Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Proc Natl Acad Sci U S A ; 115(49): E11465-E11474, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30455320

RESUMEN

A-kinase anchoring proteins (AKAPs) shape second-messenger signaling responses by constraining protein kinase A (PKA) at precise intracellular locations. A defining feature of AKAPs is a helical region that binds to regulatory subunits (RII) of PKA. Mining patient-derived databases has identified 42 nonsynonymous SNPs in the PKA-anchoring helices of five AKAPs. Solid-phase RII binding assays confirmed that 21 of these amino acid substitutions disrupt PKA anchoring. The most deleterious side-chain modifications are situated toward C-termini of AKAP helices. More extensive analysis was conducted on a valine-to-methionine variant in the PKA-anchoring helix of AKAP18. Molecular modeling indicates that additional density provided by methionine at position 282 in the AKAP18γ isoform deflects the pitch of the helical anchoring surface outward by 6.6°. Fluorescence polarization measurements show that this subtle topological change reduces RII-binding affinity 8.8-fold and impairs cAMP responsive potentiation of L-type Ca2+ currents in situ. Live-cell imaging of AKAP18γ V282M-GFP adducts led to the unexpected discovery that loss of PKA anchoring promotes nuclear accumulation of this polymorphic variant. Targeting proceeds via a mechanism whereby association with the PKA holoenzyme masks a polybasic nuclear localization signal on the anchoring protein. This led to the discovery of AKAP18ε: an exclusively nuclear isoform that lacks a PKA-anchoring helix. Enzyme-mediated proximity-proteomics reveal that compartment-selective variants of AKAP18 associate with distinct binding partners. Thus, naturally occurring PKA-anchoring-defective AKAP variants not only perturb dissemination of local second-messenger responses, but also may influence the intracellular distribution of certain AKAP18 isoforms.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/química , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas de la Membrana/metabolismo , Proteínas de Anclaje a la Quinasa A/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación Enzimológica de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Proteínas de la Membrana/genética , Modelos Moleculares , Polimorfismo de Nucleótido Simple , Unión Proteica , Conformación Proteica , Isoformas de Proteínas , Transporte de Proteínas
2.
Biochim Biophys Acta Mol Cell Res ; 1865(9): 1341-1355, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29959960

RESUMEN

L-type CaV1.2 channels are key regulators of gene expression, cell excitability and muscle contraction. CaV1.2 channels organize in clusters throughout the plasma membrane. This channel organization has been suggested to contribute to the concerted activation of adjacent CaV1.2 channels (e.g. cooperative gating). Here, we tested the hypothesis that dynamic intracellular and perimembrane trafficking of CaV1.2 channels is critical for formation and dissolution of functional channel clusters mediating cooperative gating. We found that CaV1.2 moves in vesicular structures of circular and tubular shape with diverse intracellular and submembrane trafficking patterns. Both microtubules and actin filaments are required for dynamic movement of CaV1.2 vesicles. These vesicles undergo constitutive homotypic fusion and fission events that sustain CaV1.2 clustering, channel activity and cooperative gating. Our study suggests that CaV1.2 clusters and activity can be modulated by diverse and unique intracellular and perimembrane vesicular dynamics to fine-tune Ca2+ signals.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Canales de Calcio Tipo L/metabolismo , Microtúbulos/metabolismo , Vesículas Transportadoras/metabolismo , Señalización del Calcio , Línea Celular , Membrana Celular/metabolismo , Citoplasma/metabolismo , Humanos , Activación del Canal Iónico , Transporte de Proteínas
3.
J Physiol ; 597(8): 2139-2162, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30714156

RESUMEN

KEY POINTS: Prevailing dogma holds that activation of the ß-adrenergic receptor/cAMP/protein kinase A signalling pathway leads to enhanced L-type CaV 1.2 channel activity, resulting in increased Ca2+ influx into ventricular myocytes and a positive inotropic response. However, the full mechanistic and molecular details underlying this phenomenon are incompletely understood. CaV 1.2 channel clusters decorate T-tubule sarcolemmas of ventricular myocytes. Within clusters, nanometer proximity between channels permits Ca2+ -dependent co-operative gating behaviour mediated by physical interactions between adjacent channel C-terminal tails. We report that stimulation of cardiomyocytes with isoproterenol, evokes dynamic, protein kinase A-dependent augmentation of CaV 1.2 channel abundance along cardiomyocyte T-tubules, resulting in the appearance of channel 'super-clusters', and enhanced channel co-operativity that amplifies Ca2+ influx. On the basis of these data, we suggest a new model in which a sub-sarcolemmal pool of pre-synthesized CaV 1.2 channels resides in cardiomyocytes and can be mobilized to the membrane in times of high haemodynamic or metabolic demand, to tune excitation-contraction coupling. ABSTRACT: Voltage-dependent L-type CaV 1.2 channels play an indispensable role in cardiac excitation-contraction coupling. Activation of the ß-adrenergic receptor (ßAR)/cAMP/protein kinase A (PKA) signalling pathway leads to enhanced CaV 1.2 activity, resulting in increased Ca2+ influx into ventricular myocytes and a positive inotropic response. CaV 1.2 channels exhibit a clustered distribution along the T-tubule sarcolemma of ventricular myocytes where nanometer proximity between channels permits Ca2+ -dependent co-operative gating behaviour mediated by dynamic, physical, allosteric interactions between adjacent channel C-terminal tails. This amplifies Ca2+ influx and augments myocyte Ca2+ transient and contraction amplitudes. We investigated whether ßAR signalling could alter CaV 1.2 channel clustering to facilitate co-operative channel interactions and elevate Ca2+ influx in ventricular myocytes. Bimolecular fluorescence complementation experiments reveal that the ßAR agonist, isoproterenol (ISO), promotes enhanced CaV 1.2-CaV 1.2 physical interactions. Super-resolution nanoscopy and dynamic channel tracking indicate that these interactions are expedited by enhanced spatial proximity between channels, resulting in the appearance of CaV 1.2 'super-clusters' along the z-lines of ISO-stimulated cardiomyocytes. The mechanism that leads to super-cluster formation involves rapid, dynamic augmentation of sarcolemmal CaV 1.2 channel abundance after ISO application. Optical and electrophysiological single channel recordings confirm that these newly inserted channels are functional and contribute to overt co-operative gating behaviour of CaV 1.2 channels in ISO stimulated myocytes. The results of the present study reveal a new facet of ßAR-mediated regulation of CaV 1.2 channels in the heart and support the novel concept that a pre-synthesized pool of sub-sarcolemmal CaV 1.2 channel-containing vesicles/endosomes resides in cardiomyocytes and can be mobilized to the sarcolemma to tune excitation-contraction coupling to meet metabolic and/or haemodynamic demands.


Asunto(s)
Canales de Calcio Tipo L/fisiología , Miocitos Cardíacos/fisiología , Receptores Adrenérgicos beta/fisiología , Agonistas Adrenérgicos beta/farmacología , Animales , Línea Celular , Femenino , Ventrículos Cardíacos/citología , Humanos , Isoproterenol/farmacología , Masculino , Ratones Endogámicos C57BL , Sarcolema/fisiología
4.
EMBO J ; 31(7): 1764-73, 2012 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-22343944

RESUMEN

The deregulation of brain cholesterol metabolism is typical in acute neuronal injury (such as stroke, brain trauma and epileptic seizures) and chronic neurodegenerative diseases (Alzheimer's disease). Since both conditions are characterized by excessive stimulation of glutamate receptors, we have here investigated to which extent excitatory neurotransmission plays a role in brain cholesterol homeostasis. We show that a short (30 min) stimulation of glutamatergic neurotransmission induces a small but significant loss of membrane cholesterol, which is paralleled by release to the extracellular milieu of the metabolite 24S-hydroxycholesterol. Consistent with a cause-effect relationship, knockdown of the enzyme cholesterol 24-hydroxylase (CYP46A1) prevented glutamate-mediated cholesterol loss. Functionally, the loss of cholesterol modulates the magnitude of the depolarization-evoked calcium response. Mechanistically, glutamate-induced cholesterol loss requires high levels of intracellular Ca(2+), a functional stromal interaction molecule 2 (STIM2) and mobilization of CYP46A1 towards the plasma membrane. This study underscores the key role of excitatory neurotransmission in the control of membrane lipid composition, and consequently in neuronal membrane organization and function.


Asunto(s)
Colesterol/metabolismo , Ácido Glutámico/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Transmisión Sináptica , Animales , Calcio/metabolismo , Proteínas de Unión al Calcio/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Colesterol 24-Hidroxilasa , Técnicas de Silenciamiento del Gen , Ácido Glutámico/farmacología , Hipocampo/efectos de los fármacos , Hidroxicolesteroles/metabolismo , Proteínas de la Membrana/metabolismo , Neuronas/efectos de los fármacos , Ratas , Ratas Wistar , Esteroide Hidroxilasas/genética , Esteroide Hidroxilasas/metabolismo , Molécula de Interacción Estromal 2
5.
Pflugers Arch ; 467(10): 2107-19, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25559845

RESUMEN

Calcium signaling participates in different cellular processes leading to cell migration. TRPV4, a non-selective cation channel that responds to mechano-osmotic stimulation and heat, is also involved in cell migration. However, the mechanistic involvement of TRPV4 in cell migration is currently unknown. We now report that expression of the mutant channel TRPV4-(121)AAWAA (lacking the phosphoinositide-binding site (121)KRWRK(125) and the response to physiological stimuli) altered HEK293 cell migration. Altered migration patterns included periods of fast and persistent motion followed by periods of stalling and turning, and the extension of multiple long cellular protrusions. TRPV4-WT overexpressing cells showed almost complete loss of directionality with frequent turns, no progression, and absence of long protrusions. Traction microscopy revealed higher tractions forces in the tail of TRPV4-(121)AAWAA than in TRPV4-WT expressing cells. These results are consistent with a defective and augmented tail retraction in TRPV4-(121)AAWAA- and TRPV4-WT-expressing cells, respectively. The activity of calpain, a protease implicated in focal adhesion (FA) disassembly, was decreased in TRPV4-(121)AAWAA compared with TRPV4-WT-expressing cells. Consistently, larger focal adhesions were seen in TRPV4-(121)AAWAA compared with TRPV4-WT-expressing HEK293 cells, a result that was also reproduced in T47D and U87 cells. Similarly, overexpression of the pore-dead mutant TRPV4-M680D resumed the TRPV4-(121)AAWAA phenotype presenting larger FA. The migratory phenotype obtained in HEK293 cells overexpressing TRPV4-(121)AAWAA was mimicked by knocking-down TRPC1, a cationic channel that participates in cell migration. Together, our results point to the participation of TRPV4 in the dynamics of trailing adhesions, a function that may require the interplay of TRPV4 with other cation channels or proteins present at the FA sites.


Asunto(s)
Estructuras de la Membrana Celular/metabolismo , Movimiento Celular , Canales Catiónicos TRPV/metabolismo , Sitios de Unión , Calpaína/metabolismo , Adhesión Celular , Línea Celular Tumoral , Células HEK293 , Humanos , Mutación , Fosfatidilinositoles/metabolismo , Unión Proteica , Canales Catiónicos TRPV/química , Canales Catiónicos TRPV/genética
6.
Proc Natl Acad Sci U S A ; 107(44): 19084-9, 2010 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-20956320

RESUMEN

Reduced functional bladder capacity and concomitant increased micturition frequency (pollakisuria) are common lower urinary tract symptoms associated with conditions such as cystitis, prostatic hyperplasia, neurological disease, and overactive bladder syndrome. These symptoms can profoundly affect the quality of life of afflicted individuals, but available pharmacological treatments are often unsatisfactory. Recent work has demonstrated that the cation channel TRPV4 is highly expressed in urothelial cells and plays a role in sensing the normal filling state of the bladder. In this article, we show that the development of cystitis-induced bladder dysfunction is strongly impaired in Trpv4(-/-) mice. Moreover, we describe HC-067047, a previously uncharacterized, potent, and selective TRPV4 antagonist that increases functional bladder capacity and reduces micturition frequency in WT mice and rats with cystitis. HC-067047 did not affect bladder function in Trpv4(-/-) mice, demonstrating that its in vivo effects are on target. These results indicate that TRPV4 antagonists may provide a promising means of treating bladder dysfunction.


Asunto(s)
Antineoplásicos Alquilantes/efectos adversos , Ciclofosfamida/efectos adversos , Cistitis , Moduladores del Transporte de Membrana/farmacología , Morfolinas/farmacología , Pirroles/farmacología , Canales Catiónicos TRPV/antagonistas & inhibidores , Vejiga Urinaria/fisiopatología , Urotelio/fisiopatología , Animales , Antineoplásicos Alquilantes/farmacología , Ciclofosfamida/farmacología , Cistitis/inducido químicamente , Cistitis/tratamiento farmacológico , Cistitis/metabolismo , Cistitis/fisiopatología , Humanos , Ratones , Ratones Noqueados , Ratas , Ratas Wistar , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Vejiga Urinaria/metabolismo , Micción/efectos de los fármacos , Urotelio/metabolismo
7.
Br J Neurosurg ; 25(4): 516-22, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21749185

RESUMEN

Spinal cord injury (SCI) occurs in the most productive part of life. Treatment options for treatment of chronic SCI are few and have limited impact on clinical outcome. Central nervous system (CNS) has limited intrinsic regeneration capability. The study included patients with chronic complete SCI. Previously harvested autologous mesenchymal stem cells were administered at the site of injury after a laminectomy. Follow-up was done by a neutral examiner not involved in the surgery every 3 months. One patient had improvement in motor power. Two patients had a patchy improvement in pin prick sensation below the level of injury. Three different, progressively increasing doses did not result in improvement in the clinical outcome. Though the administration of allogenic human mesenchymal stem cells is safe in patients with SCI, it may not be efficacious; especially in patients with chronic SCI.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas/métodos , Traumatismos de la Médula Espinal/terapia , Adulto , Enfermedad Crónica , Femenino , Estudios de Seguimiento , Humanos , Inyecciones Espinales , Masculino , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Persona de Mediana Edad , Proyectos Piloto , Recolección de Tejidos y Órganos , Trasplante Autólogo , Resultado del Tratamiento , Adulto Joven
8.
J Neurochem ; 110(3): 1070-81, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19500213

RESUMEN

4-Methylnitrosamino-1-(3-pyridyl)-1-butanone (NNK) is a tobacco-specific procarcinogen. We have investigated whether NNK causes inflammatory upheaval in the brain by activation of resident microglia and astrocyte and result in bystander neuronal damage. We have carried out the work in both in vitro and in vivo models. We have found that treatment with NNK causes significant activation of mouse microglial (BV2) cell line as evident by increase in reactive oxygen species and nitric oxide level. Western blot analysis has showed increase in proinflammatory signaling proteins, proinflammatory effector proteins, and other stress-related proteins. Interestingly, increased levels of proinflammatory cytokines like interleukin (IL)-6, tumor necrosis factor-alpha, monocyte chemoattractant protein 1 (MCP1), and IL-12p70 are also detected. Work from our in vivo studies has demonstrated similar increase in proinflammatory signaling and effector molecules along with the proinflammatory cytokine levels, following NNK treatment. Immunohistochemical staining of the brain sections of NNK-treated mice reveals massive microglial and astrocyte activation along with distinct foci of neuronal damage. Both in vitro and in vivo results provide strong indication that NNK causes significant upheaval of the inflammatory condition of brain and inflicts subsequent neuronal damage.


Asunto(s)
Carcinógenos/toxicidad , Microglía/efectos de los fármacos , Microglía/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Nicotiana/toxicidad , Animales , Línea Celular , Línea Celular Tumoral , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Microglía/patología , Neuronas/patología , Nitrosaminas/toxicidad
9.
Elife ; 82019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31184584

RESUMEN

The cation channel TRPA1 transduces a myriad of noxious chemical stimuli into nociceptor electrical excitation and neuropeptide release, leading to pain and neurogenic inflammation. Despite emergent evidence that TRPA1 is regulated by the membrane environment, it remains unknown whether this channel localizes in membrane microdomains or whether it interacts with cholesterol. Using total internal reflection fluorescence microscopy and density gradient centrifugation we found that mouse TRPA1 localizes preferably into cholesterol-rich domains and functional experiments revealed that cholesterol depletion decreases channel sensitivity to chemical agonists. Moreover, we identified two structural motifs in transmembrane segments 2 and 4 involved in mTRPA1-cholesterol interactions that are necessary for normal agonist sensitivity and plasma membrane localization. We discuss the impact of such interactions on TRPA1 gating mechanisms, regulation by the lipid environment, and role of this channel in sensory membrane microdomains, all of which helps to understand the puzzling pharmacology and pathophysiology of this channel.


Asunto(s)
Membrana Celular/metabolismo , Colesterol/metabolismo , Canal Catiónico TRPA1/metabolismo , Secuencia de Aminoácidos , Animales , Células CHO , Colesterol/química , Cricetinae , Cricetulus , Células HEK293 , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Microdominios de Membrana/metabolismo , Ratones , Microscopía Fluorescente/métodos , Modelos Moleculares , Unión Proteica , Dominios Proteicos , Homología de Secuencia de Aminoácido , Canal Catiónico TRPA1/química , Canal Catiónico TRPA1/genética , Proteína Fluorescente Roja
10.
J Clin Invest ; 129(8): 3140-3152, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31162142

RESUMEN

Elevated blood glucose (hyperglycemia) is a hallmark metabolic abnormality in diabetes. Hyperglycemia is associated with protein kinase A (PKA)-mediated stimulation of L-type Ca2+ channels in arterial myocytes resulting in increased vasoconstriction. However, the mechanisms by which glucose activates PKA remain unclear. Here, we showed that elevating extracellular glucose stimulates cAMP production in arterial myocytes, and that this was specifically dependent on adenylyl cyclase 5 (AC5) activity. Super-resolution imaging suggested nanometer proximity between subpopulations of AC5 and the L-type Ca2+ channel pore-forming subunit CaV1.2. In vitro, in silico, ex vivo and in vivo experiments revealed that this close association is critical for stimulation of L-type Ca2+ channels in arterial myocytes and increased myogenic tone upon acute hyperglycemia. This pathway supported the increase in L-type Ca2+ channel activity and myogenic tone in two animal models of diabetes. Our collective findings demonstrate a unique role for AC5 in PKA-dependent modulation of L-type Ca2+ channel activity and vascular reactivity during acute hyperglycemia and diabetes.


Asunto(s)
Adenilil Ciclasas/metabolismo , Arterias Cerebrales/enzimología , AMP Cíclico/metabolismo , Diabetes Mellitus Experimental/enzimología , Hiperglucemia/enzimología , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Adenilil Ciclasas/genética , Animales , Canales de Calcio Tipo L/biosíntesis , Canales de Calcio Tipo L/genética , Arterias Cerebrales/patología , AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Hiperglucemia/genética , Hiperglucemia/patología , Ratones , Ratones Noqueados , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología
11.
Elife ; 82019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30821687

RESUMEN

Elevated glucose increases vascular reactivity by promoting L-type CaV1.2 channel (LTCC) activity by protein kinase A (PKA). Yet, how glucose activates PKA is unknown. We hypothesized that a Gs-coupled P2Y receptor is an upstream activator of PKA mediating LTCC potentiation during diabetic hyperglycemia. Experiments in apyrase-treated cells suggested involvement of a P2Y receptor underlying the glucose effects on LTTCs. Using human tissue, expression for P2Y11, the only Gs-coupled P2Y receptor, was detected in nanometer proximity to CaV1.2 and PKA. FRET-based experiments revealed that the selective P2Y11 agonist NF546 and elevated glucose stimulate cAMP production resulting in enhanced PKA-dependent LTCC activity. These changes were blocked by the selective P2Y11 inhibitor NF340. Comparable results were observed in mouse tissue, suggesting that a P2Y11-like receptor is mediating the glucose response in these cells. These findings established a key role for P2Y11 in regulating PKA-dependent LTCC function and vascular reactivity during diabetic hyperglycemia.


Asunto(s)
Vasos Sanguíneos/fisiopatología , Calcio/metabolismo , Hiperglucemia , Contracción Muscular , Receptores Acoplados a Proteínas G/metabolismo , Receptores Purinérgicos/metabolismo , Animales , Señalización del Calcio , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ratones Endogámicos C57BL
12.
Drug Discov Today ; 13(13-14): 619-24, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18598919

RESUMEN

Flaviviruses are a group of arboviruses under the family of Flaviviridae. Flaviviruses are global pathogens, some of which cause neurotrophic disorders from mild febrile illness to lethal encephalitis while others cause severe hemorrhagic fever. These viruses are endemic to many parts of the world and periodic outbreaks take thousands of lives. With globalization of the world accompanied by gradual shift in global climate, the viruses are emerging in newer areas and we are in a point of exigency to decipher new drugs to combat these emerging threats. Although extensive research has taken place and vaccines for few of these viral diseases are available, still no chemotherapeutic agent has been found to deliver an absolute cure to flaviviral infections. It is important to assess the present status of flavivirus drug research and delve deep into the avenues of flavivirus drug discovery. This review outlines the chemotherapeutic agents explored till date, and highlights perspectives on a possible future breakthrough - at least to ameliorate if not to abrogate the diseases.


Asunto(s)
Antivirales/uso terapéutico , Infecciones por Flavivirus/tratamiento farmacológico , Flavivirus/patogenicidad , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Apoptosis/efectos de los fármacos , Flavivirus/efectos de los fármacos , Infecciones por Flavivirus/patología , Infecciones por Flavivirus/virología , Humanos
13.
Sci Rep ; 7(1): 14058, 2017 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-29070899

RESUMEN

Large-conductance Ca2+-activated potassium (BKCa) channels are key determinants of vascular smooth muscle excitability. Impaired BKCa channel function through remodeling of BKCa ß1 expression and function contributes to vascular complications in animal models of diabetes. Yet, whether similar alterations occur in native vascular smooth muscle from humans with type 2 diabetes is unclear. In this study, we evaluated BKCa function in vascular smooth muscle from small resistance adipose arteries of non-diabetic and clinically diagnosed type 2 diabetic patients. We found that BKCa channel activity opposes pressure-induced constriction in human small resistance adipose arteries, and this is compromised in arteries from diabetic patients. Consistent with impairment of BKCa channel function, the amplitude and frequency of spontaneous BKCa currents, but not Ca2+ sparks were lower in cells from diabetic patients. BKCa channels in diabetic cells exhibited reduced Ca2+ sensitivity, single-channel open probability and tamoxifen sensitivity. These effects were associated with decreased functional coupling between BKCa α and ß1 subunits, but no change in total protein abundance. Overall, results suggest impairment in BKCa channel function in vascular smooth muscle from diabetic patients through unique mechanisms, which may contribute to vascular complications in humans with type 2 diabetes.


Asunto(s)
Arterias/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Potenciales de Acción , Arterias/fisiopatología , Estudios de Casos y Controles , Células Cultivadas , Humanos , Potenciales de la Membrana
14.
Elife ; 52016 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-27449282

RESUMEN

Various TRP channels act as polymodal sensors of thermal and chemical stimuli, but the mechanisms whereby chemical ligands impact on TRP channel gating are poorly understood. Here we show that AITC (allyl isothiocyanate; mustard oil) and menthol represent two distinct types of ligands at the mammalian cold sensor TRPM8. Kinetic analysis of channel gating revealed that AITC acts by destabilizing the closed channel, whereas menthol stabilizes the open channel, relative to the transition state. Based on these differences, we classify agonists as either type I (menthol-like) or type II (AITC-like), and provide a kinetic model that faithfully reproduces their differential effects. We further demonstrate that type I and type II agonists have a distinct impact on TRPM8 currents and TRPM8-mediated calcium signals in excitable cells. These findings provide a theoretical framework for understanding the differential actions of TRP channel ligands, with important ramifications for TRP channel structure-function analysis and pharmacology.


Asunto(s)
Isotiocianatos/metabolismo , Mentol/metabolismo , Canales Catiónicos TRPM/agonistas , Canales Catiónicos TRPM/metabolismo , Animales , Señalización del Calcio , Cinética , Ratones , Técnicas de Placa-Clamp
15.
Nat Commun ; 7: 10489, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-26843440

RESUMEN

The cation channel TRPM8 plays a central role in the somatosensory system, as a key sensor of innocuously cold temperatures and cooling agents. Although increased functional expression of TRPM8 has been implicated in various forms of pathological cold hypersensitivity, little is known about the cellular and molecular mechanisms that determine TRPM8 abundance at the plasma membrane. Here we demonstrate constitutive transport of TRPM8 towards the plasma membrane in atypical, non-acidic transport vesicles that contain lysosomal-associated membrane protein 1 (LAMP1), and provide evidence that vesicle-associated membrane protein 7 (VAMP7) mediates fusion of these vesicles with the plasma membrane. In line herewith, VAMP7-deficient mice exhibit reduced functional expression of TRPM8 in sensory neurons and concomitant deficits in cold avoidance and icilin-induced cold hypersensitivity. Our results uncover a cellular pathway that controls functional plasma membrane incorporation of a temperature-sensitive TRP channel, and thus regulates thermosensitivity in vivo.


Asunto(s)
Membrana Celular/metabolismo , Frío , Hiperestesia/genética , Proteínas R-SNARE/genética , Células Receptoras Sensoriales/metabolismo , Canales Catiónicos TRPM/metabolismo , Vesículas Transportadoras/metabolismo , Animales , Calcio/metabolismo , Femenino , Ganglios Espinales/metabolismo , Células HEK293 , Humanos , Hiperestesia/inducido químicamente , Hiperestesia/metabolismo , Proteínas de Membrana de los Lisosomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Técnicas de Placa-Clamp , Pirimidinonas/toxicidad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ganglio del Trigémino/metabolismo
16.
Temperature (Austin) ; 2(2): 163-5, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27227018

RESUMEN

TRP ion channels are ubiquitously present in the mammalian body and take part in numerous key physiological functions, including temperature sensing, taste perception, osmo-regulation, cardiac function, renal function, development, and glucose homeostasis. The mechanisms whereby TRP channels are transported to the plasma membrane, where most of them exert their physiological actions, remains a poorly understood aspect of TRP channel biology.

17.
J Gen Physiol ; 146(1): 51-63, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26123194

RESUMEN

The transient receptor potential (TRP) channel TRPM3 is a calcium-permeable cation channel activated by heat and by the neurosteroid pregnenolone sulfate (PregS). TRPM3 is highly expressed in sensory neurons, where it plays a key role in heat sensing and inflammatory hyperalgesia, and in pancreatic ß cells, where its activation enhances glucose-induced insulin release. However, despite its functional importance, little is known about the cellular mechanisms that regulate TRPM3 activity. Here, we provide evidence for a dynamic regulation of TRPM3 by membrane phosphatidylinositol phosphates (PIPs). Phosphatidylinositol 4,5-bisphosphate (PI[4,5]P2) and ATP applied to the intracellular side of excised membrane patches promote recovery of TRPM3 from desensitization. The stimulatory effect of cytosolic ATP on TRPM3 reflects activation of phosphatidylinositol kinases (PI-Ks), leading to resynthesis of PIPs in the plasma membrane. Various PIPs directly enhance TRPM3 activity in cell-free inside-out patches, with a potency order PI(3,4,5)P3 > PI(3,5)P2 > PI(4,5)P2 ≈ PI(3,4)P2 >> PI(4)P. Conversely, TRPM3 activity is rapidly and reversibly inhibited by activation of phosphatases that remove the 5-phosphate from PIPs. Finally, we show that recombinant TRPM3, as well as the endogenous TRPM3 in insuloma cells, is rapidly and reversibly inhibited by activation of phospholipase C-coupled muscarinic acetylcholine receptors. Our results reveal basic cellular mechanisms whereby membrane receptors can regulate TRPM3 activity.


Asunto(s)
Fosfatidilinositoles/metabolismo , Canales Catiónicos TRPM/metabolismo , Adenosina Trifosfato/metabolismo , Línea Celular , Membrana Celular/metabolismo , Células HEK293 , Humanos , Hiperalgesia/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Pregnenolona/metabolismo , Células Receptoras Sensoriales/metabolismo
18.
Sci Rep ; 4: 7111, 2014 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-25407951

RESUMEN

Transient Receptor Potential (TRP) channels form a broadly expressed and functionally diverse family of cation channels involved in various (patho)physiological processes. Whereas the mechanisms that control opening of TRP channels have been extensively studied, little is known about the transport processes of TRP channels to and within the plasma membrane. Here we used Total Internal Reflection--Fluorescence Recovery after Photobleaching (TIR-FRAP) to selectively visualize and bleach the fluorescently labeled TRP channels TRPV2 and TRPM4 in close proximity of the glass-plasma membrane interface, allowing detailed analysis of their perimembrane dynamics. We show that recovery of TRPM4 occurs via 200-nm diameter transport vesicles, and demonstrate the full fusion of such vesicles with the plasma membrane. In contrast, TRPV2 recovery proceeded mainly via lateral diffusion from non-bleached areas of the plasma membrane. Analysis of the two-dimensional channel diffusion kinetics yielded 2D diffusion coefficients ranging between 0.1 and 0.3 µm(2)/s, suggesting that these TRP channels move relatively unrestricted within the plasma membrane. These data demonstrate distinct modes of TRP channel turnover at the plasma membrane and illustrate the usefulness of TIR-FRAP to monitor these processes with high resolution.


Asunto(s)
Proteínas Fluorescentes Verdes/metabolismo , Canales Catiónicos TRPM/metabolismo , Canales Catiónicos TRPV/metabolismo , Vesículas Transportadoras/metabolismo , Recuperación de Fluorescencia tras Fotoblanqueo , Expresión Génica , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Humanos , Microscopía Fluorescente/instrumentación , Microscopía Fluorescente/métodos , Plásmidos/química , Plásmidos/metabolismo , Transporte de Proteínas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPV/genética , Transfección , Vesículas Transportadoras/ultraestructura
20.
PLoS One ; 5(4): e9984, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20376308

RESUMEN

BACKGROUND: Benzo[a]pyrene (B[a]P) belongs to a class of polycyclic aromatic hydrocarbons that serve as micropollutants in the environment. B[a]P has been reported as a probable carcinogen in humans. Exposure to B[a]P can take place by ingestion of contaminated (especially grilled, roasted or smoked) food or water, or inhalation of polluted air. There are reports available that also suggests neurotoxicity as a result of B[a]P exposure, but the exact mechanism of action is unknown. METHODOLOGY/PRINCIPAL FINDINGS: Using neuroblastoma cell line and primary cortical neuron culture, we demonstrated that B[a]P has no direct neurotoxic effect. We utilized both in vivo and in vitro systems to demonstrate that B[a]P causes microglial activation. Using microglial cell line and primary microglial culture, we showed for the first time that B[a]P administration results in elevation of reactive oxygen species within the microglia thereby causing depression of antioxidant protein levels; enhanced expression of inducible nitric oxide synthase, that results in increased production of NO from the cells. Synthesis and secretion of proinflammatory cytokines were also elevated within the microglia, possibly via the p38MAP kinase pathway. All these factors contributed to bystander death of neurons, in vitro. When administered to animals, B[a]P was found to cause microglial activation and astrogliosis in the brain with subsequent increase in proinflammatory cytokine levels. CONCLUSIONS/SIGNIFICANCE: Contrary to earlier published reports we found that B[a]P has no direct neurotoxic activity. However, it kills neurons in a bystander mechanism by activating the immune cells of the brain viz the microglia. For the first time, we have provided conclusive evidence regarding the mechanism by which the micropollutant B[a]P may actually cause damage to the central nervous system. In today's perspective, where rising pollution levels globally are a matter of grave concern, our study throws light on other health hazards that such pollutants may exert.


Asunto(s)
Benzo(a)pireno/farmacología , Carcinógenos/farmacología , Microglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Benzo(a)pireno/toxicidad , Carcinógenos/toxicidad , Muerte Celular/efectos de los fármacos , Línea Celular , Células Cultivadas , Citocinas/biosíntesis , Citocinas/metabolismo , Contaminantes Ambientales/farmacología , Contaminantes Ambientales/toxicidad , Mediadores de Inflamación , Ratones , Microglía/metabolismo , Neuroblastoma/patología , Neuronas/citología , Óxido Nítrico/biosíntesis , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA