished by BAPTA-AM. As a direct test of cAMP-dependent SFK activation, intact lenses were exposed to 8-pCPT-cAMP, a cell-permeable cAMP analog. 8-pCPT-cAMP caused robust SFK activation. Using Western blot, two calcium-activated adenylyl cyclases, ADCY3 and ADCY8, were detected in lens epithelium. Conclusions: Calcium-activated adenylyl cyclases are expressed in the lens epithelium and SFK activation is linked to a rise of cAMP that occurs upon hyposmotic challenge. The findings point to cAMP as a link between TRPV4 channel-mediated calcium entry, SFK activation, and a subsequent increase of Na,K-ATPase activity.


Asunto(s)
Adenilil Ciclasas/metabolismo , Calcio/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Cristalino/enzimología , Presión Osmótica , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Familia-src Quinasas/metabolismo , Animales , Western Blotting , AMP Cíclico/fisiología , Activación Enzimática , Células Epiteliales/enzimología , Isoquinolinas/farmacología , Mediciones Luminiscentes , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/fisiología , Sulfonamidas/farmacología , Sus scrofa
7.
J Cell Physiol ; 232(6): 1489-1500, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27748508

RESUMEN

Insulin has been shown to elicit changes of Na,K-ATPase activity in various tissues. Na,K-ATPase in the nonpigmented ciliary epithelium (NPE) plays a role in aqueous humor secretion and changes of Na,K-ATPase activity impact the driving force. Because we detect a change of NPE Na,K-ATPase activity in response to insulin, studies were carried out to examine the response mechanism. Ouabain-sensitive rubidium (Rb) uptake by cultured NPE cells, measured as a functional index of Na,K-ATPase-mediated inward potassium transport, was found to increase in cells exposed for 5 min to insulin. The maximally effective concentration was 100 nM. An intrinsic increase of Na,K-ATPase activity evident as a >2-fold increase in the rate of ouabain-sensitive ATP hydrolysis in homogenates obtained from cells exposed to 100 nM insulin for 5 min was also observed. Insulin-treated cells exhibited Akt, Src family kinase (SFK), ERK1/2, and p38 activation, all of which were prevented by a pI3 kinase inhibitor LY294002. The Rb uptake and Na,K-ATPase activity response to insulin both were abolished by PP2, an SFK inhibitor which also prevented p38 and ERK1/2 but not Akt activation. The Akt inhibitor MK-2206 did not change the Na,K-ATPase response to insulin. The findings suggest insulin activates pI3K-dependent Akt and SFK signaling pathways that are separate. ERK1/2 and p38 activation is secondary to and dependent on SFK activation. The increase of Na,K-ATPase activity is dependent on activation of the SFK pathway. The findings are consistent with previous studies that indicate a link between Na,K-ATPase activity and SFK signaling. J. Cell. Physiol. 232: 1489-1500, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Cuerpo Ciliar/metabolismo , Epitelio/metabolismo , Insulina/metabolismo , Pigmentación , Transducción de Señal , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Familia-src Quinasas/metabolismo , Animales , Butadienos/farmacología , Cromonas/farmacología , Cuerpo Ciliar/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Epitelio/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Compuestos Heterocíclicos con 3 Anillos/farmacología , Imidazoles/farmacología , Insulina/farmacología , Modelos Biológicos , Morfolinas/farmacología , Nitrilos/farmacología , Ouabaína/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Pigmentación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piridinas/farmacología , Pirimidinas/farmacología , Rubidio/metabolismo , Transducción de Señal/efectos de los fármacos , Sus scrofa , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
J Ocul Pharmacol Ther ; 32(8): 504-508, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27513167

RESUMEN

To function normally, all cells must maintain ion homeostasis, establish a membrane potential, and regulate water content. These actions require active Na-K transport provided by Na,K-ATPase. The lens, however, is made up almost entirely of fiber cells that have little or no Na,K-ATPase activity. Lens ion and water homeostasis rely on Na,K-ATPase activity in a small number of cells at the periphery of epithelium monolayer. Therefore, the function of the epithelium must be integrated with the needs of the fiber mass. This suggests that a remote control mechanism may adjust Na,K-ATPase activity to match increases or decreases of ion leakage, which may occur a considerable distance away. Here, we review evidence that TRPV4 channels in the epithelium become activated when the lens is subjected to osmotic- or damage-induced swelling. This triggers a chain of events in the lens epithelium that opens connexin hemichannels, allowing ATP release that stimulates purinergic receptors, activates Src family tyrosine kinases, and increases Na,K-ATPase activity. Recent studies also revealed functional connexin hemichannels along with TRPV4 channels in nonpigmented ciliary epithelial (NPE) cells that secrete aqueous humor into the eye. Because TRPV4 channels are mechanosensitive, we speculate they might enable the NPE to respond to stimuli such as mechanical distortion associated with volume homeostasis during fluid transfer across the ciliary epithelium or changes in intraocular pressure.


Asunto(s)
Cuerpo Ciliar/metabolismo , Cristalino/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Humanos
9.
Exp Eye Res ; 132: 52-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25597520

RESUMEN

Exposure to hyposmotic solution causes release of ATP from lens cells via hemichannels. Because hemichannel opening feasibly could swamp the cells with calcium, we carried out studies to measure the magnitude of the increase in cytoplasmic calcium concentration caused by hemichannel opening. In studies on porcine lens epithelial cells in primary culture, propidium iodide (PI) uptake was measured as an index of hemichannel opening. PI uptake was increased significantly in cells exposed to hyposmotic solution. The PI increase under hyposmotic conditions was suppressed by GAP 27, a connexin inhibitor peptide. In studies on cells loaded with Fura-2, continuous exposure to hyposmotic solution caused a cytoplasmic calcium concentration increase that peaked within ∼30 s then remained elevated at or below the peak response for more than 60 min. The peak calcium concentration was 186 ± 2.3 nM compared to a baseline value of 98.0 ± 1.4 nM. The calcium concentration increased a lot further in cells exposed to A23187 (2.5 µM) or the sodium-calcium exchange inhibitor SN-6 (10 µM) added after the onset of the calcium rise in hyposmotic solution. The cytoplasmic calcium increase in hyposmotic solution was abolished by GAP 27. Calcium returned to baseline in cells exposed to hyposmotic solution then treated with GAP 27 starting 2 min after the onset of the calcium rise. The calcium increase in hyposmotic solution did not occur when calcium was eliminated from the bathing medium. The responses to hyposmotic and hyperosmotic stress were different. There was no detectable increase in calcium or PI entry in cells exposed to hyperosmotic solution (500mOsm). In summary, GAP 27-sensitive accumulation of PI by cultured lens epithelium points to connexin hemichannel opening and associated calcium entry. Even though connexins form channels with a large carrying capacity, calcium entry does not increase the cytoplasmic calcium concentration beyond a tolerable physiological range.


Asunto(s)
Calcio/metabolismo , Conexinas/fisiología , Epitelio/metabolismo , Uniones Comunicantes/metabolismo , Cristalino/metabolismo , Análisis de Varianza , Animales , Células Cultivadas , Citoplasma/metabolismo , Modelos Biológicos , Presión Osmótica , Propidio/metabolismo , Porcinos
10.
J Cell Physiol ; 229(3): 343-52, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24037816

RESUMEN

The nitric oxide (NO) donor sodium nitroprusside (SNP) is known to reduce aqueous humor (AH) secretion in the isolated porcine eye. Previously, SNP was found to inhibit Na,K-ATPase activity in nonpigmented ciliary epithelium (NPE), AH-secreting cells, through a cGMP/protein kinase G (PKG)-mediated pathway. Here we show Src family kinase (SFK) activation in the Na,K-ATPase activity response to SNP. Ouabain-sensitive (86) Rb uptake was reduced by >35% in cultured NPE cells exposed to SNP (100 µM) or exogenously added cGMP (8-Br-cGMP) (100 µM) and the SFK inhibitor PP2 (10 µM) prevented the response. Ouabain-sensitive ATP hydrolysis was reduced by ~40% in samples detected in material obtained from SNP- and 8-Br-cGMP-treated cells following homogenization, pointing to an intrinsic change of Na,K-ATPase activity. Tyrosine-10 phosphorylation of Na,K-ATPase α1 subunit was detected in SNP and L-arginine-treated cells and the response prevented by PP2. SNP elicited an increase in cell cGMP. Cells exposed to 8-Br-cGMP displayed SFK activation (phosphorylation) and inhibition of both ouabain-sensitive (86) Rb uptake and Na,K-ATPase activity that was prevented by PP2. SFK activation, which also occurred in SNP-treated cells, was suppressed by inhibitors of soluble guanylate cyclase (ODQ; 10 µM) and PKG (KT5823; 1 µM). SNP and 8-Br-cGMP also increased phosphorylation of ERK1/2 and p38 MAPK and the response prevented by PP2. However, U0126 did not prevent SNP or 8-Br-cGMP-induced inhibition of Na,K-ATPase activity. Taken together, the results suggest that NO activates guanylate cyclase to cause a rise in cGMP and subsequent PKG-dependent SFK activation. Inhibition of Na,K-ATPase activity depends on SFK activation.


Asunto(s)
Cuerpo Ciliar/enzimología , Células Epiteliales/enzimología , Óxido Nítrico/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Familia-src Quinasas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Células Cultivadas , Cuerpo Ciliar/efectos de los fármacos , GMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Activación Enzimática , Activadores de Enzimas/farmacología , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Guanilato Ciclasa/metabolismo , Hidrólisis , Donantes de Óxido Nítrico/farmacología , Fosforilación , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores , Porcinos , Factores de Tiempo , Tirosina , Familia-src Quinasas/antagonistas & inhibidores
11.
Invest Ophthalmol Vis Sci ; 55(1): 187-97, 2014 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-24282227

RESUMEN

PURPOSE: The nonpigmented ciliary epithelium (NPE) is rich in soluble adenylyl cyclase (sAC), a proposed cytoplasmic bicarbonate sensor. Here, we examine the contribution of sAC to an increase in cyclic AMP (cAMP) and changes in a key ion transporter, H(+)-ATPase, in NPE exposed to acetazolamide, a carbonic anhydrase inhibitor (CAI). METHODS: Cyclic AMP was measured by radioimmunoassay in primary cultured porcine NPE. The pH-sensitive dye BCECF was used to examine cytoplasmic pH regulation. Subcellular protein translocation was examined by Western blot. RESULTS: A transient cAMP increase, detectable within minutes of acetazolamide treatment, was prevented by KH7, a specific sAC inhibitor. Following 10-minute exposure to acetazolamide, the abundance of H(+)-ATPase B1 subunit and sAC was doubled in a plasma membrane-rich fraction, suggesting subcellular translocation. Similar evidence of H(+)-ATPase translocation was observed in NPE exposed to 8-Bromoadenosine 3',5'-cyclic monophosphate (8-Br-cAMP). Consistent with increased capacity for proton export, acetazolamide increased the rate of pH recovery from acidification. KH7 and bafilomycin A1, an inhibitor of H(+)-ATPase, both prevented the stimulatory effect of acetazolamide on pH recovery. In a parallel study, H(+)-ATPase abundance was found to be higher in the plasma membrane of HEK293 cells that overexpress sAC compared to the normal HEK293 cells. HEK cells that overexpress sAC and had higher H(+)-ATPase abundance displayed a faster rate of pH recovery and greater sensitivity to KH7. CONCLUSIONS: Acetazolamide increases cAMP in a response that involves activation of sAC. Subcellular translocation of H(+)-ATPase and an increase in the capacity for proton export by acetazolamide-treated NPE cells is a cAMP-dependent response.


Asunto(s)
Acetazolamida/farmacología , Adenilil Ciclasas/metabolismo , Cuerpo Ciliar/enzimología , Células Epiteliales/efectos de los fármacos , Animales , Inhibidores de Anhidrasa Carbónica/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células Cultivadas , Cuerpo Ciliar/citología , Cuerpo Ciliar/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/enzimología , Humanos , Transporte Iónico , Porcinos
12.
Indian J Biochem Biophys ; 51(6): 449-56, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25823216

RESUMEN

Aneurysms develop as a result of chronic inflammation of vascular bed, where progressive destruction of structural proteins, especially elastin and collagen of smooth muscle cells has been shown to manifest. The underlying mechanisms are an increase in local production of proinflammatory cytokines and subsequent increase in proteases, especially matrix metalloproteinases (MMPs) that degrade the structural proteins. The plasminogen system: urokinase-type PA (u-PA), tissue-type PA (t-PA) and plasminogen activator inhibitor-1 (PAI-1) and the MMPs system-MMPs and TIMPs contribute to the progression and development of aneurysms. Recent studies suggest that aneurysms may be genetically determined. To date, most observable candidate genes for aneurysm (elastin, collagen, fibrillin, MMPs and TIMPs) have been explored with little substantiation of the underlying cause and effect. Recently, overexpression of the MMP-2 gene has been suggested as an important phenomenon for aneurysm formation. Along with MMPs, matrix formation also depends on JNK (c-Jun N-terminal kinase) as its activation plays important role in downregulating several genes of matrix production. Under stress, activation of JNK by various stimuli, such as angiotensin II, tumor necrosis factor-α and interleukin-1ß has been noted significantly in vascular smooth muscle cells. Several therapeutic indications corroborate that inhibition of MMP-2 and JNK is useful in preventing progression of vascular aneurysms. This review deals with the role of proteases in the progression of vascular aneurysm.


Asunto(s)
Aneurisma/inmunología , Vasos Sanguíneos/inmunología , Citocinas/inmunología , Modelos Cardiovasculares , Modelos Inmunológicos , Péptido Hidrolasas/inmunología , Transducción de Señal/inmunología , Animales , Activación Enzimática , Humanos
13.
Arch Biochem Biophys ; 540(1-2): 133-44, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24184446

RESUMEN

Treatment of bovine pulmonary artery smooth muscle cells (BPASMCs) with U46619 attenuated isoproterenol caused stimulation of adenyl cyclase activity and cAMP production. Pretreatment with SQ29548 (Tp receptor antagonist), apocynin (NADPH oxidase inhibitor) and Go6976 (PKC-α inhibitor) eliminated U46619 caused attenuation of isoproterenol stimulated adenyl cyclase activity. Pretreatment with SQ29548 and apocynin prevented U46619 induced increase in NADPH oxidase activity, PKC-α activity and Giα phosphorylation. However, pretreatment with CZI, a PKC-ζ inhibitor, markedly, but not completely, inhibited U46619 induced increase in NADPH oxidase activity, PKC-α activity, Giα phosphorylation and also significantly eliminated U46619 caused attenuation of isoproterenol stimulated adenyl cyclase activity. Pretreatment with Go6976 inhibited U46619 induced increase in Giα phosphorylation, but not PKC-ζ activity and NADPH oxidase activity. Pretreatment with pertussis toxin eliminated U46619 caused attenuation of isoproterenol stimulated adenyl cyclase activity without any discernible change in PKC-ζ, NADPH oxidase and PKC-α activities. Transfection of the cells with Tp, PKC-ζ and PKC-α siRNA duplexes corroborate the findings observed with their respective pharmacological inhibitors on the responses produced by U46619. Taken together, we suggest involvement of PKC-ζ in U46619 caused attenuation of isoproterenol stimulated ß-adrenergic response, which is regulated by NADPH oxidase-PKCα-Giα axis in pulmonary artery smooth muscle cells.


Asunto(s)
Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , NADPH Oxidasas/metabolismo , Proteína Quinasa C-alfa/metabolismo , Proteína Quinasa C/metabolismo , Receptores Adrenérgicos beta/metabolismo , Adenilil Ciclasas/metabolismo , Antagonistas Adrenérgicos beta/farmacología , Animales , Bovinos , AMP Cíclico/biosíntesis , Inhibidores Enzimáticos/farmacología , Isoenzimas/metabolismo , Isoproterenol/farmacología , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/metabolismo , NADPH Oxidasas/antagonistas & inhibidores , Arteria Pulmonar/citología
14.
Cell Signal ; 25(2): 512-26, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23159577

RESUMEN

In the context of cross-talk between transmembrane signaling pathways, we studied the loci within the ß-adrenergic receptor/G protein/adenyl cyclase system at which PKC exerts regulatory effects of peroxynitrite (ONOO(-)) on isoproterenol stimulated adenyl cyclase activity in pulmonary artery smooth muscle cells. Treatment of the cells with ONOO(-) stimulated PKC-α activity and that subsequently increased p(38)MAPK phosphorylation. Pretreatment with Go6976 (PKC-α inhibitor) and SB203580 (p(38)MAPK inhibitor) eliminated ONOO(-) caused inhibition on isoproterenol stimulated adenyl cyclase activity. Pretreatment with Go6976, but not SB203580, prevented ONOO(-) induced increase in PKC-α activity. Studies using genetic inhibitors of PKC-α (PKC-α siRNA) and p(38)MAPK (p(38)MAPK siRNA) also corroborated the findings obtained with their pharmacological inhibitors in eliminating the attenuation of ONOO(-) effect on isoproterenol stimulated adenyl cyclase activity. This inhibitory effect of ONOO(-) was found to be eliminated upon pretreatment of the cells with pertussis toxin thereby pointing to a G(i) dependent mechanism. This hypothesis was reinforced by G(i)α phosphorylation as well as by the observation of the loss of the ability of Gpp(NH)p (a measure of G(i) mediated response) to stimulate adenyl cyclase activity upon ONOO(-) treatment to the cells. We suggest the existence of a pertussis toxin sensitive G protein (G(i))-mediated mechanism in isoproterenol stimulated adenyl cyclase activity, which is regulated by PKCα-p(38)MAPK axis dependent phosphorylation of its α-subunit (G(i)α) in the pulmonary artery smooth muscle cells.


Asunto(s)
Antagonistas Adrenérgicos beta/farmacología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Ácido Peroxinitroso/farmacología , Proteína Quinasa C-alfa/metabolismo , Receptores Adrenérgicos beta/química , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Adenilil Ciclasas/metabolismo , Animales , Carbazoles/farmacología , Bovinos , Línea Celular , AMP Cíclico/metabolismo , Isoproterenol/farmacología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Toxina del Pertussis/farmacología , Fosforilación , Proteína Quinasa C-alfa/antagonistas & inhibidores , Proteína Quinasa C-alfa/genética , Arteria Pulmonar , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores Adrenérgicos beta/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética
15.
Arch Biochem Biophys ; 523(2): 169-80, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22568895

RESUMEN

We have recently reported that treatment of bovine pulmonary artery smooth muscle cells with the thromboxane A(2) mimetic, U46619 stimulated NADPH oxidase derived O(2)(·-) level, which subsequently caused marked increase in [Ca(2+)](i)[17]. Herein, we demonstrated that O(2)(·-)-mediated increase in [Ca(2+)](i) stimulates an aprotinin sensitive proteinase activity, which proteolytically activates PKC-α under U46619 treatment to the cells. The activated PKC-α then phosphorylates p(38)MAPK and that subsequently caused G(i)α phosphorylation leading to stimulation of cPLA(2) activity in the cell membrane.


Asunto(s)
Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/enzimología , NADPH Oxidasas/metabolismo , Fosfolipasas A2 Citosólicas/metabolismo , Arteria Pulmonar/citología , Superóxidos/metabolismo , Animales , Materiales Biomiméticos/farmacología , Calcio/metabolismo , Bovinos , Membrana Celular/efectos de los fármacos , Membrana Celular/enzimología , Membrana Celular/metabolismo , Activación Enzimática/efectos de los fármacos , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Proteína Quinasa C-alfa/genética , Proteína Quinasa C-alfa/metabolismo , Tromboxano A2/metabolismo , Vasoconstrictores/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
Am J Physiol Cell Physiol ; 302(12): C1751-61, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22492652

RESUMEN

In several tissues, transient receptor potential vanilloid 4 (TRPV4) channels are involved in the response to hyposmotic challenge. Here we report TRPV4 protein in porcine lens epithelium and show that TRPV4 activation is an important step in the response of the lens to hyposmotic stress. Hyposmotic solution (200 mosM) elicited ATP release from intact lenses and TRPV4 antagonists HC 067047 and RN 1734 prevented the release. In isosmotic solution, the TRPV4 agonist GSK1016790A (GSK) elicited ATP release. When propidium iodide (PI) (MW 668) was present in the bathing medium, GSK and hyposmotic solution both increased PI entry into the epithelium of intact lenses. Increased PI uptake and ATP release in response to GSK and hyposmotic solution were abolished by a mixture of agents that block connexin and pannexin hemichannels, 18α-glycyrrhetinic acid and probenecid. Increased Na-K-ATPase activity occurred in the epithelium of lenses exposed to GSK and 18α-glycyrrhetinic acid + probenecid prevented the response. Hyposmotic solution caused activation of Src family kinase and increased Na-K-ATPase activity in the lens epithelium and TRPV4 antagonists prevented the response. Ionomycin, which is known to increase cytoplasmic calcium, elicited ATP release, the magnitude of which was no greater when lenses were exposed simultaneously to ionomycin and hyposmotic solution. Ionomycin-induced ATP release was significantly reduced in calcium-free medium. TRPV4-mediated calcium entry was examined in Fura-2-loaded cultured lens epithelium. Hyposmotic solution and GSK both increased cytoplasmic calcium that was prevented by TRPV4 antagonists. The cytoplasmic calcium rise in response to hyposmotic solution or GSK was abolished when calcium was removed from the bathing solution. The findings are consistent with hyposmotic shock-induced TRPV4 channel activation which triggers hemichannel-mediated ATP release. The results point to TRPV4-mediated calcium entry that causes a cytoplasmic calcium increase which is an essential early step in the mechanism used by the lens to sense and respond to hyposmotic stress.


Asunto(s)
Adenosina Trifosfato/metabolismo , Conexinas/metabolismo , Epitelio/enzimología , Cristalino/enzimología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Calcio/metabolismo , Conexina 43/metabolismo , Conexinas/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Activación Enzimática , Epitelio/efectos de los fármacos , Proteínas del Ojo/metabolismo , Ácido Glicirretínico/análogos & derivados , Ácido Glicirretínico/farmacología , Cristalino/efectos de los fármacos , Leucina/análogos & derivados , Leucina/farmacología , Morfolinas/farmacología , Presión Osmótica , Probenecid/farmacología , Propidio/metabolismo , Pirroles/farmacología , Sulfonamidas/farmacología , Porcinos , Canales Catiónicos TRPV/efectos de los fármacos , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Familia-src Quinasas/metabolismo
17.
PLoS One ; 6(8): e23835, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21887327

RESUMEN

Apoptosis resistance is a hallmark of cancer cells. Typically, bile acids induce apoptosis. However during gastrointestinal (GI) tumorigenesis the cancer cells develop resistance to bile acid-induced cell death. To understand how bile acids induce apoptosis resistance we first need to identify the molecular pathways that initiate apoptosis in response to bile acid exposure. In this study we examined the mechanism of deoxycholic acid (DCA)-induced apoptosis, specifically the role of Na(+)/H(+) exchanger (NHE) and Na(+) influx in esophageal cells. In vitro studies revealed that the exposure of esophageal cells (JH-EsoAd1, CP-A) to DCA (0.2 mM-0.5 mM) caused lysosomal membrane perturbation and transient cytoplasmic acidification. Fluorescence microscopy in conjunction with atomic absorption spectrophotometry demonstrated that this effect on lysosomes correlated with influx of Na(+), subsequent loss of intracellular K(+), an increase of Ca(2+) and apoptosis. However, ethylisopropyl-amiloride (EIPA), a selective inhibitor of NHE, prevented Na(+), K(+) and Ca(2+) changes and caspase 3/7 activation induced by DCA. Ouabain and amphotericin B, two drugs that increase intracellular Na(+) levels, induced similar changes as DCA (ion imbalance, caspase3/7 activation). On the contrary, DCA-induced cell death was inhibited by medium with low a Na(+) concentrations. In the same experiments, we exposed rat ileum ex-vivo to DCA with or without EIPA. Severe tissue damage and caspase-3 activation was observed after DCA treatment, but EIPA almost fully prevented this response. In summary, NHE-mediated Na(+) influx is a critical step leading to DCA-induced apoptosis. Cells tolerate acidification but evade DCA-induced apoptosis if NHE is inhibited. Our data suggests that suppression of NHE by endogenous or exogenous inhibitors may lead to apoptosis resistance during GI tumorigenesis.


Asunto(s)
Apoptosis/efectos de los fármacos , Cationes/metabolismo , Ácido Desoxicólico/farmacología , Esófago/patología , Neoplasias Gastrointestinales/patología , Intercambiadores de Sodio-Hidrógeno/fisiología , Ácidos y Sales Biliares/farmacología , Células Cultivadas , Neoplasias Gastrointestinales/etiología , Humanos , Hidrógeno/metabolismo , Concentración de Iones de Hidrógeno , Lisosomas , Sodio/metabolismo , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores
18.
Am J Physiol Renal Physiol ; 299(1): F77-90, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20427472

RESUMEN

Recent investigations demonstrate increased Na/H exchanger-1 (NHE-1) activity and plasma levels of ouabain-like factor in spontaneously hypertensive rats. At nanomolar concentrations, ouabain increases Na-K-ATPase activity, induces cell proliferation, and activates complex signaling cascades. We hypothesize that the activity of NHE-1 and Na-K-ATPase are interdependent. To test whether treatment with picomolar ouabain regulates Na-K-ATPase through an NHE-1-dependent mechanism, we examined the role of NHE-1 in ouabain-mediated stimulation of Na-K-ATPase in kidney proximal tubule cell lines [opossum kidney (OK), HK-2, HKC-5, and HKC-11] and rat kidney basolateral membranes. Ouabain stimulated Na-K-ATPase activity and tyrosine phosphorylation in cells that express NHE-1 (OK, HKC-5, and HKC-11) but not in HK-2 cells that express very low levels of NHE-1. Inhibition of NHE-1 with 5 microM EIPA, a NHE-1-specific inhibitor, prevented ouabain-mediated stimulation of (86)Rb uptake and Na-K-ATPase phosphorylation in OK, HKC-5, and HKC-11 cells. Expression of wild-type NHE-1 in HK2 cells restored regulation of Na-K-ATPase by picomolar ouabain. Treatment with picomolar ouabain increased membrane expression of Na-K-ATPase and enhanced NHE-1-Na-K-ATPase alpha1-subunit association. Treatment with ouabain (1 microg x kg body wt(-1) x day(-1)) increased Na-K-ATPase activity, expression, phosphorylation, and association with NHE-1 increased in rat kidney cortical basolateral membranes. Eight days' treatment with ouabain (1 microg x kg body wt(-1) x day(-1)) resulted in increased blood pressure in these rats. These results suggest that the association of NHE-1 with Na-K-ATPase is critical for ouabain-mediated regulation of Na-K-ATPase and that these effects may play a role in cardioglycoside-stimulated hypertension.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Ouabaína/farmacología , Intercambiadores de Sodio-Hidrógeno/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Adenosina Trifosfato/metabolismo , Amilorida/análogos & derivados , Amilorida/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Proteínas de Transporte de Catión/antagonistas & inhibidores , Proteínas de Transporte de Catión/genética , Línea Celular , Relación Dosis-Respuesta a Droga , Activación Enzimática , Humanos , Hidrólisis , Túbulos Renales Proximales/enzimología , Zarigüeyas , Fosforilación , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Intercambiador 1 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Intercambiadores de Sodio-Hidrógeno/genética , Factores de Tiempo , Transfección , Tirosina
19.
Invest Ophthalmol Vis Sci ; 51(6): 3129-38, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20071675

RESUMEN

PURPOSE: Elevated intraocular pressure is associated with glaucomatous optic nerve damage. Other investigators have shown functional changes in optic nerve head astrocytes subjected to elevated hydrostatic pressure (HP) for 1 to 5 days. Recently, the authors reported ERK1/2, p90(RSK) and NHE1 phosphorylation after 2 hours. Here they examine calcium responses at the onset of HP to determine what precedes ERK1/2 phosphorylation. METHODS: Cytoplasmic calcium concentration ([Ca(2+)](i)) was measured in cultured rat optic nerve astrocytes loaded with fura-2. The cells were placed in a closed imaging chamber and subjected to an HP increase of 15 mm Hg. Protein phosphorylation was detected by Western blot analysis. RESULTS: The increase of HP caused an immediate slow increase in [Ca(2+)](i). The response persisted in calcium-free solution and when nickel chloride (4 mM) was added to suppress channel-mediated calcium entry. Previous depletion of the ER calcium stores by cyclopiazonic acid abolished the HP-induced calcium level increase. The HP-induced increase persisted in cells exposed to xestospongin C, an inhibitor of IP3R-mediated calcium release. In contrast, ryanodine receptor (RyR) antagonist ruthenium red (10 microM) or dantrolene (25 microM) inhibited the HP-induced calcium increase. The HP-induced calcium increase was abolished when ryanodine-sensitive calcium stores were pre-depleted with caffeine (3 mM). HP caused ERK1/2 phosphorylation. The magnitude of the ERK1/2 phosphorylation response was reduced by ruthenium red and dantrolene. CONCLUSIONS: Increasing HP causes calcium release from a ryanodine-sensitive cytoplasmic store and subsequent ERK1/2 activation. Calcium store release appears to be a required early step in the initial astrocyte response to an HP increase.


Asunto(s)
Astrocitos/metabolismo , Calcio/metabolismo , Disco Óptico/metabolismo , Rojo de Rutenio/farmacología , Animales , Animales Recién Nacidos , Western Blotting , Bloqueadores de los Canales de Calcio/farmacología , Células Cultivadas , Citoplasma/metabolismo , Dantroleno/farmacología , Femenino , Fura-2/metabolismo , Presión Hidrostática , Masculino , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Quinasa 6 Activada por Mitógenos/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley
20.
Invest Ophthalmol Vis Sci ; 50(12): 5851-8, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19608532

RESUMEN

PURPOSE: To better understand how nitric oxide (NO) alters the function of the nonpigmented ciliary epithelium (NPE), studies were performed to determine the influence of NO on sodium-hydrogen exchanger (NHE) activity. METHODS: Cytoplasmic pH (pH(i)) was measured in cultured porcine NPE loaded with BCECF (2',7'-bis(2-carboxyl)-5(6)-carboxyfluorescein-acetoxyethyl ester). Na-H exchanger (NHE) was examined by immunolocalization. RESULTS: In cells acidified by 5 minutes of exposure to 20 mM ammonium chloride, pH(i) recovery was partially inhibited by sodium nitroprusside (SNP), an NO donor, and l-arginine, the endogenous substrate for NO synthase. SNP and dimethyl amiloride (DMA), an NHE inhibitor, inhibited pH(i) recovery to a similar degree. In bicarbonate-free buffer SNP+DMA elicited no additional change in pH(i) recovery beyond that elicited by DMA alone. This suggests that SNP causes NHE inhibition. the SNP's effect on pH(i) recovery was mimicked by 8-pCPT-cGMP but suppressed by ODQ and H-8. Ouabain alone reduced pH(i) recovery, but SNP+ouabain caused significant further reduction. Immunolocalization studies revealed NHE1 and -4 in native and cultured NPE. CONCLUSIONS: NHE1 and -4 are expressed at the NPE basolateral margin. The findings suggest the NHE is inhibited by NO which acts via a cGMP and protein kinase G signaling pathway. The NHE response does not appear to be the consequence of NO-induced Na,K-ATPase inhibition. Because NO synthases are expressed in porcine NPE, NO could act as an autocrine regulator of NHE activity. Although NHE inhibitors are known to lower intraocular pressure (IOP), further studies are needed to understand whether changes in NHE activity contribute to the IOP-lowering effect of NO donors.


Asunto(s)
Cuerpo Ciliar/citología , Células Epiteliales/metabolismo , Óxido Nítrico/fisiología , Intercambiadores de Sodio-Hidrógeno/metabolismo , Animales , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Epitelio , Fluoresceínas/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Colorantes Fluorescentes/metabolismo , Concentración de Iones de Hidrógeno , Donantes de Óxido Nítrico/farmacología , Nitroprusiato/farmacología , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...