Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
PLoS One ; 19(3): e0300150, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38457438

RESUMEN

During hypoxia accumulation of lactate may be a key factor in acidosis-induced tissue damage. Binding of hexokinase (HK) to the outer membrane of mitochondria may have a protective effect under these conditions. We have investigated the regulation of lactate metabolism by hexokinases (HKs), using HEK293 cells in which the endogenous hexokinases have been knocked down to enable overexpression of wild type and mutant HKs. To assess the real-time changes in intracellular lactate levels the cells were also transfected with a lactate specific FRET probe. In the HKI/HKII double knockdown HEK cells, addition of extracellular pyruvate caused a large and sustained decrease in lactate. Upon inhibition of the mitochondrial electron transfer chain by NaCN this effect was reversed as a rapid increase in lactate developed which was followed by a slow and sustained increase in the continued presence of the inhibitor. Incubation of the HKI/HKII double knockdown HEK cells with the inhibitor of the malic enzyme, ME1*, blocked the delayed accumulation of lactate evoked by NaCN. With replacement by overexpression of HKI or HKII the accumulation of intracellular lactate evoked by NaCN was prevented. Blockage of the pentose phosphate pathway with the inhibitor 6-aminonicotinamide (6-AN) abolished the protective effect of HK expression, with NaCN causing again a sustained increase in lactate. The effect of HK was dependent on HK's catalytic activity and interaction with the mitochondrial outer membrane (MOM). Based on these data we propose that transformation of glucose into G6P by HK activates the pentose phosphate pathway which increases the production of NADPH, which then blocks the activity of the malic enzyme to transform malate into pyruvate and lactate.


Asunto(s)
Hexoquinasa , Ácido Láctico , Humanos , Hexoquinasa/genética , Hexoquinasa/metabolismo , Ácido Láctico/metabolismo , Células HEK293 , Mitocondrias/metabolismo , Piruvatos/metabolismo
2.
PLoS One ; 18(11): e0286660, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37917627

RESUMEN

Lactate is a mitochondrial substrate for many tissues including neuron, muscle, skeletal and cardiac, as well as many cancer cells, however little is known about the processes that regulate its utilization in mitochondria. Based on the close association of Hexokinases (HK) with mitochondria, and the known cardio-protective role of HK in cardiac muscle, we have investigated the regulation of lactate and pyruvate metabolism by hexokinases (HKs), utilizing wild-type HEK293 cells and HEK293 cells in which the endogenous HKI and/or HKII have been knocked down to enable overexpression of wild type and mutant HKs. To assess the real-time changes in intracellular lactate levels the cells were transfected with a lactate specific FRET probe. In the HKI/HKII double knockdown cells, addition of extracellular pyruvate caused a large and sustained decrease in lactate. This decrease was rapidly reversed upon inhibition of the malate aspartate shuttle by aminooxyacetate, or inhibition of mitochondrial oxidative respiration by NaCN. These results suggest that in the absence of HKs, pyruvate-dependent activation of the TCA cycle together with the malate aspartate shuttle facilitates lactate transformation into pyruvate and its utilization by mitochondria. With replacement by overexpression of HKI or HKII the cellular response to pyruvate and NaCN was modified. With either hexokinase present, both the decrease in lactate due to the addition of pyruvate and the increase following addition of NaCN were either transient or suppressed altogether. Blockage of the pentose phosphate pathway with the inhibitor 6-aminonicotinamide (6-AN), abolished the effects of HK replacement. These results suggest that blocking of the malate aspartate shuttle by HK may involve activation of the pentose phosphate pathway and increased NADPH production.


Asunto(s)
Ácido Láctico , Ácido Pirúvico , Humanos , Hexoquinasa/metabolismo , Malatos/metabolismo , Ácido Aspártico/metabolismo , Células HEK293
3.
Proc Natl Acad Sci U S A ; 117(18): 9857-9864, 2020 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-32300017

RESUMEN

Vitamin A has diverse biological functions and is essential for human survival at every point from embryogenesis to adulthood. Vitamin A and its derivatives have been used to treat human diseases including vision diseases, skin diseases, and cancer. Both insufficient and excessive vitamin A uptake are detrimental, but how its transport is regulated is poorly understood. STRA6 is a multitransmembrane domain cell-surface receptor and mediates vitamin A uptake from plasma retinol binding protein (RBP). STRA6 can mediate both cellular vitamin A influx and efflux, but what regulates these opposing activities is unknown. To answer this question, we purified and identified STRA6-associated proteins in a native mammalian cell type that takes up vitamin A through STRA6 using mass spectrometry. We found that the major protein repeatedly identified as STRA6-associated protein is calmodulin, consistent with the cryogenic electron microscopy (cryo-EM) study of zebrafish STRA6 associated with calmodulin. Using radioactivity-based, high-performance liquid chromatography (HPLC)-based and real-time fluorescence techniques, we found that calmodulin profoundly affects STRA6's vitamin A transport activity. Increased calcium/calmodulin promotes cellular vitamin A efflux and suppresses vitamin A influx through STRA6. Further mechanistic studies revealed that calmodulin enhances the binding of apo-RBP to STRA6, and this enhancement is much more pronounced for apo-RBP than holo-RBP. This study revealed that calmodulin regulates STRA6's vitamin A influx or efflux activity by modulating its preferential interaction with apo-RBP or holo-RBP. This molecular mechanism of regulating vitamin A transport may point to new directions to treat human diseases associated with insufficient or excessive vitamin A uptake.


Asunto(s)
Transporte Biológico/genética , Calmodulina/genética , Proteínas de la Membrana/genética , Proteínas Plasmáticas de Unión al Retinol/genética , Vitamina A/metabolismo , Animales , Apoproteínas/genética , Apoproteínas/metabolismo , Calcio/metabolismo , Bovinos , Línea Celular , Cromatografía Líquida de Alta Presión , Microscopía por Crioelectrón , Humanos , Proteínas de la Membrana/metabolismo , Unión Proteica/genética , Receptores de Superficie Celular/genética , Proteínas Plasmáticas de Unión al Retinol/metabolismo , Vitamina A/genética , Pez Cebra/genética
4.
Nat Commun ; 10(1): 5444, 2019 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-31784510

RESUMEN

Elevated glucose consumption is fundamental to cancer, but selectively targeting this pathway is challenging. We develop a high-throughput assay for measuring glucose consumption and use it to screen non-small-cell lung cancer cell lines against bioactive small molecules. We identify Milciclib that blocks glucose consumption in H460 and H1975, but not in HCC827 or A549 cells, by decreasing SLC2A1 (GLUT1) mRNA and protein levels and by inhibiting glucose transport. Milciclib blocks glucose consumption by targeting cyclin-dependent kinase 7 (CDK7) similar to other CDK7 inhibitors including THZ1 and LDC4297. Enhanced PIK3CA signaling leads to CDK7 phosphorylation, which promotes RNA Polymerase II phosphorylation and transcription. Milciclib, THZ1, and LDC4297 lead to a reduction in RNA Polymerase II phosphorylation on the SLC2A1 promoter. These data indicate that our high-throughput assay can identify compounds that regulate glucose consumption and that CDK7 is a key regulator of glucose consumption in cells with an activated PI3K pathway.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Transportador de Glucosa de Tipo 1/efectos de los fármacos , Glucosa/metabolismo , Neoplasias Pulmonares/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Células A549 , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I/efectos de los fármacos , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Fenilendiaminas/farmacología , Fosforilación , Pirazoles/farmacología , Pirimidinas/farmacología , Quinazolinas/farmacología , ARN Polimerasa II/efectos de los fármacos , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal , Triazinas/farmacología , Quinasa Activadora de Quinasas Ciclina-Dependientes
5.
Elife ; 62017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29231167

RESUMEN

The heart switches its energy substrate from glucose to fatty acids at birth, and maternal hyperglycemia is associated with congenital heart disease. However, little is known about how blood glucose impacts heart formation. Using a chemically defined human pluripotent stem-cell-derived cardiomyocyte differentiation system, we found that high glucose inhibits the maturation of cardiomyocytes at genetic, structural, metabolic, electrophysiological, and biomechanical levels by promoting nucleotide biosynthesis through the pentose phosphate pathway. Blood glucose level in embryos is stable in utero during normal pregnancy, but glucose uptake by fetal cardiac tissue is drastically reduced in late gestational stages. In a murine model of diabetic pregnancy, fetal hearts showed cardiomyopathy with increased mitotic activity and decreased maturity. These data suggest that high glucose suppresses cardiac maturation, providing a possible mechanistic basis for congenital heart disease in diabetic pregnancy.


Asunto(s)
Células Madre Embrionarias/citología , Glucosa/farmacología , Desarrollo de Músculos/efectos de los fármacos , Miocardio/citología , Miocitos Cardíacos/citología , Nucleótidos/biosíntesis , Animales , Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Vía de Pentosa Fosfato , Embarazo , Edulcorantes/farmacología
6.
Physiol Rep ; 5(3)2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28193781

RESUMEN

The processes controlling targeting of glucose transporters to apical and basolateral membranes of polarized cells are complex and not-well understood. We have engineered SGLT1 and GLUT4 constructs linked to fluorescent proteins to highlight the differences in transporter expression and trafficking, in real time, in different cell types. Activity was assessed in parallel using a FRET glucose sensor. In COS cells and HEK cells, SGLT1 was distributed between the plasma membrane and intracellular compartments, but there was little expression in CHO cells. Trafficking was investigated using the lysosome inhibitors NH4Cl (10 mmol/L) and chloroquine (150 µmol/L) and the proteasome inhibitors MG-262 (1 µmol/L) and lactacystin (5 µmol/L). Lysosome inhibitors caused SGLT1 accumulation into intracellular bodies, whereas proteasome inhibitors induced SGLT1 accumulation in the plasma membrane, even in CHO cells. Our data suggest that a fraction of SGLT1 is rapidly degraded by lysosomes and never reached the plasma membrane; another fraction reaches the membrane and is subsequently degraded by lysosomes following internalization. The latter process is regulated by the ubiquitin/proteasome pathway, acting at a late stage of the lysosomal pathway. Using the cholesterol inhibitor MßCD (3 mmol/L), a dominant negative dynamin (K44A) and caveolin, we showed that SGLT1 internalization is lipid raft-mediated, but caveolin-independent. In contrast, GLUT4 internalization is dynamin-dependent, but cholesterol-independent. The physiological relevance of these data is discussed in terms of differential membrane compartmentalization of the transporters and expression under stress conditions.


Asunto(s)
Microdominios de Membrana/metabolismo , Proteolisis , Transportador 1 de Sodio-Glucosa/metabolismo , Animales , Células CHO , Células COS , Polaridad Celular , Chlorocebus aethiops , Cricetulus , Transportador de Glucosa de Tipo 4/metabolismo , Células HEK293 , Humanos , Proteínas Luminiscentes , Lisosomas/metabolismo , Microscopía Fluorescente/métodos , Complejo de la Endopetidasa Proteasomal/metabolismo , Transporte de Proteínas
7.
J Mol Cell Cardiol ; 78: 107-15, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25264175

RESUMEN

As mediators of the first enzymatic step in glucose metabolism, hexokinases (HKs) orchestrate a variety of catabolic and anabolic uses of glucose, regulate antioxidant power by generating NADPH for glutathione reduction, and modulate cell death processes by directly interacting with the voltage-dependent anion channel (VDAC), a regulatory component of the mitochondrial permeability transition pore (mPTP). Here we summarize the current state-of-knowledge about HKs and their role in protecting the heart from ischemia/reperfusion (I/R) injury, reviewing: 1) the properties of different HK isoforms and how their function is regulated by their subcellular localization; 2) how HKs modulate glucose metabolism and energy production during I/R; 3) the molecular mechanisms by which HKs influence mPTP opening and cellular injury during I/R; and 4) how different metabolic and HK profiles correlate with susceptibility to I/R injury and cardioprotective efficacy in cancer cells, neonatal hearts, and normal, hypertrophied and failing adult hearts, and how these difference may guide novel therapeutic strategies to limit I/R injury in the heart. This article is part of a Special Issue entitled "Mitochondria: From Basic Mitochondrial Biology to Cardiovascular Disease".


Asunto(s)
Hexoquinasa/metabolismo , Miocardio/metabolismo , Animales , Glucosa/metabolismo , Cardiopatías/metabolismo , Humanos , Isoenzimas , Metaboloma , Mitocondrias Cardíacas/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Daño por Reperfusión Miocárdica/metabolismo , Neoplasias/metabolismo
8.
J Immunol ; 192(1): 110-22, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24307733

RESUMEN

Orai1 is the pore subunit of Ca(2+) release-activated Ca(2+) (CRAC) channels that stimulate downstream signaling pathways crucial for T cell activation. CRAC channels are an attractive therapeutic target for alleviation of autoimmune diseases. Using high-throughput chemical library screening targeting Orai1, we identified a novel class of small molecules that inhibit CRAC channel activity. One of these molecules, compound 5D, inhibited CRAC channel activity by blocking ion permeation. When included during differentiation, Th17 cells showed higher sensitivity to compound 5D than Th1 and Th2 cells. The selectivity was attributable to high dependence of promoters of retinoic-acid-receptor-related orphan receptors on the Ca(2+)-NFAT pathway. Blocking of CRAC channels drastically decreased recruitment of NFAT and histone modifications within key gene loci involved in Th17 differentiation. The impairment in Th17 differentiation by treatment with CRAC channel blocker was recapitulated in Orai1-deficient T cells, which could be rescued by exogenous expression of retinoic-acid-receptor-related orphan receptors or a constitutive active mutant of NFAT. In vivo administration of CRAC channel blockers effectively reduced the severity of experimental autoimmune encephalomyelitis by suppression of differentiation of inflammatory T cells. These results suggest that CRAC channel blockers can be considered as chemical templates for the development of therapeutic agents to suppress inflammatory responses.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio , Receptores Nucleares Huérfanos/metabolismo , Células Th17/citología , Células Th17/metabolismo , Animales , Bloqueadores de los Canales de Calcio/química , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/genética , Diferenciación Celular/efectos de los fármacos , Línea Celular , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Humanos , Iones/metabolismo , Ratones , Factores de Transcripción NFATC/metabolismo , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Proteína ORAI1 , Receptores Nucleares Huérfanos/deficiencia , Receptores Nucleares Huérfanos/genética , Regiones Promotoras Genéticas , Unión Proteica , Elementos de Respuesta , Bibliotecas de Moléculas Pequeñas , Células TH1/citología , Células TH1/inmunología , Células TH1/metabolismo , Células Th17/inmunología , Células Th2/citología , Células Th2/inmunología , Células Th2/metabolismo
9.
J Gen Physiol ; 142(4): 425-36, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24081983

RESUMEN

In mammalian tumor cell lines, localization of hexokinase (HK) isoforms to the cytoplasm or mitochondria has been shown to control their anabolic (glycogen synthesis) and catabolic (glycolysis) activities. In this study, we examined whether HK isoform differences could explain the markedly different metabolic profiles between normal adult and neonatal cardiac tissue. We used a set of novel genetically encoded optical imaging tools to track, in real-time in isolated adult (ARVM) and neonatal (NRVM) rat ventricular myocytes, the subcellular distributions of HKI and HKII, and the functional consequences on glucose utilization. We show that HKII, the predominant isoform in ARVM, dynamically translocates from mitochondria and cytoplasm in response to removal of extracellular glucose or addition of iodoacetate (IAA). In contrast, HKI, the predominant isoform in NRVM, is only bound to mitochondria and is not displaced by the above interventions. In ARVM, overexpression of HKI, but not HKII, increased glycolytic activity. In neonatal rat ventricular myocytes (NVRM), knockdown of HKI, but not HKII, decreased glycolytic activity. In conclusion, differential interactions of HKI and HKII with mitochondria underlie the different metabolic profiles of ARVM and NRVM, accounting for the markedly increased glycolytic activity of NRVM.


Asunto(s)
Glucosa/metabolismo , Ventrículos Cardíacos/crecimiento & desarrollo , Hexoquinasa/metabolismo , Mitocondrias/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Citoplasma/metabolismo , Glucólisis , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/metabolismo , Isoenzimas/metabolismo , Miocitos Cardíacos/enzimología , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley
10.
Channels (Austin) ; 7(5): 354-63, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23454861

RESUMEN

Store-operated Ca(2+) entry (SOCE) is a widespread mechanism to elevate the intracellular Ca(2+) concentrations and stimulate downstream signaling pathways affecting proliferation, secretion, differentiation and death in different cell types. In immune cells, immune receptor stimulation induces intracellular Ca(2+) store depletion that subsequently activates Ca(2+)-release-activated-Ca(2+) (CRAC) channels, a prototype of store-operated Ca(2+) (SOC) channels. CRAC channel opening leads to activation of diverse downstream signaling pathways affecting proliferation, differentiation, cytokine production and cell death. Recent identification of STIM1 as the endoplasmic reticulum Ca(2+) sensor and Orai1 as the pore subunit of CRAC channels has provided the much-needed molecular tools to dissect the mechanism of activation and regulation of CRAC channels. In this review, we discuss the recent advances in understanding the associating partners and posttranslational modifications of Orai1 and STIM1 proteins that regulate diverse aspects of CRAC channel function.


Asunto(s)
Canales de Calcio/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Animales , Humanos , Activación del Canal Iónico , Proteína ORAI1 , Procesamiento Proteico-Postraduccional , Molécula de Interacción Estromal 1
11.
J Biol Chem ; 286(40): 35318-28, 2011 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-21865174

RESUMEN

Orai1, the pore subunit of Ca(2+) release-activated Ca(2+) channels, has four transmembrane segments (TMs). The first segment, TMI, lines the pore and plays an important role in channel activation and ion permeation. TMIII, on the other hand, does not line the pore but still regulates channel gating and permeation properties. To understand the role of TMIII, we have mutated and characterized several residues in this domain. Mutation of Trp-176 to Cys (W176C) and Gly-183 to Ala (G183A) had dramatic effects. Unlike wild-type channels, which exhibit little outward current and are activated by STIM1, W176C mutant channels exhibited a large outward current at positive potentials and were constitutively active in the absence of STIM1. G183A mutant channels also exhibited substantial outward currents but were active only in the presence of 2-aminoethoxydiphenyl borate (2-APB), irrespective of STIM1. With W176C mutant channels inward, monovalent currents were blocked by Ca(2+) with a high affinity similar to the wild type, but the Ca(2+)-dependent blocking of outward currents differed in the two cases. Although a 50% block of the WT outward current required 250 µm Ca(2+), more than 6 mm was necessary to have the same effect on W176C mutant channels. In the presence of extracellular Ca(2+), W176C and G183A outward currents developed slowly in a voltage-dependent manner, whereas they developed almost instantaneously in the absence of Ca(2+). These changes in permeation and gating properties mimic the changes induced by mutations of Glu-190 in TMIII and Asp-110/Asp-112 in the TMI/TMII loop. On the basis of these data, we propose that TMIII maintains negatively charged residues at or near the selectivity filter in a conformation that facilitates Ca(2+) inward currents and prevents outward currents of monovalent cations. In addition, to controlling selectivity, TMIII may also stabilize channel gating in a closed state in the absence of STIM1 in a Trp-176-dependent manner.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Factores de Transcripción NFATC/metabolismo , Secuencia de Aminoácidos , Animales , Señalización del Calcio , Membrana Celular/metabolismo , Humanos , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Mutación , Proteína ORAI1 , Técnicas de Placa-Clamp , Homología de Secuencia de Aminoácido , Transducción de Señal
12.
PLoS One ; 6(3): e17674, 2011 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-21408025

RESUMEN

BACKGROUND: The first step in glucose metabolism is conversion of glucose to glucose 6-phosphate (G-6-P) by hexokinases (HKs), a family with 4 isoforms. The two most common isoforms, HKI and HKII, have overlapping tissue expression, but different subcellular distributions, with HKI associated mainly with mitochondria and HKII associated with both mitochondrial and cytoplasmic compartments. Here we tested the hypothesis that these different subcellular distributions are associated with different metabolic roles, with mitochondrially-bound HK's channeling G-6-P towards glycolysis (catabolic use), and cytoplasmic HKII regulating glycogen formation (anabolic use). METHODOLOGY/PRINCIPAL FINDINGS: To study subcellular translocation of HKs in living cells, we expressed HKI and HKII linked to YFP in CHO cells. We concomitantly recorded the effects on glucose handling using the FRET based intracellular glucose biosensor, FLIPglu-600 mM, and glycogen formation using a glycogen-associated protein, PTG, tagged with GFP. Our results demonstrate that HKI remains strongly bound to mitochondria, whereas HKII translocates between mitochondria and the cytosol in response to glucose, G-6-P and Akt, but not ATP. Metabolic measurements suggest that HKI exclusively promotes glycolysis, whereas HKII has a more complex role, promoting glycolysis when bound to mitochondria and glycogen synthesis when located in the cytosol. Glycogen breakdown upon glucose removal leads to HKII inhibition and dissociation from mitochondria, probably mediated by increases in glycogen-derived G-6-P. CONCLUSIONS/SIGNIFICANCE: These findings show that the catabolic versus anabolic fate of glucose is dynamically regulated by extracellular glucose via signaling molecules such as intracellular glucose, G-6-P and Akt through regulation and subcellular translocation of HKII. In contrast, HKI, which activity and regulation is much less sensitive to these factors, is mainly committed to glycolysis. This may be an important mechanism by which HK's allow cells to adapt to changing metabolic conditions to maintain energy balance and avoid injury.


Asunto(s)
Hexoquinasa/metabolismo , Animales , Células CHO , Carbonil Cianuro p-Trifluorometoxifenil Hidrazona/farmacología , Permeabilidad de la Membrana Celular/efectos de los fármacos , Cricetinae , Cricetulus , Citosol/efectos de los fármacos , Citosol/enzimología , Glucosa/metabolismo , Glucosa/farmacología , Transportador de Glucosa de Tipo 1/metabolismo , Glucosa-6-Fosfato/metabolismo , Glucógeno/metabolismo , Hexoquinasa/antagonistas & inhibidores , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/enzimología , Factores de Tiempo , Transfección
13.
Proc Natl Acad Sci U S A ; 108(4): 1699-704, 2011 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-21209335

RESUMEN

Cytoplasmic Ca(2+) is known to regulate Na(+)-Ca(2+) exchanger (NCX) activity by binding to two adjacent Ca(2+)-binding domains (CBD1 and CBD2) located in the large intracellular loop between transmembrane segments 5 and 6. We investigated Ca(2+)-dependent movements as changes in FRET between exchanger proteins tagged with CFP or YFP at position 266 within the large cytoplasmic loop. Data indicate that the exchanger assembles as a dimer in the plasma membrane. Addition of Ca(2+) decreases the distance between the cytoplasmic loops of NCX pairs. The Ca(2+)-dependent movements detected between paired NCXs were abolished by mutating the Ca(2+) coordination sites in CBD1 (D421A, E451A, and D500V), whereas disruption of the primary Ca(2+) coordination site in CBD2 (E516L) had no effect. Thus, the Ca(2+)-induced conformational changes of NCX dimers arise from the movement of CBD1. FRET studies of CBD1, CBD2, and CBD1-CBD2 peptides displayed Ca(2+)-dependent movements with different apparent affinities. CBD1-CBD2 showed a Ca(2+)-dependent phenotype mirroring full-length NCX but distinct from both CBD1 and CBD2.


Asunto(s)
Calcio/metabolismo , Multimerización de Proteína , Intercambiador de Sodio-Calcio/química , Intercambiador de Sodio-Calcio/metabolismo , Animales , Sitios de Unión/genética , Calcio/farmacología , Membrana Celular/metabolismo , Citoplasma/metabolismo , Perros , Femenino , Transferencia Resonante de Energía de Fluorescencia , Células HEK293 , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Potenciales de la Membrana , Mutación , Oocitos/metabolismo , Oocitos/fisiología , Conformación Proteica/efectos de los fármacos , Intercambiador de Sodio-Calcio/genética , Xenopus laevis
14.
J Biol Chem ; 285(7): 5066-75, 2010 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-20007711

RESUMEN

Store-operated Ca(2+) entry (SOCE) due to activation of Ca(2+) release-activated Ca(2+) (CRAC) channels leads to sustained elevation of cytoplasmic Ca(2+) and activation of lymphocytes. CRAC channels consisting of four pore-forming Orai1 subunits are activated by STIM1, an endoplasmic reticulum Ca(2+) sensor that senses intracellular store depletion and migrates to plasma membrane proximal regions to mediate SOCE. One of the fundamental properties of CRAC channels is their Ca(2+)-dependent fast inactivation. To identify the domains of Orai1 involved in fast inactivation, we have mutated residues in the Orai1 intracellular loop linking transmembrane segment II to III. Mutation of four residues, V(151)SNV(154), at the center of the loop (MutA) abrogated fast inactivation, leading to increased SOCE as well as higher CRAC currents. Point mutation analysis identified five key amino acids, N(153)VHNL(157), that increased SOCE in Orai1 null murine embryonic fibroblasts. Expression or direct application of a peptide comprising the entire intracellular loop or the sequence N(153)VHNL(157) blocked CRAC currents from both wild type (WT) and MutA Orai1. A peptide incorporating the MutA mutations had no blocking effect. Concatenated Orai1 constructs with four MutA monomers exhibited high CRAC currents lacking fast inactivation. Reintroduction of a single WT monomer (MutA-MutA-MutA-WT) was sufficient to fully restore fast inactivation, suggesting that only a single intracellular loop can block the channel. These data suggest that the intracellular loop of Orai1 acts as an inactivation particle, which is stabilized in the ion permeation pathway by the N(153)VHNL(157) residues. These results along with recent reports support a model in which the N terminus and the selectivity filter of Orai1 as well as STIM1 act in concert to regulate the movement of the intracellular loop and evoke fast inactivation.


Asunto(s)
Canales de Calcio/metabolismo , Animales , Canales de Calcio/química , Canales de Calcio/efectos de los fármacos , Canales de Calcio/genética , Canales de Calcio/fisiología , Línea Celular , Células Cultivadas , Células HeLa , Humanos , Ratones , Mutagénesis , Proteína ORAI1 , Técnicas de Placa-Clamp , Péptidos/química , Péptidos/genética , Péptidos/farmacología , Relación Estructura-Actividad
15.
J Physiol ; 586(7): 1833-48, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18276733

RESUMEN

Inwardly rectifying potassium (Kir) channels are gated by the interaction of their cytoplasmic regions with membrane-bound phosphatidylinositol-4,5-bisphosphate (PIP(2)). In the present study, we examined how PIP(2) interaction regulates channel availability and channel openings to various subconductance levels (sublevels) as well as the fully open state in the strong inward rectifier Kir2.1 channel. Various Kir2.1 channel constructs were expressed in Xenopus oocytes and single channel or macroscopic currents were recorded from inside-out patches. The wild-type (WT) channel rarely visited the subconductance levels under control conditions. However, upon reducing Kir2.1 channel interaction with PIP(2) by a variety of interventions, including PIP(2) antibodies, screening PIP(2) with neomycin, or mutating PIP(2) binding sites (e.g. K188Q), visitation to the sublevels was markedly increased before channels were converted to an unavailable mode in which they did not open. No channel activity was detected in channels with the double mutation K188A/R189A, a mutant which exhibits extremely weak interaction with PIP(2). By linking subunits together in tandem dimers or tetramers containing mixtures of WT and K188A/R189A subunits, we demonstrate that one functional PIP(2)-interacting WT subunit is sufficient to convert channels from the unavailable to the available mode with a high open probability dominated by the fully open state, with similar kinetics as tetrameric WT channels. Occasional openings to sublevels become progressively less frequent as the number of WT subunits increases. Quantitative analysis reveals that the interaction of PIP(2) with WT subunits exerts strong positive cooperativity in both converting the channels from the unavailable to the available mode, and in promoting the fully open state over sublevels. We conclude that the interaction of PIP(2) with only one Kir2.1 subunit is sufficient for the channel to become available and to open to its full conductance state. Interaction with additional subunits exerts positive cooperativity at multiple levels to further enhance channel availability and promote the fully open state.


Asunto(s)
Fosfatidilinositol 4,5-Difosfato/fisiología , Canales de Potasio de Rectificación Interna/fisiología , Subunidades de Proteína/fisiología , Animales , Anticuerpos/farmacología , Femenino , Ratones , Modelos Estructurales , Mutación/genética , Neomicina/farmacología , Oocitos/citología , Técnicas de Placa-Clamp , Canales de Potasio de Rectificación Interna/química , Canales de Potasio de Rectificación Interna/genética , Conformación Proteica , Inhibidores de la Síntesis de la Proteína/farmacología , Xenopus laevis
16.
Pflugers Arch ; 456(2): 307-22, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18071748

RESUMEN

To study intracellular glucose homeostasis, the glucose nanosensor FLIPglu-600 microM, which undergoes changes in fluorescence resonance energy transfer (FRET) upon interaction with glucose, was expressed in four mammalian cell lines: COS-7, CHO, HEK293, and C2C12. Upon addition of extracellular glucose, the intracellular FRET ratio decreased rapidly as intracellular glucose increased. The kinetics were fast (tau=5 to 15 s) in COS and C2C12 cells and slow (tau=20 to 40 s) in HEK and CHO cells. Upon removal of extracellular glucose, the FRET ratio returned to its initial value at similar rates (tau=15 to 40 s) in all cell types. In all cell types, the glucose uptake FRET signal was blocked by the glucose transporter (GLUTx) inhibitor cytochalasin B and was not affected by the Na/glucose transporter inhibitor phlorizin. Glucose clearance was inhibited by the glycolytic inhibitor iodoacetate. Using beta-escin to permeabilize the cell, we found that the glucose gradient across the membrane was strongly dependent on the rates of glucose uptake versus glucose clearance. With 10 mM extracellular glucose and a high rate of glucose clearance, intracellular glucose level fell below 100 muM when glucose uptake rate was low, whereas it exceeded 0.5 mM when glucose uptake was high. Cells cultured in high glucose maintained lower basal intracellular glucose levels than cells cultured in low glucose, attributed to "reciprocal regulation" of glycolysis and gluconeogenesis. Basal glucose level also increased with elevated temperatures. Experiments performed with C2C12 cells demonstrated a shift from fast glucose uptake to slow glucose uptake in the absence of insulin during differentiation.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia/instrumentación , Glucosa/farmacocinética , Hemostasis/fisiología , Riñón/metabolismo , Mioblastos Esqueléticos/metabolismo , Ovario/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Células CHO , Células COS , Línea Celular , Chlorocebus aethiops , Cricetinae , Cricetulus , Citocalasina B/farmacología , Inhibidores Enzimáticos/farmacología , Femenino , Transferencia Resonante de Energía de Fluorescencia/métodos , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Hemostasis/efectos de los fármacos , Humanos , Yodoacetatos/farmacología , Riñón/citología , Riñón/efectos de los fármacos , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/efectos de los fármacos , Nanoestructuras , Ovario/citología , Ovario/efectos de los fármacos , Florizina/farmacología , Temperatura
17.
Prog Biophys Mol Biol ; 94(3): 320-35, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16837026

RESUMEN

All members of the inwardly rectifying potassium channels (Kir1-7) are regulated by the membrane phospholipid, phosphatidylinosital-4,5-bisphosphate (PIP(2)). Some are also modulated by other regulatory factors or ligands such as ATP and G-proteins, which give them their common names, such as the ATP sensitive potassium (K(ATP)) channel and the G-protein gated potassium channel. Other more non-specific regulators include polyamines, kinases, pH and Na(+) ions. Recent studies have demonstrated that PIP(2) acts cooperatively with other regulatory factors to modulate Kir channels. Here we review how PIP(2) and co-factors modulate channel activities in each subfamily of the Kir channels.


Asunto(s)
Ligandos , Fosfatidilinositol 4,5-Difosfato/fisiología , Canales de Potasio de Rectificación Interna/metabolismo , Secuencia de Aminoácidos , Animales , Humanos , Datos de Secuencia Molecular , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/fisiología
18.
J Physiol ; 571(Pt 2): 303-17, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16373383

RESUMEN

ATP-sensitive K+ channels composed of the pore-forming protein Kir6.2 and the sulphonylurea receptor SUR1 are inhibited by ATP and activated by Phosphatidylinositol Bisphosphate (PIP2). Residues involved in binding of these ligands to the Kir6.2 cytoplasmic domain have been identified, and it has been hypothesized that gating mechanisms involve conformational changes in the regions of the bundle crossing and/or the selectivity filter of Kir6.2. Regulation of Kir6.2 by SUR1, however, is not well-understood, even though this process is ATP and PIP2 dependent. In this study, we investigated the relationship between channel regulation by SUR1 and PIP2 by comparing a number of single and double mutants known to affect open probability (P(o)), PIP2 affinity, and sulphonylurea and MgADP sensitivity. When coexpressed with SUR1, the Kir6.2 mutant C166A, which is characterized by a P(o) value close to 0.8, exhibits no sulphonylurea or MgADP sensitivity. However, when P(o) was reduced by combining mutations at the PIP2-sensitive residues R176 and R177 with C166A, sulphonylurea and MgADP sensitivities were restored. These effects correlated with a dramatic decrease in PIP2 affinity, as assessed by PIP2-induced channel reactivation and inhibition by neomycin, an antagonist of PIP2 binding. Based on macroscopic and single-channel data, we propose a model in which entry into the high-P(o) bursting state by the C166A mutation or by SUR1 depends on the interaction of PIP2 with R176 and R177 and, to a lesser extent, R54. In conjunction with this PIP2-dependent process, SUR1 also regulates channel activity via a PIP2-independent, but MgADP-dependent process.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/fisiología , Adenosina Trifosfato/farmacología , Fosfatidilinositol 4,5-Difosfato/farmacología , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/fisiología , Canales de Potasio/metabolismo , Receptores de Droga/fisiología , Adenosina Difosfato/farmacología , Línea Celular , Membrana Celular/fisiología , Relación Dosis-Respuesta a Droga , Humanos , Activación del Canal Iónico/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Modelos Biológicos , Mutación Puntual , Canales de Potasio/genética , Canales de Potasio/fisiología , Canales de Potasio de Rectificación Interna/química , Receptores de Sulfonilureas , Transfección
19.
J Gen Physiol ; 126(6): 541-9, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16316973

RESUMEN

Phosphatidylinosital-4,5-bisphosphate (PIP2) acts as an essential factor regulating the activity of all Kir channels. In most Kir members, the dependence on PIP2 is modulated by other factors, such as protein kinases (in Kir1), G(betagamma) (in Kir3), and the sulfonylurea receptor (in Kir6). So far, however, no regulator has been identified in Kir2 channels. Here we show that polyamines, which cause inward rectification by selectively blocking outward current, also regulate the interaction of PIP2 with Kir2.1 channels to maintain channel availability. Using spermine and diamines as polyamine analogs, we demonstrate that both spontaneous and PIP2 antibody-induced rundown of Kir2.1 channels in excised inside-out patches was markedly slowed by long polyamines; in contrast, polyamines with shorter chain length were ineffective. In K188Q mutant channels, which have a low PIP2 affinity, application PIP2 (10 microM) was unable to activate channel activity in the absence of polyamines, but markedly activated channels in the presence of long diamines. Using neomycin as a measure of PIP2 affinity, we found that long polyamines were capable of strengthening either the wild type or K188Q channels' interaction with PIP2. The negatively charged D172 residue inside the transmembrane pore region was critical for the shift of channel-PIP2 binding affinity by long polyamines. Sustained pore block by polyamines was neither sufficient nor necessary for this effect. We conclude that long polyamines serve a dual role as both blockers and coactivators (with PIP2) of Kir2.1 channels.


Asunto(s)
Fosfatidilinositol 4,5-Difosfato/metabolismo , Poliaminas/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Animales , Membrana Celular/metabolismo , Electrofisiología , Femenino , Transporte Iónico , Oocitos/metabolismo , Potasio/metabolismo , Xenopus
20.
J Physiol ; 568(Pt 3): 931-40, 2005 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16166157

RESUMEN

ATP-sensitive K (K(ATP)) channels are composed of Kir6, the pore-forming protein, and the sulphonylurea receptor SUR, a regulatory protein. We and others have previously shown that positively charged residues in the C terminus of Kir6.2, including R201 and K185, interact with the alpha and beta phosphate groups of ATP, respectively, to induce channel closure. A positively charged residue in the N terminus, R50, is also important, and has been proposed to interact with either the gamma or beta phosphate group of ATP. To examine this issue, we systematically mutated R50 to residues of different size, charge and hydropathy, and examined the effects on adenine nucleotide sensitivity in the absence and presence of SUR1. In the absence of SUR1, only the size of residue 50 significantly altered ATP sensitivity, with smaller side chains decreasing ATP sensitivity. In the presence of SUR1, however, hydrophathy and charge also played a role. Hydrophilic residues decreased ATP sensitivity more than hydrophobic residues for small size residues, and, surprisingly, negatively charged residues E and D preserved ATP sensitivity and increased ADP sensitivity relative to the wild-type residue R. These observations suggest that a negative charge near position 50, due to either mutation of R50 or the interaction of the gamma phosphate group of ATP with R50, facilitates closure of the ATP-dependent gate. Mutation of the nearby positively charged residue R54, known to be involved in stabilizing channel opening via electrostatic interactions with phosphatidylinositol 4,5-bisphosphate (PIP2), also caused increased ADP sensitivity as compared with ATP, suggesting a loss of function of ATP's gamma phosphate. Based on these results, we propose that a phosphate group or a negative charge at position 50 initiates channel closure by destabilizing the electrostatic interactions between negative phosphate groups of PIP2 and residues such as R54.


Asunto(s)
Adenosina Trifosfato/química , Adenosina Trifosfato/farmacología , Activación del Canal Iónico/fisiología , Riñón/metabolismo , Fosfatos/metabolismo , Canales de Potasio de Rectificación Interna/química , Canales de Potasio de Rectificación Interna/fisiología , Animales , Sitios de Unión , Línea Celular , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Unión Proteica , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA