Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Am J Physiol Renal Physiol ; 315(6): F1626-F1636, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30207167

RESUMEN

Transient receptor potential cation channel subfamily V member 4 (TRPV4)-mediated Ca2+ signaling induces early activation of small/intermediate Ca2+-activated K+ channels, SK3 (KCNN3) and IK1 (KCNN4), which leads to membrane hyperpolarization and enhanced Ca2+ influx, which is critical for subsequent activation of the large conductance Ca2+-activated K+ channel BK (KCNMA1) and K+ secretion in kidney cortical collecting duct (CCD) cells. The focus of the present study was to determine if such coordinated hierarchical/sequential activation of these channels in CCD was orchestrated within caveolae, a known microcompartment underlying selective Ca2+-signaling events in other cells. In K+-secreting mouse principal cell (PC) line, mCCDcl1 cells, knockdown of caveolae caveolin-1 (CAV-1) depressed TRPV4-mediated Ca2+ signaling and activation of SK3, intermediate conductance channel (IK1), and BK. Immunofluorescence colocalization analysis and coimmunoprecipitation assays demonstrated direct coupling of TRPV4 with each of the KCa channels in both mCCDcl1 and whole mouse kidney homogenates. Likewise, extending this analysis to CAV-1 demonstrates colocalization and direct coupling of CAV-1 with TRPV4, SK3, IK1, and BK, providing strong support for coupling of the channels in caveolae microdomains. Furthermore, differential expression of CAV-1 along the CCD was apparent where CAV-1 was strongly expressed within and along the cell borders of kidney PCs and intercalated cells (ICs), although significantly less in ICs. It is concluded that caveolae provide a key microdomain in PCs and ICs for coupling of TRPV4 with SK3, IK1, and BK that directly contributes to TRPV4-mediated Ca2+ signaling in these domains leading to rapid and sequential coupling of TRPV4-SK3/IK1-BK that may play a central role in mediating Ca2+-dependent regulation of BK and K+ secretion.


Asunto(s)
Señalización del Calcio , Caveolas/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Túbulos Renales Colectores/metabolismo , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Caveolina 1/genética , Caveolina 1/metabolismo , Línea Celular , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Túbulos Renales Colectores/citología , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Potenciales de la Membrana , Ratones Endogámicos C57BL , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/genética , Canales Catiónicos TRPV/genética
2.
J Allergy Clin Immunol ; 141(2): 608-619.e7, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28807414

RESUMEN

BACKGROUND: Chronic itch is a highly debilitating symptom that underlies many medical disorders with no universally effective treatments. Although unique neuronal signaling cascades in the sensory ganglia and spinal cord have been shown to critically promote the pathogenesis of chronic itch, the role of skin-associated cells remains poorly understood. OBJECTIVE: We sought to examine the cutaneous mechanisms underlying transient receptor potential vanilloid 4 (TRPV4)-mediated allergic and nonallergic chronic itch. METHODS: Expression of TRPV4 in chronic itch and healthy control skin preparations was examined by using real-time RT-PCR. Trpv4eGFP mice were used to study the expression and function of TRPV4 in the skin by means of immunofluorescence staining, flow cytometry, calcium imaging, and patch-clamp recordings. Genetic and pharmacologic approaches were used to examine the role and underlying mechanisms of TRPV4 in mouse models of dry skin-associated chronic itch and spontaneous scratching associated with squaric acid dibutylester-induced allergic contact dermatitis. RESULTS: TRPV4 is selectively expressed by dermal macrophages and epidermal keratinocytes in mice. Lineage-specific deletion of TRPV4 in macrophages and keratinocytes reduces allergic and nonallergic chronic itch in mice, respectively. Importantly, TRPV4 expression is significantly increased in skin biopsy specimens from patients with chronic idiopathic pruritus in comparison with skin from healthy control subjects. Moreover, TRPV4-dependent chronic itch requires 5-hydroxytryptamine (5-HT) signaling secondary to activation of distinct 5-HT receptors in mice with allergic and those with nonallergic chronic itch conditions. CONCLUSION: Our study reveals previously unrecognized mechanisms by which TRPV4-expressing epithelial and immune cells in the skin critically and dynamically mediate chronic itch and unravels novel targets for therapeutics in the setting of chronic itch.


Asunto(s)
Dermatitis Alérgica por Contacto/inmunología , Dermis/inmunología , Regulación de la Expresión Génica/inmunología , Queratinocitos/inmunología , Macrófagos/inmunología , Prurito/inmunología , Canales Catiónicos TRPV/inmunología , Animales , Enfermedad Crónica , Dermatitis Alérgica por Contacto/genética , Dermatitis Alérgica por Contacto/patología , Dermis/patología , Femenino , Regulación de la Expresión Génica/genética , Humanos , Queratinocitos/patología , Macrófagos/patología , Masculino , Ratones , Ratones Noqueados , Prurito/genética , Prurito/patología , Canales Catiónicos TRPV/genética
3.
Am J Physiol Renal Physiol ; 312(6): F1081-F1089, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28274924

RESUMEN

The large-conductance Ca2+-activated K+ channel, BK (KCNMA1), is expressed along the connecting tubule (CNT) and cortical collecting duct (CCD) where it underlies flow- and Ca2+-dependent K+ secretion. Its activity is partially under the control of the mechanosensitive transient receptor potential vanilloid type 4 (TRPV4) Ca2+-permeable channel. Recently, we identified three small-/intermediate-conductance Ca2+-activated K+ channels, SK1 (KCNN1), SK3 (KCNN3), and IK1 (KCNN4), with notably high Ca2+-binding affinities, that are expressed in CNT/CCD and may be regulated by TRPV4-mediated Ca2+ influx. The K+-secreting CCD mCCDcl1 cells, which express these channels, were used to determine whether SK1/3 and IK1 are activated on TRPV4 stimulation and whether they contribute to Ca2+ influx and activation of BK. Activation of TRPV4 (GSK1016790A) modestly depolarized the membrane potential and robustly increased intracellular Ca2+, [Ca2+]i Inhibition of both SK1/3 and IK1 by application of apamin and 1-[(2-chlorophenyl)diphenylmethyl]-1H-pyrazole (TRAM-34), respectively, further depolarized the membrane potential and markedly suppressed the TRPV4-mediated rise in [Ca2+]i Application of BK inhibitor iberiotoxin after activation of TRPV4 without apamin/TRAM-34 also reduced [Ca2+]i and further intensified membrane depolarization, demonstrating BK involvement. However, the BK-dependent effects on [Ca2+]i and membrane potential were largely abolished by pretreatment with apamin and TRAM-34, identical to that observed by separately suppressing TRPV4-mediated Ca2+ influx, demonstrating that SK1/3-IK1 channels potently contribute to TRPV4-mediated BK activation. Our data indicate a direct correlation between TRPV4-mediated Ca2+ signal and BK activation but where early activation of SK1/3 and IK1 channels are critical to sufficiently enhanced Ca2+ entry and [Ca2+]i levels required for activation of BK.


Asunto(s)
Calcio/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Túbulos Renales Colectores/metabolismo , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Potasio/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Agonistas de los Canales de Calcio/farmacología , Señalización del Calcio , Células Cultivadas , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/antagonistas & inhibidores , Túbulos Renales Colectores/efectos de los fármacos , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/antagonistas & inhibidores , Potenciales de la Membrana , Ratones , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/antagonistas & inhibidores , Canales Catiónicos TRPV/agonistas
4.
Kidney Int ; 91(6): 1398-1409, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28187982

RESUMEN

To maintain potassium homeostasis, kidneys exert flow-dependent potassium secretion to facilitate kaliuresis in response to elevated dietary potassium intake. This process involves stimulation of calcium-activated large conductance maxi-K (BK) channels in the distal nephron, namely the connecting tubule and the collecting duct. Recent evidence suggests that the TRPV4 channel is a critical determinant of flow-dependent intracellular calcium elevations in these segments of the renal tubule. Here, we demonstrate that elevated dietary potassium intake (five percent potassium) increases renal TRPV4 mRNA and protein levels in an aldosterone-dependent manner and causes redistribution of the channel to the apical plasma membrane in native collecting duct cells. This, in turn, leads to augmented TRPV4-mediated flow-dependent calcium ion responses in freshly isolated split-opened collecting ducts from mice fed the high potassium diet. Genetic TRPV4 ablation greatly diminished BK channel activity in collecting duct cells pointing to a reduced capacity to excrete potassium. Consistently, elevated potassium intake induced hyperkalemia in TRPV4 knockout mice due to deficient renal potassium excretion. Thus, regulation of TRPV4 activity in the distal nephron by dietary potassium is an indispensable component of whole body potassium balance.


Asunto(s)
Hiperpotasemia/metabolismo , Túbulos Renales/metabolismo , Potasio en la Dieta/metabolismo , Eliminación Renal , Canales Catiónicos TRPV/metabolismo , Adaptación Fisiológica , Animales , Calcio/metabolismo , Predisposición Genética a la Enfermedad , Homeostasis , Hiperpotasemia/genética , Hiperpotasemia/fisiopatología , Túbulos Renales/fisiopatología , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Potasio en la Dieta/administración & dosificación , Receptores de Mineralocorticoides/metabolismo , Canales Catiónicos TRPV/deficiencia , Canales Catiónicos TRPV/genética
6.
PLoS One ; 11(5): e0155006, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27159616

RESUMEN

The voltage- and Ca2+-activated, large conductance K+ channel (BK, maxi-K) is expressed in the collecting duct system of kidney where it underlies flow- and Ca2+-dependent K+ excretion. To determine if other Ca2+-activated K+ channels (KCa) may participate in this process, mouse kidney and the K+-secreting mouse cortical collecting duct (CCD) cell line, mCCDcl1, were assessed for TRPV4 and KCa channel expression and cross-talk. qPCR mRNA analysis and immunocytochemical staining demonstrated TRPV4 and KCa expression in mCCDcl1 cells and kidney connecting tubule (CNT) and CCD. Three subfamilies of KCa channels were revealed: the high Ca2+-binding affinity small-conductance SK channels, SK1and SK3, the intermediate conductance channel, IK1, and the low Ca2+-binding affinity, BK channel (BKα subunit). Apparent expression levels varied in CNT/CCD where analysis of CCD principal cells (PC) and intercalated cells (IC) demonstrated differential staining: SK1:PCIC, IK1:PC>IC, BKα:PC = IC, and TRPV4:PC>IC. Patch clamp analysis and fluorescence Ca2+ imaging of mCCDcl1 cells demonstrated potent TRPV4-mediated Ca2+ entry and strong functional cross-talk between TRPV4 and KCa channels. TRPV4-mediated Ca2+ influx activated each KCa channel, as evidenced by selective inhibition of KCa channels, with each active KCa channel enhancing Ca2+ entry (due to membrane hyperpolarization). Transepithelial electrical resistance (TEER) analysis of confluent mCCDcl1 cells grown on permeable supports further demonstrated this cross-talk where TRPV4 activation induce a decrease in TEER which was partially restored upon selective inhibition of each KCa channel. It is concluded that SK1/SK3 and IK1 are highly expressed along with BKα in CNT and CCD and are closely coupled to TRPV4 activation as observed in mCCDcl1 cells. The data support a model in CNT/CCD segments where strong cross talk between TRPV4-mediated Ca2+ influx and each KCa channel leads to enhance Ca2+ entry which will support activation of the low Ca2+-binding affinity BK channel to promote BK-mediated K+ secretion.


Asunto(s)
Túbulos Renales Colectores/metabolismo , Canales de Potasio Calcio-Activados/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Línea Celular , Ratones , Ratones Endogámicos C57BL
7.
PLoS One ; 9(4): e95149, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24762817

RESUMEN

The Ca2+-activated, maxi-K (BK) K+ channel, with low Ca2+-binding affinity, is expressed in the distal tubule of the nephron and contributes to flow-dependent K+ secretion. In the present study we demonstrate that the Ca2+-activated, SK3 (KCa2.3) K+ channel, with high Ca2+-binding affinity, is also expressed in the mouse kidney (RT-PCR, immunoblots). Immunohistochemical evaluations using tubule specific markers demonstrate significant expression of SK3 in the distal tubule and the entire collecting duct system, including the connecting tubule (CNT) and cortical collecting duct (CCD). In CNT and CCD, main sites for K+ secretion, the highest levels of expression were along the apical (luminal) cell membranes, including for both principal cells (PCs) and intercalated cells (ICs), posturing the channel for Ca2+-dependent K+ secretion. Fluorescent assessment of cell membrane potential in native, split-opened CCD, demonstrated that selective activation of the Ca2+-permeable TRPV4 channel, thereby inducing Ca2+ influx and elevating intracellular Ca2+ levels, activated both the SK3 channel and the BK channel leading to hyperpolarization of the cell membrane. The hyperpolarization response was decreased to a similar extent by either inhibition of SK3 channel with the selective SK antagonist, apamin, or by inhibition of the BK channel with the selective antagonist, iberiotoxin (IbTX). Addition of both inhibitors produced a further depolarization, indicating cooperative effects of the two channels on Vm. It is concluded that SK3 is functionally expressed in the distal nephron and collecting ducts where induction of TRPV4-mediated Ca2+ influx, leading to elevated intracellular Ca2+ levels, activates this high Ca2+-affinity K+ channel. Further, with sites of expression localized to the apical cell membrane, especially in the CNT and CCD, SK3 is poised to be a key pathway for Ca2+-dependent regulation of membrane potential and K+ secretion.


Asunto(s)
Túbulos Renales Distales/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Canales Catiónicos TRPV/fisiología , Animales , Acuaporina 2/metabolismo , Señalización del Calcio , Expresión Génica , Riñón/citología , Riñón/fisiología , Potenciales de la Membrana , Ratones Endogámicos C57BL , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/genética
8.
Nat Struct Mol Biol ; 21(2): 180-8, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24440983

RESUMEN

FGFR2-expressing human cancer cells with low concentrations of the adaptor protein Grb2 show high prevalence for metastatic outcome. In nonstimulated cells, the SH3 domain (and not the SH2 domains) of Plcγ1 directly competes for a binding site at the very C terminus of FGFR2 with the C-terminal SH3 domain of Grb2. Reduction of Grb2 concentration permits Plcγ1 access to the receptor. Recruitment of Plcγ1 in this way is sufficient to upregulate phospholipase activity. This results in elevated phosphatidylinositol 4,5-bisphosphate turnover and intracellular calcium levels, thus leading to increased cell motility and promotion of cell-invasive behavior in the absence of extracellular receptor stimulation. Therefore, metastatic outcome can be dictated by the constitutive competition between Grb2 and Plcγ1 for the phosphorylation-independent binding site on FGFR2.


Asunto(s)
Proteína Adaptadora GRB2/fisiología , Fosfolipasa C gamma/fisiología , Fosfolipasas/fisiología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Sitios de Unión , Unión Competitiva , Línea Celular Tumoral , Proteína Adaptadora GRB2/metabolismo , Células HEK293 , Humanos , Modelos Genéticos , Invasividad Neoplásica/genética , Fosfolipasa C gamma/metabolismo , Estructura Terciaria de Proteína
9.
Pain ; 154(10): 2130-2141, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23811042

RESUMEN

Chronic neuropathic pain is often a severe and inadequately treated consequence of spinal cord injury (SCI). Recent findings suggest that SCI pain is promoted by spontaneous activity (SA) generated chronically in cell bodies of primary nociceptors in dorsal root ganglia (DRG). Many nociceptors express transient receptor potential V1 (TRPV1) channels, and in a preceding study most dissociated DRG neurons exhibiting SA were excited by the TRPV1 activator, capsaicin. The present study investigated roles of TRPV1 channels in behavioral hypersensitivity and nociceptor SA after SCI. Contusive SCI at thoracic segment T10 increased expression of TRPV1 protein in lumbar DRG 1 month after injury and enhanced capsaicin-evoked ion currents and Ca2+ responses in dissociated small DRG neurons. A major role for TRPV1 channels in pain-related behavior was indicated by the ability of a specific TRPV1 antagonist, AMG9810, to reverse SCI-induced hypersensitivity of hind limb withdrawal responses to mechanical and thermal stimuli at a dose that did not block detection of noxious heat. Similar reversal of behavioral hypersensitivity was induced by intrathecal oligodeoxynucleotides antisense to TRPV1, which knocked down TRPV1 protein and reduced capsaicin-evoked currents. TRPV1 knockdown also decreased the incidence of SA in dissociated nociceptors after SCI. Prolonged application of very low concentrations of capsaicin produced nondesensitizing firing similar to SA, and this effect was enhanced by prior SCI. These results show that TRPV1 makes important contributions to pain-related hypersensitivity long after SCI, and suggest a role for TRPV1-dependent enhancement of nociceptor SA that offers a promising target for treating chronic pain after SCI.


Asunto(s)
Hiperalgesia/metabolismo , Hiperalgesia/psicología , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/psicología , Canales Catiónicos TRPV/fisiología , Animales , Técnicas de Silenciamiento del Gen/métodos , Hiperalgesia/fisiopatología , Masculino , Potenciales de la Membrana/fisiología , Ratas , Ratas Sprague-Dawley
10.
J Biol Chem ; 288(28): 20306-14, 2013 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-23709216

RESUMEN

We have recently documented that the Ca(2+)-permeable TRPV4 channel, which is abundantly expressed in distal nephron cells, mediates cellular Ca(2+) responses to elevated luminal flow. In this study, we combined Fura-2-based [Ca(2+)]i imaging with immunofluorescence microscopy in isolated split-opened distal nephrons of C57BL/6 mice to probe the molecular determinants of TRPV4 activity and subcellular distribution. We found that activation of the PKC pathway with phorbol 12-myristate 13-acetate significantly increased [Ca(2+)]i responses to flow without affecting the subcellular distribution of TRPV4. Inhibition of PKC with bisindolylmaleimide I diminished cellular responses to elevated flow. In contrast, activation of the PKA pathway with forskolin did not affect TRPV4-mediated [Ca(2+)]i responses to flow but markedly shifted the subcellular distribution of the channel toward the apical membrane. These actions were blocked with the specific PKA inhibitor H-89. Concomitant activation of the PKA and PKC cascades additively enhanced the amplitude of flow-induced [Ca(2+)]i responses and greatly increased basal [Ca(2+)]i levels, indicating constitutive TRPV4 activation. This effect was precluded by the selective TRPV4 antagonist HC-067047. Therefore, the functional status of the TRPV4 channel in the distal nephron is regulated by two distinct signaling pathways. Although the PKA-dependent cascade promotes TRPV4 trafficking and translocation to the apical membrane, the PKC-dependent pathway increases the activity of the channel on the plasma membrane.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Nefronas/metabolismo , Proteína Quinasa C/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Calcio/metabolismo , Colforsina/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Activación Enzimática/efectos de los fármacos , Fura-2/química , Fura-2/metabolismo , Técnicas In Vitro , Indoles/farmacología , Isoquinolinas/farmacología , Túbulos Renales Colectores/metabolismo , Maleimidas/farmacología , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Morfolinas/farmacología , Nefronas/efectos de los fármacos , Perfusión/métodos , Ésteres del Forbol/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/efectos de los fármacos , Pirroles/farmacología , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología , Canales Catiónicos TRPV/antagonistas & inhibidores
11.
Methods Mol Biol ; 998: 371-84, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23529445

RESUMEN

Transient receptor potential (TRP) channels are expressed in almost every segment of renal nephron from the glomerulus to the inner medullary collecting duct. Serving as a route for Ca(2+) entry from the intratubular space into cells in response to external cues, TRP channels modulate water-electrolyte transport, thus determining functional properties of the renal tubule. In this chapter, we discuss technical aspects of using Ca(2+) imaging to monitor activity of TRP channels in situ, namely, in the freshly isolated distal nephrons, with a special emphasis on the mechanosensitive TRPV4 channel and its role in tubular flow sensing.


Asunto(s)
Calcio/metabolismo , Imagen Molecular/métodos , Nefronas/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Animales , Inmunohistoquímica , Ratones , Nefronas/citología , Permeabilidad , Ratas , Estrés Mecánico
12.
J Am Soc Nephrol ; 24(4): 604-16, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23411787

RESUMEN

The molecular mechanism of cyst formation and expansion in autosomal recessive polycystic kidney disease (ARPKD) is poorly understood, but impaired mechanosensitivity to tubular flow and dysfunctional calcium signaling are important contributors. The activity of the mechanosensitive Ca(2+)-permeable TRPV4 channel underlies flow-dependent Ca(2+) signaling in murine collecting duct (CD) cells, suggesting that this channel may contribute to cystogenesis in ARPKD. Here, we developed a method to isolate CD-derived cysts and studied TRPV4 function in these cysts laid open as monolayers and in nondilated split-open CDs in a rat model of ARPKD. In freshly isolated CD-derived cyst monolayers, we observed markedly impaired TRPV4 activity, abnormal subcellular localization of the channel, disrupted TRPV4 glycosylation, decreased basal [Ca(2+)]i, and loss of flow-mediated [Ca(2+)]i signaling. In contrast, nondilated CDs of these rats exhibited functional TRPV4 with largely preserved mechanosensitive properties. Long-term systemic augmentation of TRPV4 activity with a selective TRPV4 activator significantly attenuated the renal manifestations of ARPKD in a time-dependent manner. At the cellular level, selective activation of TRPV4 restored mechanosensitive Ca(2+) signaling as well as the function and subcellular distribution of TRPV4. In conclusion, the functional status of TRPV4, which underlies mechanosensitive Ca(2+) signaling in CD cells, inversely correlates with renal cystogenesis in ARPKD. Augmenting TRPV4 activity may have therapeutic potential in ARPKD.


Asunto(s)
Calcio/metabolismo , Quistes/metabolismo , Túbulos Renales Colectores/patología , Riñón Poliquístico Autosómico Recesivo/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Quistes/fisiopatología , Túbulos Renales Colectores/metabolismo , Túbulos Renales Colectores/fisiopatología , Riñón Poliquístico Autosómico Recesivo/fisiopatología , Ratas , Ratas Sprague-Dawley , Transducción de Señal
13.
Pflugers Arch ; 465(2): 177-86, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23207579

RESUMEN

Kidneys are complex highly organized paired organs of nearly one million nephrons each. They rigorously process about 180 l of plasma daily to keep whole body homeostasis. To effectively perform such a titanic work, kidneys rely on mechanisms able to sense dynamic changes in composition and flow rates of protourine along the renal tubule. It is envisioned that Ca(2+)-permeable transient receptor potential (TRP) channels, and specifically mechanosensitive TRPV4, can serve to interpret these external mechanical cues in the form of elevated intracellular Ca(2+) concentration. This, in turn, initiates multiple cellular responses and adaptation mechanisms. The current review summarizes up-to-date knowledge about the sites of TRPV4 expression in renal tissue as well as discusses the functional role of the channel in cellular responses to hypotonicity and tubular flow. We will also provide insights as to how TRPV4 fits into classical polycystin mechanosensory complex in cilia and will speculate about previously underappreciated clinical implication of pharmacological TRPV4 targeting in treatment of polycystic kidney disease.


Asunto(s)
Riñón/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Calcio/metabolismo , Expresión Génica , Humanos , Riñón/fisiología , Enfermedades Renales Poliquísticas/metabolismo , Canales Catiónicos TRPV/genética
14.
Biochim Biophys Acta ; 1821(7): 994-1002, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22521764

RESUMEN

Nonsteroidal anti-inflammatory drugs (NSAIDs) are one of the most widely consumed pharmaceuticals, yet both the mechanisms involved in their therapeutic actions and side-effects, notably gastrointestinal (GI) ulceration/bleeding, have not been clearly defined. In this study, we have used a number of biochemical, structural, computational and biological systems including; Fourier Transform InfraRed (FTIR). Nuclear Magnetic Resonance (NMR) and Surface Plasmon Resonance (SPR) spectroscopy, and cell culture using a specific fluorescent membrane probe, to demonstrate that NSAIDs have a strong affinity to form ionic and hydrophobic associations with zwitterionic phospholipids, and specifically phosphatidylcholine (PC), that are reversible and non-covalent in nature. We propose that the pH-dependent partition of these potent anti-inflammatory drugs into the phospholipid bilayer, and possibly extracellular mono/multilayers present on the luminal interface of the mucus gel layer, may result in profound changes in the hydrophobicity, fluidity, permeability, biomechanical properties and stability of these membranes and barriers. These changes may not only provide an explanation of how NSAIDs induce surface injury to the GI mucosa as a component in the pathogenic mechanism leading to peptic ulceration and bleeding, but potentially an explanation for a number of (COX-independent) biological actions of this family of pharmaceuticals. This insight also has proven useful in the design and development of a novel class of PC-associated NSAIDs that have reduced GI toxicity while maintaining their essential therapeutic efficacy to inhibit pain and inflammation.


Asunto(s)
Antiinflamatorios no Esteroideos/química , Aspirina/química , Mucosa Gástrica/efectos de los fármacos , Ibuprofeno/química , Naproxeno/química , Fosfatidilcolinas/química , Antiinflamatorios no Esteroideos/farmacología , Aspirina/farmacología , Línea Celular Tumoral , Permeabilidad de la Membrana Celular/efectos de los fármacos , Colorantes Fluorescentes , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patología , Humanos , Concentración de Iones de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Ibuprofeno/farmacología , Membrana Dobles de Lípidos/química , Espectroscopía de Resonancia Magnética , Simulación de Dinámica Molecular , Naproxeno/farmacología , Compuestos de Piridinio , Espectroscopía Infrarroja por Transformada de Fourier , Resonancia por Plasmón de Superficie
15.
J Biol Chem ; 287(12): 8782-91, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22298783

RESUMEN

The TRPV4 Ca(2+)-permeable channel is sensitive to mechanical stimuli. In the current study we have employed immunocytochemical staining in kidney slices and functional assessments (Ca(2+) imaging) in isolated, split-opened, tubule segments to define TRPV4 sites of expression and flow-dependent function in the collecting duct system. Staining patterns revealed strong expression of TRPV4 along the entire collecting duct system with highest levels at the apical (luminal)/subapical region of the principal cells (PCs), the dominant cell type, with more diffuse staining in intercalated cells (ICs). Using fluorescence Ca(2+) imaging and the selective TRPV4 agonist, GSK1016790A, we demonstrated functional TRPV4 channels in PCs and ICs of split-opened cortical collecting ducts and connecting tubules. The agonist was ineffective in inducing a rise in [Ca(2+)](i) in the absence of extracellular Ca(2+) or in tubules from TRPV4-deficient animals. Most importantly, a 10-fold elevation in luminal (apical) fluid flow induced a rapid and sustained influx of Ca(2+) that was abolished by the TRPV channel inhibitor, ruthenium red, or in tubules isolated from TRPV4 deficient animals. We concluded that TRPV4 is highly expressed along the entire collecting duct system where it appears to function as a sensor/transducer of flow-induce mechanical stresses.


Asunto(s)
Túbulos Renales/metabolismo , Mecanotransducción Celular , Canales Catiónicos TRPV/metabolismo , Animales , Ratones , Estrés Mecánico , Canales Catiónicos TRPV/genética
16.
Cell Calcium ; 51(2): 131-9, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22204737

RESUMEN

The mouse cortical collecting duct (CCD) M-1 cells were grown to confluency on coverslips to assess the interaction between TRPV4 and Ca(2+)-activated K(+) channels. Immunocytochemistry demonstrated strong expression of TRPV4, along with the CCD marker, aquaporin-2, and the Ca(2+)-activated K(+) channels, the small conductance SK3 (K(Ca)2.3) channel and large conductance BKα channel (K(Ca)1.1). TRPV4 overexpression studies demonstrated little physical dependency of the K(+) channels on TRPV4. However, activation of TRPV4 by hypotonic swelling (or GSK1016790A, a selective agonist) or inhibition by the selective antagonist, HC-067047, demonstrated a strong dependency of SK3 and BK-α activation on TRPV4-mediated Ca(2+) influx. Selective inhibition of BK-α channel (Iberiotoxin) or SK3 channel (apamin), thereby depolarizing the cells, further revealed a significant dependency of TRPV4-mediated Ca(2+) influx on activation of both K(+) channels. It is concluded that a synergistic cross-talk exists between the TRPV4 channel and SK3 and BK-α channels to provide a tight functional regulation between the channel groups. This cross-talk may be progressive in nature where the initial TRPV4-mediated Ca(2+) influx would first activate the highly Ca(2+)-sensitive SK3 channel which, in turn, would lead to enhanced Ca(2+) influx and activation of the less Ca(2+)-sensitive BK channel.


Asunto(s)
Calcio/metabolismo , Túbulos Renales Colectores/metabolismo , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Apamina/farmacología , Acuaporina 2/biosíntesis , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Túbulos Renales Colectores/citología , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Leucina/análogos & derivados , Leucina/farmacología , Ratones , Péptidos/farmacología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/agonistas , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/antagonistas & inhibidores , Sulfonamidas/farmacología
17.
Brain Res ; 1436: 1-12, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22192412

RESUMEN

The microvessels of the brain are very sensitive to mechanical stresses such as observed in traumatic brain injury (TBI). Such stresses can quickly lead to dysfunction of the microvessel endothelial cells, including disruption of blood-brain barrier (BBB). It is now evident that elevation of cytosolic calcium levels ([Ca2+]i) can compromise the BBB integrity, however the mechanism by which mechanical injury can produce a [Ca2+]i increase in brain endothelial cells is unclear. To assess the effects of mechanical/stretch injury on [Ca2+]i signaling, mouse brain microvessel endothelial cells (bEnd3) were grown to confluency on elasticized membranes and [Ca2+]i monitored using fura 2 fluorescence imaging. Application of an injury, using a pressure/stretch pulse of 50 ms, induced a rapid transient increase in [Ca2+]i. In the absence of extracellular Ca2+, the injury-induced [Ca2+]i transient was greatly reduced, but not fully eliminated, while unloading of Ca2+ stores by thapsigargin treatment in the absence of extracellular Ca2+ abolished the injury transient. Application of LOE-908 and amiloride, TRPC and TRPP2 channel blockers, respectively, both reduced the transient [Ca2+]i increase. Further, siRNA knockdown assays directed at TRPC1 and TRPP2 expression also resulted in a reduction of the injury-induced [Ca2+]i response. In addition, stretch injury induced increases of NO production and actin stress fiber formation, both of which were markedly reduced upon treatment with LOE908 and/or amiloride. We conclude that mechanical injury of brain endothelial cells induces a rapid influx of calcium, mediated by TRPC1 and TRPP2 channels, which leads to NO synthesis and actin cytoskeletal rearrangement.


Asunto(s)
Barrera Hematoencefálica/citología , Barrera Hematoencefálica/metabolismo , Calcio/metabolismo , Células Endoteliales/metabolismo , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPP/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Señalización del Calcio , Línea Celular , Técnicas de Silenciamiento del Gen , Ratones , Óxido Nítrico/metabolismo , Estrés Mecánico
18.
Cell Calcium ; 50(6): 502-9, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21930300

RESUMEN

There is controversy as to whether TRP channels participate in mediating store-operated current (I(SOC)) and store-operated Ca(2+) entry (SOCE). Our recent study has demonstrated that TRPC1 forms heteromeric channels with TRPV4 in vascular endothelial cells and that Ca(2+) store depletion enhances the vesicle trafficking of heteromeric TRPV4-C1 channels, causing insertion of more channels into the plasma membrane in vascular endothelial cells. In the present study, we determined whether the enhanced TRPV4-C1 insertion to the plasma membrane could contribute to SOCE and I(SOC). We found that thapsigargin-induced SOCE was much lower in aortic endothelial cells derived from trpv4(-/-) or trpc1(-/-) knockout mice when compared to that of wild-type mice. In human umbilical vein endothelial cells (HUVECs), thapsigargin-induced SOCE was markedly reduced by knocking down the expression of TRPC1 and/or TRPV4 with respective siRNAs. Brefeldin A, a blocker of vesicular translocation, inhibited the SOCE. These results suggest that an enhanced vesicular trafficking of heteromeric TRPV4-C1 channels contributes to SOCE in vascular endothelial cells. Vascular tension studies suggest that such an enhanced trafficking of TRPV4-C1 channels may play a role in thapsigargin-induced vascular relaxation in rat small mesenteric arteries.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Brefeldino A/farmacología , Canales de Calcio/genética , Línea Celular , Fura-2 , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones , Mutación , Técnicas de Placa-Clamp , ARN Interferente Pequeño , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/fisiología , Tapsigargina/farmacología
19.
PLoS One ; 6(8): e22824, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21850238

RESUMEN

Mechanical forces are known to induce increases of [Ca(2+)](i) in the aldosterone-sensitive distal nephron (ASDN) cells to regulate epithelial transport. At the same time, mechanical stress stimulates ATP release from ASDN cells. In this study, we combined ratiometric Fura-2 based monitoring of [Ca(2+)](i) in freshly isolated split-opened ASDN with targeted deletion of P2Y2 and TRPV4 in mice to probe a role for purinergic signaling in mediating mechano-sensitive responses in ASDN cells. ATP application causes a reproducible transient Ca(2+) peak followed by a sustained plateau. Individual cells of the cortical collecting duct (CCD) and the connecting tubule (CNT) respond to purinergic stimulation with comparative elevations of [Ca(2+)](i). Furthermore, ATP-induced Ca(2+)-responses are nearly identical in both principal (AQP2-positive) and intercalated (AQP2-negative) cells as was confirmed using immunohistochemistry in split-opened ASDN. UTP application produces elevations of [Ca(2+)](i) similar to that observed with ATP suggesting a dominant role of P2Y2-like receptors in generation of [Ca(2+)](i) response. Indeed, genetic deletion of P2Y2 receptors decreases the magnitude of ATP-induced and UTP-induced Ca(2+) responses by more than 70% and 90%, respectively. Both intracellular and extracellular sources of Ca(2+) appeared to contribute to the generation of ATP-induced Ca(2+) response in ASDN cells. Importantly, flow- and hypotonic-induced Ca(2+) elevations are markedly blunted in P2Y2 -/- mice. We further demonstrated that activation of mechano-sensitive TRPV4 channel plays a major role in the sustained [Ca(2+)](i) elevation during purinergic stimulation. Consistent with this, ATP-induced Ca(2+) plateau are dramatically attenuated in TRV4 -/- mice. Inhibition of TRPC channels with 10 µM BTP2 also decreased ATP-induced Ca(2+) plateau whilst to a lower degree than that observed with TRPV4 inhibition/genetic deletion. We conclude that stimulation of purinergic signaling by mechanical stimuli leads to activation of TRPV4 and, to a lesser extent, TRPCs channels, and this is an important component of mechano-sensitive response of the ASDN.


Asunto(s)
Aldosterona/metabolismo , Calcio/metabolismo , Mecanotransducción Celular/fisiología , Nefronas/metabolismo , Receptores Purinérgicos P2Y2/metabolismo , Canales Catiónicos TRPV/metabolismo , Adenosina Trifosfato/farmacología , Animales , Inmunohistoquímica , Técnicas In Vitro , Masculino , Mecanotransducción Celular/efectos de los fármacos , Mecanotransducción Celular/genética , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Nefronas/citología , Nefronas/efectos de los fármacos , Receptores Purinérgicos P2Y2/genética , Canales Catiónicos TRPV/genética , Uridina Trifosfato/farmacología
20.
Am J Physiol Renal Physiol ; 300(5): F1105-15, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21325499

RESUMEN

Activation of the renal kallikrein-kinin system results in natriuresis and diuresis, suggesting its possible role in renal tubular sodium transport regulation. Here, we used patch-clamp electrophysiology to directly assess the effects of bradykinin (BK) on the epithelial Na(+) channel (ENaC) activity in freshly isolated split-opened murine aldosterone-sensitive distal nephrons (ASDNs). BK acutely inhibits ENaC activity by reducing channel open probability (P(o)) in a dose-dependent and reversible manner. Inhibition of B2 receptors with icatibant (HOE-140) abolished BK actions on ENaC. In contrast, activation of B1 receptors with the selective agonist Lys-des-Arg(9)-BK failed to reproduce BK actions on ENaC. This is consistent with B2 receptors playing a critical role in mediating BK signaling to ENaC. BK has little effect on ENaC P(o) when G(q/11) was inhibited with Gp antagonist 2A. Moreover, inhibition of phospholipase C (PLC) with U73122, but not saturation of cellular cAMP levels with the membrane-permeable nonhydrolysable cAMP analog 8-cpt-cAMP, prevents BK actions on ENaC activity. This argues that BK stimulates B2 receptors with subsequent activation of G(q/11)-PLC signaling cascade to acutely inhibit ENaC activity. Activation of BK signaling acutely depletes apical PI(4,5)P(2) levels. However, inhibition of Ca(2+) pump SERCA of the endoplasmic reticulum with thapsigargin does not prevent BK signaling to ENaC. Furthermore, caffeine, while producing a similar rise in [Ca(2+)](i) as in response to BK stimulation, fails to recapitulate BK actions on ENaC. Therefore, we concluded that BK acutely inhibits ENaC P(o) in mammalian ASDN via stimulation of B2 receptors and following depletion of PI(4,5)P(2), but not increases in [Ca(2+)](i).


Asunto(s)
Aldosterona/metabolismo , Bradiquinina/farmacología , Bloqueadores del Canal de Sodio Epitelial , Activación del Canal Iónico/efectos de los fármacos , Natriuresis/efectos de los fármacos , Nefronas/efectos de los fármacos , Bloqueadores de los Canales de Sodio/farmacología , Sodio/metabolismo , Absorción , Animales , Técnicas Biosensibles , Cafeína/farmacología , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Relación Dosis-Respuesta a Droga , Canales Epiteliales de Sodio/metabolismo , Estrenos/farmacología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Hidrólisis , Calidina/análogos & derivados , Calidina/farmacología , Masculino , Potenciales de la Membrana , Ratones , Ratones Endogámicos C57BL , Nefronas/metabolismo , Técnicas de Placa-Clamp , Fosfatidilinositol 4,5-Difosfato/metabolismo , Inhibidores de Fosfodiesterasa/farmacología , Pirrolidinonas/farmacología , Receptor de Bradiquinina B1/agonistas , Receptor de Bradiquinina B1/metabolismo , Receptor de Bradiquinina B2/agonistas , Receptor de Bradiquinina B2/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/antagonistas & inhibidores , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Transducción de Señal/efectos de los fármacos , Tapsigargina/farmacología , Tionucleótidos/farmacología , Técnicas de Cultivo de Tejidos , Fosfolipasas de Tipo C/antagonistas & inhibidores , Fosfolipasas de Tipo C/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA