Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 147
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 500(2): 204-210, 2018 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-29634926

RESUMEN

Recently, the modulation of cellular inflammatory responses via endogenous regulators became a major focus of medically relevant investigations. Prostaglandins (PGs) are attractive regulatory molecules, but their synthesis and mechanisms of action in brain cells are still unclear. Astrocytes are involved in manifestation of neuropathology and their proliferation is an important part of astrogliosis, a cellular neuroinflammatory response. The aims of our study were to measure synthesis of PGs by astrocytes, and evaluate their influence on proliferation in combination with addition of inflammatory pathway inhibitors. With UPLC-MS/MS analysis we detected primary PGs (1410 ±â€¯36 pg/mg PGE2, 344 ±â€¯24 PGD2) and cyclopentenone PGs (cyPGs) (87 ±â€¯17 15d-PGJ2, 308 ±â€¯23 PGA2) in the extracellular medium after 24-h lipopolysaccharide (LPS) stimulation of astrocytes. PGs reduced astrocytic proliferation with the following order of potencies (measured as inhibition at 20 µM): most potent 15d-PGJ2 (90%) and PGA2 (80%), > PGD2 (40%) > 15d-PGA2 (20%) > PGE2 (5%), the least potent. However, PGF2α and 2-cyclopenten-1-one, and ciglitazone and rosiglitazone (synthetic agonists of PPARγ) had no effect. Combinations of cyPGs with SC-560 or NS-398 (specific anti-inflammatory inhibitors of cyclooxygenase-1 and -2, respectively) were not effective; while GW9662 (PPARγ antagonist) or MK-741 (inhibitor of multidrug resistance protein-1, MRP1, and CysLT1 receptors) amplified the inhibitory effect of PGA2 and 15d-PGJ2. Although concentrations of individual PGs and cyPGs are low, all of them, as well as primary PGs suppress proliferation. Thus, the effects are potentially additive, and activated PGs synthesis suppresses proliferation in astrocytes.


Asunto(s)
Astrocitos/citología , Astrocitos/metabolismo , Ciclopentanos/metabolismo , Prostaglandinas/biosíntesis , Animales , Astrocitos/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cromatografía Liquida , Lipopolisacáridos/farmacología , PPAR gamma/agonistas , PPAR gamma/metabolismo , Prostaglandina D2/análogos & derivados , Prostaglandina D2/química , Prostaglandina D2/metabolismo , Prostaglandinas A/química , Prostaglandinas A/metabolismo , Ratas Wistar , Espectrometría de Masas en Tándem
2.
Biochim Biophys Acta ; 1852(5): 925-36, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25583114

RESUMEN

X-linked adrenoleukodystrophy (X-ALD) is a severe neurodegenerative disorder resulting from defective ABCD1 transport protein. ABCD1 mediates peroxisomal uptake of free very-long-chain fatty acids (VLCFA) as well as their CoA-esters. Consequently, VLCFA accumulate in patients' plasma and tissues, which is considered as pathogenic X-ALD triggering factor. Clinical symptoms are mostly manifested in neural tissues and adrenal gland. Here, we investigate astrocytes from wild-type control and a genetic X-ALD mouse model (Abcd1-knockout), exposed to supraphysiological VLCFA (C22:0, C24:0 and C26:0) concentrations. They exhibit multiple impairments of energy metabolism. Furthermore, brain mitochondria from Abcd1(-/-) mice and wild-type control respond similarly to VLCFA with increased ROS generation, impaired oxidative ATP synthesis and diminished Ca(2+) uptake capacity, suggesting that a defective ABCD1 exerts no adaptive pressure on mitochondria. In contrast, astrocytes from Abcd1(-/-) mice respond more sensitively to VLCFA than wild-type control astrocytes. Moreover, long-term application of VLCFA induces high ROS generation, and strong in situ depolarization of mitochondria, and, in Abcd1(-/-) astrocytes, severely diminishes the capability to revert oxidized pyridine nucleotides to NAD(P)H. In addition, observed differences in responses of mitochondria and astrocytes to the hydrocarbon chain length of VLCFA suggest that detrimental VLCFA activities in astrocytes involve defective cellular functions other than mitochondria. In summary, we clearly demonstrate that VLCFA increase the vulnerability of Abcd1(-/-) astrocytes.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/deficiencia , Adrenoleucodistrofia/metabolismo , Astrocitos/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Ácidos Grasos/farmacología , Mitocondrias/efectos de los fármacos , Miembro 1 de la Subfamilia D de Transportador de Casetes de Unión al ATP , Transportadoras de Casetes de Unión a ATP/genética , Adrenoleucodistrofia/genética , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Astrocitos/metabolismo , Calcio/metabolismo , Calcio/farmacocinética , Células Cultivadas , Ácidos Grasos/química , Transporte Iónico/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Mitocondrias/metabolismo , NADP/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Espectrometría de Fluorescencia , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
3.
Neurochem Res ; 41(6): 1250-62, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26842930

RESUMEN

The current study demonstrates that in hippocampal neurons mitochondrial Ca(2+) processing supports Ca(2+) influx via ionotropic glutamate (Glu) receptors. We define mitochondrial Ca(2+) processing as Ca(2+) uptake via mitochondrial Ca(2+) uniporter (MCU) combined with subsequent Ca(2+) release via mitochondrial Na(+)/Ca(2+) exchanger (NCX). Our tool is to measure the Ca(2+) influx rate in primary hippocampal co-cultures, i.e. neurons and astrocytes, by fluorescent digital microscopy, using a Fura-2-quenching method where we add small amounts of Mn(2+) in the superfusion medium. Thus, Ca(2+) influx is measured with Mn(2+) in the bath. Ru360 as inhibitor of mitochondrial Ca(2+) uptake through MCU strongly reduces the rate of Ca(2+) influx in Glu-stimulated primary hippocampal neurons. Similarly, the Ca(2+) influx rate in Glu-stimulated neurons declines after suppression of potential-dependent MCU, when we depolarize mitochondria with rotenone. With inhibition of Ca(2+) release from mitochondria via NCX using CGP-37157 the Ca(2+) influx via N-methyl-D-aspartate (NMDA)- and kainate-sensitive receptors is slowed down. Working jointly as mitochondrial Ca(2+) processing unit, MCU and NCX, apparently sustain the Ca(2+) throughput of activated Glu-sensitive receptors. Our results revise the role frequently attributed to mitochondria in neuronal Ca(2+) homeostasis, where mitochondria function mainly as Ca(2+) buffer, and prevent excessively high cytosolic Ca(2+) concentration increase during neuronal activity. The mechanism to control Ca(2+) influx in neurons, as discovered in this study, highlights mitochondrial Ca(2+) processing as a promising pharmacological target. We discuss this pathway in relation to the endoplasmic reticulum-related mechanisms of Ca(2+) processing.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Mitocondrias/metabolismo , Neuronas/metabolismo , Receptores de Glutamato/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Animales , Astrocitos/metabolismo , Señalización del Calcio/fisiología , Técnicas de Cocultivo , Femenino , Hipocampo/citología , Hipocampo/metabolismo , Masculino , Ratas , Ratas Wistar
4.
J Neurochem ; 134(1): 113-24, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25818681

RESUMEN

Peroxisome proliferator-activated receptors (PPAR)-α and -γ in astrocytes play important roles in inflammatory brain pathologies. Understanding the regulation of both activity and expression levels of PPARs is an important neuroscience issue. Toll-like receptor (TLR) agonists are inflammatory stimuli that could modulate PPAR, but the mechanisms of their control in astrocytes are poorly understood. In the present study, we report that lipopolysaccharide, peptidoglycan, and flagellin, which are agonists of TLR4, TLR1/2, and TLR5, respectively, exert time- and nuclear factor kappa-light-chain-enhancer of activated B cells-dependent suppression of mRNA, protein and activity of PPARα and PPARγ. In naïve astrocytes, PPARα and PPARγ mRNA have short turnover time (half-life about 30 min for PPARα, 75 min for PPARγ) with a nearly two-fold stabilization after TLR-activation. p38 inhibition abolished TLR-induced stabilization. The levels of PPARα and PPARγ mRNA, and protein and DNA-binding activity could be modified using c-Jun N-terminal Kinase and p38 inhibitors. In addition, the expression levels of both PPARα and PPARγ isotypes were induced after inhibition of protein synthesis. This induction signifies participation of additional regulatory proteins with short life-time. They are p38-sensitive for PPARα and c-Jun N-terminal Kinase-sensitive for PPARγ. Thus, PPARα and PPARγ are regulated in astrocytes on mRNA and protein levels, mRNA stability, and DNA-binding activity during TLR-mediated responses. Astrocytes have the triad of PPARα, PPARß/δ, and PPARγ in regulation of proinflammatory responses. Activation of Toll-like receptors (TLR) leads to PPARß/δ overexpression, PPARα and PPARγ suppression via TLR/NF-κB pathway on mRNA, protein and activity levels. Mitogen-activated protein kinases (MAPK) p38 and JNK are involved in regulation of PPAR expression. p38 MAPK plays a special role in stabilization of PPAR mRNA.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , PPAR alfa/fisiología , PPAR gamma/fisiología , Receptores Toll-Like/agonistas , Receptores Toll-Like/metabolismo , Animales , Astrocitos/efectos de los fármacos , Encéfalo/efectos de los fármacos , Células Cultivadas , Femenino , Masculino , Peptidoglicano/farmacología , Ratas , Ratas Wistar
5.
Cell Tissue Res ; 359(3): 817-27, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25519044

RESUMEN

The protease-activated receptors are a group of unique G protein-coupled receptors, including PAR-1, PAR-2, PAR-3 and PAR-4. PAR-2 is activated by multiple trypsin-like serine proteases, including trypsin, tryptase and coagulation proteases. The clusters of phosphorylation sites in the PAR-2 carboxyl tail are suggested to be important for the binding of adaptor proteins to initiate intracellular signaling to Ca(2+) and mitogen-activated protein kinases. To explore the functional role of PAR-2 carboxyl tail in controlling intracellular Ca(2+), ERK and AKT signaling, a series of truncated mutants containing different clusters of serines/threonines were generated and expressed in HEK293 cells. Firstly, we observed that lack of the complete C-terminus of PAR-2 in a mutated receptor gave a relatively low level of localization on the cell plasma membrane. Secondly, the shortened carboxyl tail containing 13 amino acids was sufficient for receptor internalization. Thirdly, the cells expressing truncation mutants showed deficits in their capacity to couple to intracellular Ca(2+) and ERK and AKT signaling upon trypsin challenge. In addition, HEK293 cells carrying different PAR-2 truncation mutants displayed decreased levels of cell survival after long-lasting trypsin stimulation. In summary, the PAR-2 carboxyl tail was found to control the receptor localization, internalization, intracellular Ca(2+) responses and signaling to ERK and AKT. The latter can be considered to be important for cell death control.


Asunto(s)
Espacio Intracelular/metabolismo , Receptor PAR-2/química , Receptor PAR-2/metabolismo , Transducción de Señal , Animales , Calcio/metabolismo , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Endocitosis/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HEK293 , Humanos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Receptor PAR-2/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad , Tripsina/farmacología
6.
Purinergic Signal ; 11(4): 441-54, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26407872

RESUMEN

This review describing the role of P2Y receptors in neuropathological conditions focuses on obvious differences between results demonstrating either a role in neuroprotection or in neurodegeneration, depending on in vitro and in vivo models. Such critical juxtaposition puts special emphasis on discussions of beneficial and detrimental effects of P2Y receptor agonists and antagonists in these models. The mechanisms reported to underlie the protection in vitro include increased expression of oxidoreductase genes, like carbonyl reductase and thioredoxin reductase; increased expression of inhibitor of apoptosis protein-2; extracellular signal-regulated kinase- and Akt-mediated antiapoptotic signaling; increased expression of Bcl-2 proteins, neurotrophins, neuropeptides, and growth factors; decreased Bax expression; non-amyloidogenic APP shedding; and increased neurite outgrowth in neuronal cells. Animal studies investigating the influence of P2Y receptors in middle cerebral artery occlusion (MCAO) models for stroke prove beneficial effects of P2Y receptor antagonists. In MCAO mice and rats, the application of broad-range P2 receptor antagonists decreased the infarct volume and improved neurological outcome. Moreover, antagonists of the P2Y1 receptor, one of the most abundant P2Y receptor subtypes in brain tissue, decreased neuronal loss and improved spatial memory in rats after traumatic brain injury (TBI). Currently available data show a discrepancy between in vitro and in vivo models concerning the benefits of P2Y receptor activation in pathological conditions. In vitro models demonstrate protection by P2Y receptor agonists, but in vivo P2Y receptor activation deteriorates the outcome after MCAO and controlled cortical impact brain injury, a TBI model. To broaden the scope of the review, we additionally discuss publications that demonstrate detrimental effects of P2Y receptor agonists in vitro and publications showing protective effects of agonists in vivo. All these studies help to better understand the significant role of P2Y receptors especially in stroke models and to develop pharmacological strategies for the treatment of stroke.


Asunto(s)
Encefalopatías/fisiopatología , Enfermedades Neurodegenerativas/fisiopatología , Neuronas , Neuroprotección , Receptores Purinérgicos P2Y , Accidente Cerebrovascular/fisiopatología , Animales , Modelos Animales de Enfermedad , Humanos
7.
Mol Cell Neurosci ; 59: 127-34, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24513118

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) are important members of the nuclear receptor superfamily. Ligands of these nuclear receptors (PPARα, ß/δ and γ) belong to a wide range of lipophilic substances. In spite of the proven neuroprotective efficacy of PPARß/δ in models of neurological diseases, the biology of PPARß/δ in the brain has been much less investigated than that of PPARα and PPARγ. In the present study, we test the hypothesis that neuroprotection induced by PPARß/δ could rely on the regulation of ceramide metabolism. We found that preincubation of neural cells with the PPARß/δ agonist L-165041 exerts significant protection against ceramide-induced cell death. Most importantly, L-165041 protects against ceramide-induced cell death not only before the insult, but also after the onset of the insult. To identify the mechanism of protection, we show that L-165041 upregulates ceramide kinase (CerK) expression levels in neural cells. Consistent with that, we detected that pharmacological inhibition of CerK reduces the protective effects of L-165041. To further decipher the mechanism of protection, gene knockdown in astrocytes was studied. Knockdown of PPARß/δ and CerK in astrocytes was used to verify that the protective effects of L-165041 are CerK- and PPARß/δ-dependent. We demonstrate that in CerK- or PPARß/δ-knockdown astrocytes, addition of L-165041 has no protective effect. Thus, we conclude that PPARß/δ protects neural cells against ceramide-induced cell death via induction and activation of CerK.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Ceramidas/toxicidad , Neuronas/metabolismo , PPAR delta/metabolismo , PPAR-beta/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Animales , Astrocitos/efectos de los fármacos , Encéfalo/citología , Células Cultivadas , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , PPAR delta/genética , PPAR-beta/genética , Fenoxiacetatos/farmacología , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Ratas , Ratas Wistar
8.
J Neurochem ; 129(4): 602-13, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24524250

RESUMEN

The P2Y11 nucleotide receptor detects high extracellular ATP concentrations. Mutations of the human P2RY11 gene can play a role in brain autoimmune responses, and the P2Y11 receptor alanine-87-threonine (A87T) polymorphism has been suggested to affect immune-system functions. We investigated receptor functionality of the P2Y11 A87T mutant using HEK293 and 1321N1 astrocytoma cells. In HEK293 cells, the P2Y11 receptor agonist 3'-O-(4-benzoylbenzoyl)adenosine 5'-triphosphate (BzATP) was completely inactive in evoking intracellular calcium release while the potency of ATP was reduced. ATP was also less potent in triggering cAMP generation. However, 1321N1 astrocytoma cells, which lack any endogenous P2Y1 receptors, did not display a reduction. Only when 1321N1 cells were co-transfected with P2Y11 A87T and P2Y1 receptors, the calcium responses to the P2Y11 receptor-specific agonist BzATP were reduced. It is already known that P2Y1 and P2Y11 receptors interact. We thus conclude that the physiological impact of A87T mutation of the P2Y11 receptor derives from detrimental effects on P2Y1 -P2Y11 receptor interaction. We additionally investigated alanine-87-serine and alanine-87-tyrosine P2Y11 receptor mutants. Both mutations rescue the response to BzATP in HEK293 cells, thus ruling out polarity of amino acid-87 to be the molecular basis for altered receptor characteristics. We further found that the P2Y11 A87T receptor shows complete loss of nucleotide-induced internalization in HEK293 cells. Thus, we demonstrate impaired signaling of the P2Y11 A87T-mutated receptors when co-operating with P2Y1 receptors.


Asunto(s)
Sustitución de Aminoácidos , Señalización del Calcio/efectos de los fármacos , Polimorfismo de Nucleótido Simple , Receptores Purinérgicos P2Y1/metabolismo , Receptores Purinérgicos P2/genética , Adenosina Difosfato/análogos & derivados , Adenosina Difosfato/farmacología , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Astrocitoma/patología , Autoinmunidad , Encéfalo/inmunología , Línea Celular Tumoral , AMP Cíclico/metabolismo , Células HEK293 , Humanos , Mutagénesis Sitio-Dirigida , Mapeo de Interacción de Proteínas , Agonistas Purinérgicos/farmacología , Receptores Purinérgicos P2/metabolismo , Relación Estructura-Actividad , Tionucleótidos/farmacología , Transfección
9.
J Neurochem ; 131(2): 163-76, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24923354

RESUMEN

Ca(2+) -independent phospholipase A2 (iPLA2 ) is hypothesized to control mitochondrial reactive oxygen species (ROS) generation. Here, we modulated the influence of iPLA2 -induced liberation of non-esterified free fatty acids on ROS generation associated with the electron transport chain. We demonstrate enzymatic activity of membrane-associated iPLA2 in native, energized rat brain mitochondria (RBM). Theoretically, enhanced liberation of free fatty acids by iPLA2 modulates mitochondrial ROS generation, either attenuating the reversed electron transport (RET) or deregulating the forward electron transport of electron transport chain. For mimicking such conditions, we probed the effect of docosahexaenoic acid (DHA), a major iPLA2 product on ROS generation. We demonstrate that the adenine nucleotide translocase partly mediates DHA-induced uncoupling, and that low micromolar DHA concentrations diminish RET-dependent ROS generation. Uncoupling proteins have no effect, but the adenine nucleotide translocase inhibitor carboxyatractyloside attenuates DHA-linked uncoupling effect on RET-dependent ROS generation. Under physiological conditions of forward electron transport, low micromolar DHA stimulates ROS generation. Finally, exposure of RBM to the iPLA2 inhibitor bromoenol lactone (BEL) enhanced ROS generation. BEL diminished RBM glutathione content. BEL-treated RBM exhibits reduced Ca(2+) retention capacity and partial depolarization. Thus, we rebut the view that iPLA2 attenuates oxidative stress in brain mitochondria. However, the iPLA2 inhibitor BEL has detrimental activities on energy-dependent mitochondrial functions. The Ca(2+) -independent phospholipase A2 (iPLA2 ), a FFA (free fatty acids)-generating membrane-attached mitochondrial phospholipase, is potential to regulate ROS (reactive oxygen species) generation by mitochondria. FFA can either decrease reversed electron transport (RET)-linked or enhance forward electron transport (FET)-linked ROS generation. In the physiological mode of FET, iPLA2 activity increases ROS generation. The iPLA2 inhibitor BEL exerts detrimental effects on energy-dependent mitochondrial functions.


Asunto(s)
Encéfalo/metabolismo , Calcio/metabolismo , Ácidos Docosahexaenoicos/metabolismo , Mitocondrias/metabolismo , Naftalenos/metabolismo , Fosfolipasas A2/metabolismo , Pironas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Encéfalo/efectos de los fármacos , Calcio/farmacología , Ácidos Docosahexaenoicos/farmacología , Transporte de Electrón/efectos de los fármacos , Transporte de Electrón/fisiología , Mitocondrias/efectos de los fármacos , Naftalenos/farmacología , Pironas/farmacología , Ratas , Ratas Wistar
10.
J Neurochem ; 130(4): 563-74, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24806616

RESUMEN

Peroxisome proliferator-activated receptor ß/δ (PPARß/δ) is a potential regulator of neuroinflammation. Toll-like receptors (TLR) are innate immunity-related receptors of inflammatory stimuli. In the present report, we evaluate the molecular mechanisms of regulation of mRNA, protein, and transcriptional activity levels of PPARß/δ by agonists of TLR4, TLR1/2, and TLR5, using lipopolysaccharide (LPS), peptidoglycan, and flagellin, respectively. We found that these stimuli increase the PPARß/δ levels in astrocytes. Expression and activity of PPARß/δ are separately regulated by inhibitors of p38, MEK1/2, extracellular signal-regulated kinases 1/2, and c-Jun N-terminal Kinase mitogen-activated protein kinases. The LPS-induced kinetics of PPARß/δ expression is similar to that of the proinflammatory gene cyclooxygenase 2. Moreover, for both genes the expression depends on nuclear factor kappa-light-chain-enhancer of activated B cells and p38, and is induced after inhibition of protein synthesis. The up-regulation of the expression after inhibition of protein synthesis signifies the participation of a labile protein in regulation of PPARß/δ expression. In contrast to cyclooxygenase 2, the cycloheximide-sensitive PPARß/δ expression was not responsive to nuclear factor kappa-light-chain-enhancer of activated B cells inhibition. Measurements of PPARß/δ mRNA stability showed that the PPARß/δ mRNA levels are regulated post-transcriptionally. We found that in LPS-stimulated astrocytes, the half-life of PPARß/δ mRNA was 50 min. Thus, we demonstrate that PPARß/δ expression and activity are regulated in TLR agonist-stimulated astrocytes by mechanisms that are widely used for regulation of proinflammatory genes. Protein expression level of nuclear receptor PPARß/δ is important for functions of this transcription factor. We investigate the regulatory mechanisms of PPARß/δ in rat primary astrocytes stimulated by agonists of toll-like receptors (TLR): TLR4, TLR1/2, and TLR5. Expression, activity, mRNA stability, and superinduction of PPARß/δ were up-regulated after TLR stimulation. These processes are sensitive to MAPKs and NF-kB inhibitors. Superinduction is up-regulation of mRNA expression after inhibition of protein synthesis.


Asunto(s)
Astrocitos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , PPAR delta/biosíntesis , Inhibidores de Proteínas Quinasas/farmacología , Receptores Toll-Like/agonistas , Animales , Astrocitos/efectos de los fármacos , Western Blotting , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Inducción Enzimática/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Lipopolisacáridos/farmacología , PPAR delta/efectos de los fármacos , PPAR delta/genética , Cultivo Primario de Células , Procesamiento Proteico-Postraduccional/efectos de los fármacos , ARN/biosíntesis , ARN/genética , ARN/aislamiento & purificación , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Transcripción Genética/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
11.
Hum Mol Genet ; 21(12): 2807-14, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22442204

RESUMEN

Infantile neuroaxonal dystrophy (INAD; OMIM #no. 256600) is an inherited degenerative nervous system disorder characterized by nerve abnormalities in brain, spinal cord and peripheral nerves. About 85% of INAD patients carry mutations in the PLA2G6 gene that encodes for a Ca(2+)-independent phospholipase A(2) (VIA iPLA(2)), but how these mutations lead to disease is unknown. Besides regulating phospholipid homeostasis, VIA iPLA(2) is emerging with additional non-canonical functions, such as modulating store-regulated Ca(2+) entry into cells, and mitochondrial functions. In turn, defective Ca(2+) regulation could contribute to the development of INAD. Here, we studied possible changes in ATP-induced Ca(2+) signaling in astrocytes derived from two mutant strains of mice. The first strain carries a hypomorphic allele of the Pla2g6 that reduces transcript levels to 5-10% of that observed in wild-type mice. The second strain carries a point mutation in Pla2g6 that results in inactive VIA iPLA(2) protein with postulated gain in toxicity. Homozygous mice from both strains develop pathology analogous to that observed in INAD patients. The nucleotide ATP is the most important transmitter inducing Ca(2+) signals in astroglial networks. We demonstrate here a severe disturbance in Ca(2+) responses to ATP in astrocytes derived from both mutant mouse strains. The duration of the Ca(2+) responses in mutant astrocytes was significantly reduced when compared with values observed in control cells. We also show that the reduced Ca(2+) responses are probably due to a reduction in capacitative Ca(2+) entry (2.3-fold). Results suggest that altered Ca(2+) signaling could be a central mechanism in the development of INAD pathology.


Asunto(s)
Astrocitos/metabolismo , Calcio/metabolismo , Fosfolipasas A2 Grupo VI/genética , Mutación , Distrofias Neuroaxonales/genética , Adenosina Trifosfato/farmacología , Animales , Astrocitos/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Expresión Génica , Genotipo , Fosfolipasas A2 Grupo VI/metabolismo , Humanos , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Distrofias Neuroaxonales/metabolismo , Distrofias Neuroaxonales/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Biol Chem ; 395(11): 1321-40, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24854535

RESUMEN

Eukaryotic cells express numerous ArfGAPs (ADP-ribosylation factor GTPase-activating proteins). There is increasing knowledge about the function of the brain-specific protein ADAP1 [ArfGAP with dual pleckstrin homology (PH) domain] as well as about its biochemical properties. The ADAP subfamily, also designated centaurin-α, has an N-terminal ArfGAP domain followed by two PH domains. The mammalian ADAP subfamily consists of two identified isoforms, ADAP1 and ADAP2 (centaurin-α1 and -α2). ADAP1 is highly expressed in neurons. We highlight the functional roles of ADAP1 in neuronal differentiation and neurodegeneration. Because of interactions with different proteins and phosphoinositol-lipids, ADAP1 can function as a scaffolding protein in several signal transduction pathways. Firstly, ADAP1 mediates cytoskeletal crosstalk. This is indicated by multiple interactions of ADAP1 with components of the actin and microtubule cytoskeleton. Secondly, regulation of neuronal polarity formation and axon specification by ADAP1 is suggested by crystal structural data obtained for human ADAP1, and the complexes of ADAP1-Ins(1,3,4,5)P4 and/or the forkhead-associated domain of the kinesin KIF13B. These structures support the concept that a KIF13B-ADAP1 complex enhances the local accumulation of PtdIns(3,4,5)P3 at the tips of neurites, and thus favors neuronal polarity. Thirdly, recent evidence unravels a pathological role of ADAP1 because upregulation of ADAP1 by amyloid ß-peptide causes ADAP1-Ras-ERK-dependent translocation of Elk-1 to mitochondria. This impairs mitochondrial functions with subsequent synaptic dysfunction and exacerbates neurodegeneration, as in Alzheimer's disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Neurogénesis , Neuronas/citología , Proteínas Adaptadoras Transductoras de Señales/análisis , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencia de Aminoácidos , Animales , Regulación del Desarrollo de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/análisis , Proteínas del Tejido Nervioso/genética , Enfermedades Neurodegenerativas/genética , Neuronas/metabolismo , Mapas de Interacción de Proteínas
13.
J Bioenerg Biomembr ; 46(2): 135-45, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24532135

RESUMEN

In our previous studies phosphorylation of several membrane-bound proteins in brain and liver mitochondria were found to be regulated by Ca(2+) as a second messenger. One of the proteins, the 46 kDa phosphoprotein was found to be highly phosphorylated when Ca(2+)-induced permeability transition pore (mPTP) was opened in rat brain mitochondria (RBM). In the present study the 46 kDa phosphoprotein was identified as 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNPase) after purification by 2D diagonal electrophoresis following mass spectrometric analysis and Western blot probed with anti-CNP antibody. CNPase was discovered in immunoprecipitates of mitochondria, phosphorylated under both conditions (control and with opened mPTP). Status phosphorylation of CNPase was found to be higher in the inmmunoprecipiates of calcium-overloaded RBM. The phospohoserine and phosphotyrosine residues were detected in phosphorylated 46 kDa band (CNPase) as well as in CNPase immunoprecipitates indicating possible participation of tyrosine and serine protein kinases in phosphorylation of CNPase in mitochondria. The levels of phospo-Ser and phospho-Tyr were increased in RBM with mPTP opened. It was found that CNPase substrate, 2',3'-cAMP (5 µM) and, a non-competitive CNPase inhibitor, atractyloside (5 µM), were able to increase the level of CNPase phosphorylation in calcium-overloaded mitochondria, while CsA (mPTP blocker) was able to strong suppress the phosphorylation of the enzyme. Collectively, our results provide evidence that Ca(2+)-stimulated and mPTP-associated CNPase phosphorylation might be an important stage of mPTP regulation in mitochondria, revealing a new function of CNPase outside of myelin structure.


Asunto(s)
2',3'-Nucleótido Cíclico 3'-Fosfodiesterasa/química , Encéfalo/metabolismo , Calcio/química , Mitocondrias/química , 2',3'-Nucleótido Cíclico 3'-Fosfodiesterasa/metabolismo , Animales , Química Encefálica , Calcio/metabolismo , Mitocondrias/metabolismo , Fosfoproteínas , Fosforilación , Ratas
14.
Arch Biochem Biophys ; 558: 87-94, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-24995971

RESUMEN

Ca(2+)-induced permeability transition pore (mPTP) opening in isolated rat brain mitochondria is promoted through targeting of connexin43. After a threshold Ca(2+) load, mitochondrial membrane potential drops and efflux of accumulated Ca(2+) from the mitochondrial matrix occurs, indicating the mPTP opening. Specific antibodies were used to assess the role of the translocator protein (18kDa; TSPO) and connexin43 in swelling of isolated rat liver and brain mitochondria induced by carbenoxolone and the endogenous TSPO ligand protoporphyrin IX. Mitochondrial membrane potential, Ca(2+) transport and oxygen consumption were determined using selective electrodes. All the parameters were detected simultaneously in a chamber with the selective electrodes. The phosphorylation state of mitochondrial protein targets was assessed. We report that Ca(2+)-induced mitochondrial swelling was strengthened in the presence of both carbenoxolone and protoporphyrin IX. The carbenoxolone- and protoporphyrin IX-accelerated mPTP induction in brain mitochondria was completely prevented by antibodies specific for the mitochondrial translocator protein (TSPO). The anti-TSPO antibodies were more effective than anti-сonnexin43 antibodies. Moreover, carbenoxolone-stimulated phosphorylation of mitochondrial proteins was inhibited by anti-TSPO antibodies. Taken together, the data suggests that, in addition to acting via connexion43, carbenoxolone may exert its effect on mPTP via mitochondrial outer membrane TSPO.


Asunto(s)
Carbenoxolona/farmacología , Proteínas Portadoras/metabolismo , Conexina 43/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/química , Receptores de GABA-A/metabolismo , Animales , Encéfalo/citología , Calcio/farmacología , Hígado/citología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Fosforilación/efectos de los fármacos , Conformación Proteica/efectos de los fármacos , Protoporfirinas/farmacología , Ratas , Ratas Wistar
15.
Biol Chem ; 394(12): 1553-70, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24021597

RESUMEN

Overwhelming evidence shows that oxidative stress is a major cause in development of brain disorders. Low activity of the reactive oxygen species (ROS)-degrading system as well as high levels of oxidative damage markers have been observed in brain tissue of patients with neurodegenerative and other brain diseases to a larger extent than in healthy individuals. Many studies aimed to develop effective and safe antioxidant strategies for the therapy or prevention of brain diseases. Nevertheless, it became clear that rigorous suppression of ROS is deleterious for normal cell functioning. Thus, approaches that can regulate the ROS levels over a wide range, from inhibition to induction, will be a powerful tool for neuroprotection. A most prominent target for such ROS management is the family of peroxisome proliferator-activated receptors (PPARs). All three members (PPAR-α, -ß/δ and -γ) of this nuclear receptor subfamily form a tightly connected triad. For individual PPAR isoforms, neuroprotective properties have been well proven. Their involvement in regulation of ROS production and degradation underlies the therapeutic effects. Nevertheless, the current paradigms of the involvement of PPAR in neuroprotective therapy ignore such interconnections of PPARs and aim at antioxidant effects of individual PPAR isoforms, but do not take into account the necessity of careful regulation of ROS levels. The present review (i) summarizes the data, which support the concept of the PPAR triad in brain, (ii) demonstrates that usage of the PPAR triad allows the regulation of PPAR-dependent genes over a wide range, from inhibition to upregulation, and (iii) summarizes the known data concerning the PPAR triad involvement in regulation of ROS. Our report opens new directions in the field of PPAR/ROS-related neuroscience research.


Asunto(s)
Encéfalo/metabolismo , Receptores Activados del Proliferador del Peroxisoma/fisiología , Especies Reactivas de Oxígeno/metabolismo , Animales , Humanos , PPAR alfa/fisiología , PPAR delta/fisiología , PPAR gamma/fisiología , PPAR-beta/fisiología , Transducción de Señal
16.
Bioorg Med Chem ; 20(18): 5483-95, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22901672

RESUMEN

P2Y(6) nucleotide receptor (P2Y(6)-R) plays important physiological roles, such as insulin secretion and reduction of intraocular pressure. However, this receptor is still lacking potent and selective agonists to be used as potential drugs. Here, we synthesized uracil nucleotides and dinucleotides, substituted at the C5 and/or P(α) position with methoxy and/or borano groups, 18-22. Compound 18A, R(p) isomer of 5-OMe-UDP(α-B), is the most potent and P2Y(6)-R selective agonist currently known (EC(50) 0.008 µM) being 19-fold more potent than UDP and showing no activity at uridine nucleotide receptors, P2Y(2)- and P2Y(4)-R. Analogue 18A was highly chemically stable under conditions mimicking gastric juice acidity (t(1/2) = 16.9 h). It was more stable to hydrolysis by nucleotide pyrophosphatases (NPP1,3) than UDP (15% and 28% hydrolysis by NPP1 and NPP3, respectively, vs 50% and 51% hydrolysis of UDP) and metabolically stable in blood serum (t(1/2) = 17 vs 2.4, 11.9, and 21 h for UDP, 5-OMe-UDP, and UDP(α-B), respectively). This newly discovered highly potent and physiologically stable P2Y(6)-R agonist may be of future therapeutic potential.


Asunto(s)
Receptores Purinérgicos P2/metabolismo , Uridina Trifosfato/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad , Uridina Trifosfato/síntesis química , Uridina Trifosfato/química
17.
Am J Physiol Cell Physiol ; 300(3): C542-9, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21178110

RESUMEN

Many Ca(2+)-regulated intracellular processes are involved in the development of neuroinflammation. However, the changes of Ca(2+) signaling in the brain under inflammatory conditions were hardly studied. ATP-induced Ca(2+) signaling is a central event of signal transmission in astrocytic networks. We investigated primary astrocytes after proinflammatory stimulation with lipopolysaccharide (LPS; 100 ng/ml) for 6-24 h. We reveal that Ca(2+) responses to purinergic ATP stimulation are significantly increased in amplitude and duration after stimulation with LPS. We detected that increased amplitudes of Ca(2+) responses to ATP in LPS-treated astrocytes can be explained by substantial increase of Ca(2+) load in stores in endoplasmic reticulum. The mechanism implies enhanced Ca(2+) store refilling due to the amplification of capacitative Ca(2+) entry. The reason for the increased duration of Ca(2+) responses in LPS-treated cells is also the amplified capacitative Ca(2+) entry. Next, we established that the molecular mechanism for the LPS-induced amplification of Ca(2+) responses in astrocytes is increased expression and activity of VIA phospholipase A(2) (VIA iPLA(2)). Indeed, both gene silencing with specific small interfering RNA and pharmacological inhibition of VIA iPLA(2) with S-bromoenol lactone reduced the load of the Ca(2+) stores and caused a decrease in the amplitudes of Ca(2+) responses in LPS-treated astrocytes to values, which were comparable with those in untreated cells. Our findings highlight a novel regulatory role of VIA iPLA(2) in development of inflammation in brain. We suggest that this enzyme might be a possible target for treatment of pathologies related to brain inflammation.


Asunto(s)
Astrocitos/enzimología , Señalización del Calcio/fisiología , Gliosis/enzimología , Fosfolipasas A2 Grupo VI/metabolismo , Mediadores de Inflamación/farmacología , Lipopolisacáridos/farmacología , Regulación hacia Arriba/fisiología , Animales , Animales Recién Nacidos , Calcio/fisiología , Células Cultivadas , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Regulación Enzimológica de la Expresión Génica/fisiología , Silenciador del Gen/fisiología , Gliosis/patología , Fosfolipasas A2 Grupo VI/antagonistas & inhibidores , Fosfolipasas A2 Grupo VI/genética , Ratas , Regulación hacia Arriba/genética
18.
Am J Physiol Cell Physiol ; 300(3): C707-20, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21148408

RESUMEN

Carbenoxolone (Cbx), a substance from medicinal licorice, is used for antiinflammatory treatments. We investigated the mechanism of action of Cbx on Ca(2+)-induced permeability transition pore (PTP) opening in synaptic and nonsynaptic rat brain mitochondria (RBM), as well as in rat liver mitochondria (RLM), in an attempt to identify the molecular target of Cbx in mitochondria. Exposure to threshold Ca(2+) load induced PTP opening, as seen by sudden Ca(2+) efflux from the mitochondrial matrix and membrane potential collapse. In synaptic RBM, Cbx (1 µM) facilitated the Ca(2+)-induced, cyclosporine A-sensitive PTP opening, while in nonsynaptic mitochondria the Cbx threshold concentration was higher. A well-known molecular target of Cbx is the connexin (Cx) family, gap junction proteins. Moreover, Cx43 was previously found in heart mitochondria and attributed to the preconditioning mechanism of protection. Thus, we hypothesized that Cx43 might be a target for Cbx in brain mitochondria. For the first time, we detected Cx43 by Western blot in RBM, but Cx43 was absent in RLM. Interestingly, two anti-Cx43 antibodies, directed against amino acids 252 to 270 of rat Cx43, abolished the Cbx-induced enhancement of PTP opening in total RBM and in synaptic mitochondria, but not in RLM. In total RBM and in synaptic mitochondria, PTP caused dephosphorylation of Cx43 at serine 368. The phosphorylation level of serine 368 was decreased at threshold calcium concentration and additionally in the combined presence of Cbx in synaptic mitochondria. In conclusion, active mitochondrial Cx43 appears to counteract the Ca(2+)-induced PTP opening and thus might inhibit the PTP-ensuing mitochondrial demise and cell death. Consequently, we suggest that activity of Cx43 in brain mitochondria represents a novel molecular target for protection.


Asunto(s)
Encéfalo/metabolismo , Señalización del Calcio/fisiología , Carbenoxolona/farmacología , Conexina 43/metabolismo , Membranas Intracelulares/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/fisiología , Animales , Antiulcerosos/farmacología , Encéfalo/ultraestructura , Señalización del Calcio/efectos de los fármacos , Membranas Intracelulares/efectos de los fármacos , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/fisiología , Poro de Transición de la Permeabilidad Mitocondrial , Permeabilidad/efectos de los fármacos , Ratas , Ratas Wistar
19.
J Neurochem ; 118(3): 354-64, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21615407

RESUMEN

We previously demonstrated that αB-crystallin and protease-activated receptor (PAR) are involved in protection of astrocytes against C2-ceramide- and staurosporine-induced cell death [Li et al. (2009) J. Neurochem.110, 1433-1444]. Here, we further investigated the mechanism of cytoprotection by αB-crystallin. Our current data revealed that after down-regulation of αB-crystallin by siRNA, cell death caused by C2-ceramide and staurosporine is increased. Furthermore, we investigated the mechanism of cytoprotection of astrocytes by intracellular αB-crystallin. Application of specific inhibitors of p38 and extracellular regulated kinase (ERK) abrogates the protection of astrocytes by over-expression of αB-crystallin. Thus, p38 and ERK contribute to protective processes by αB-crystallin. To reveal the molecular mechanism of αB-crystallin-mediated cytoprotection, we mimicked phosphorylation or unphosphorylation of αB-crystallin. In these experiments, we found that the phosphorylation of αB-crystallin at Ser45 and Ser59 is required for protection. Ser19 phosphorylation of αB-crystallin does not contribute to protection. Moreover, we detected that PAR-2 activation increases the phosphorylation level of αB-crystallin at Ser59, but does not affect the expression level of αB-crystallin. Thus, endogenous αB-crystallin has protective capacity employing a mechanism, which involves regulation of the phosphorylation status of αB-crystallin and p38 and ERK activity. Moreover, we report that PAR-2 activation evokes the phosphorylation of αB-crystallin to increase astrocytes survival.


Asunto(s)
Astrocitos/efectos de los fármacos , Fármacos Neuroprotectores , Serina/química , Esfingosina/análogos & derivados , Estaurosporina/antagonistas & inhibidores , Estaurosporina/toxicidad , Cadena B de alfa-Cristalina/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Animales , Western Blotting , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Fosforilación , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Receptor PAR-2/genética , Receptor PAR-2/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esfingosina/antagonistas & inhibidores , Esfingosina/toxicidad , Cadena B de alfa-Cristalina/química , Proteínas Quinasas p38 Activadas por Mitógenos/química
20.
J Neurochem ; 119(6): 1194-204, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21988180

RESUMEN

Challenge of protease-activated receptors induces cytosolic Ca(2+) concentration ([Ca(2+) ](c)) increase, mitogen-activated protein kinase activation and reactive oxygen species (ROS) formation with a bandwidth of responses in individual cells. We detected in this study in situ the thrombin-induced [Ca(2+) ](c) rise and ROS formation in dissociated hippocampal astrocytes and neurons in a mixed culture. In identified cells, single cell responses were correlated with extracellular-regulated kinase (ERK)1/2 phosphorylation level. On average, in astrocytes, thrombin induced a transient [Ca(2+) ](c) rise with concentration-dependent increase in amplitude and extrusion rate and high ERK1/2 phosphorylation level. Correlation analysis of [Ca(2+) ](c) response characteristics of single astrocytes reveals that astrocytes with nuclear phosphoERK1/2 localization have a smaller Ca(2+) amplitude and extrusion rate compared with cells with a cytosolic phosphoERK1/2 localization. In naive neurons, without thrombin challenge, variable ERK1/2 phosphorylation patterns are observed. ROS were detected by hydroethidine. Only in neurons with increased ERK1/2 phosphorylation level, we see sustained intracellular rise in fluorescence of the dye lasting over several minutes. ROS formation was abolished by pre-incubation with the NADPH oxidase inhibitor apocynin. Additionally, thrombin induced an immediate, transient hydroethidine fluorescence increase. This was interpreted as NADPH oxidase-mediated O(2) (•-) -release into the extracellular milieu, because it was decreased by pre-incubation with apocynin, and could be eluted by superfusion. In conclusion, the phosphorylation status of ERK1/2 determines the thrombin-dependent [Ca(2+) ](c) increase and ROS formation and, thus, influences the capacity of thrombin to regulate neuroprotection or neurodegeneration.


Asunto(s)
Astrocitos/metabolismo , Calcio/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hipocampo/citología , Neuronas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Proteína Ácida Fibrilar de la Glía/metabolismo , Hemostáticos/farmacología , Neuronas/efectos de los fármacos , Fosfopiruvato Hidratasa/metabolismo , Fosforilación/efectos de los fármacos , Cloruro de Potasio/farmacología , Ratas , Trombina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA