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










Base de datos
Intervalo de año de publicación
1.
Neurochem Int ; 148: 105050, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33945834

RESUMEN

Neurons spurn hydrogen-rich fatty acids for energizing oxidative ATP synthesis, contrary to other cells. This feature has been mainly attributed to a lower yield of ATP per reduced oxygen, as compared to glucose. Moreover, the use of fatty acids as hydrogen donor is accompanied by severe ß-oxidation-associated ROS generation. Neurons are especially susceptible to detrimental activities of ROS due to their poor antioxidative equipment. It is also important to note that free fatty acids (FFA) initiate multiple harmful activities inside the cells, particularly on phosphorylating mitochondria. Several processes enhance FFA-linked lipotoxicity in the cerebral tissue. Thus, an uptake of FFA from the circulation into the brain tissue takes place during an imbalance between energy intake and energy expenditure in the body, a situation similar to that during metabolic syndrome and fat-rich diet. Traumatic or hypoxic brain injuries increase hydrolytic degradation of membrane phospholipids and, thereby elevate the level of FFA in neural cells. Accumulation of FFA in brain tissue is markedly associated with some inherited neurological disorders, such as Refsum disease or X-linked adrenoleukodystrophy (X-ALD). What are strategies protecting neurons against FFA-linked lipotoxicity? Firstly, spurning the ß-oxidation pathway in mitochondria of neurons. Secondly, based on a tight metabolic communication between neurons and astrocytes, astrocytes donate metabolites to neurons for synthesis of antioxidants. Further, neuronal autophagy of ROS-emitting mitochondria combined with the transfer of degradation-committed FFA for their disposal in astrocytes, is a potent protective strategy against ROS and harmful activities of FFA. Finally, estrogens and neurosteroids are protective as triggers of ERK and PKB signaling pathways, consequently initiating the expression of various neuronal survival genes via the formation of cAMP response element-binding protein (CREB).


Asunto(s)
Encéfalo/metabolismo , Ácidos Grasos/toxicidad , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Encéfalo/efectos de los fármacos , Metabolismo Energético/fisiología , Ácidos Grasos/metabolismo , Humanos , Mitocondrias/efectos de los fármacos , Neuronas/metabolismo , Estrés Oxidativo/fisiología
2.
Neurochem Int ; 131: 104513, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31369777

RESUMEN

Astrocytes play a vital role in regulating central nervous system inflammation, energy metabolism and brain homeostasis. Unlike macrophages and microglia, which are cells of myeloid ancestry, astrocytes are of ectodermal origin. However, regulatory specificities of signaling pathways connecting inflammatory and metabolic processes are still largely unknown. We analyzed firstly cellular responses to toll-like receptor (TLR) agonists and secondly, modulation of the mRNA of the three isoforms of the transcription factors PPARs (peroxisome proliferator-activated receptors) in primary rat astrocytes exposed to normal glucose (5.5 mM) and high glucose (25 mM). Cell culturing of rat brain astrocytes for 2 days in high glucose did not alter cellular morphology, but i) enhanced the release of TNFα that was induced by TLR4 agonist LPS or TLR3 agonist PIC and the synthesis of prostaglandin E2 (PGE2), ii) changed the signaling pathways of TLR4/MAPK (increase in p38 MAPK, and decrease in JNK activities at early stages of TLR activation) and iii) modulated mRNA expression of PPARs. High glucose cultivation reduced PPARα and PPARß mRNA levels, without altering PPARγ mRNA level and changed the sensitivity of expressions to agonists of TLR1/2 (PGN), TLR4 (LPS), TLR3 (PIC), and TLR5 (FGN). Differences between low and high glucose-adapted cells were obtained for agonists of TLR1/2 (PPARα, PPARß), TLR4 (PPAR ß), TLR3 (PPARα). In the TLR4/p38/PPARß signaling pathway, there was a stimulatory connection in normal glucose but an inhibitory connection in high glucose. TLR4/JNK/activated PPARß, TLR4/JNK/inhibited PPARγ both in cells adapted to normal or high glucose, but PPARα expression was not affected. As PPARs in astrocytes are involved in inflammatory processes in the form of the recently published PPAR triad, the changes in expression revealed here are most likely resulting in implications of high glucose in inflammatory processes. Our data underline the complexity of multiple regulatory interactions between inflammatory responses and energy metabolism in astrocytes.


Asunto(s)
Astrocitos/fisiología , Glucosa/farmacología , Sistema de Señalización de MAP Quinasas/fisiología , Receptores Toll-Like/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Animales , Astrocitos/efectos de los fármacos , Células Cultivadas , Medios de Cultivo , Metabolismo Energético/efectos de los fármacos , Lipopolisacáridos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , PPAR alfa/metabolismo , PPAR gamma/metabolismo , Cultivo Primario de Células , Ratas , Receptor Toll-Like 4/agonistas , Receptores Toll-Like/agonistas , Factor de Necrosis Tumoral alfa/metabolismo
3.
Neurochem Int ; 118: 82-90, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29702146

RESUMEN

Control of decay of mRNA containing the adenine-uridine rich elements (AREs) is an important post-transcriptional mechanism involved in the regulation of inflammatory gene expression. Two widely recognized proteins in this machinery are HuR (human antigen R) - a protein that stabilizes ARE-containing mRNA and TTP (tristetraprolin) - a protein that shortens half-lives of ARE-containing mRNA. Although HuR and TTP regulation mechanisms have been well studied in cells of hematopoietic origin, there are no respective data in astrocytes, cells of ectodermal origin which play an important role in neuroinflammation. Therefore we evaluated the existence of TTP and HuR in primary astrocytes and characterized the features of their regulation after stimulation by the proinflammatory stimuli thrombin, ATP, and agonists of TLR4, TLR2. All proinflammatory stimuli increased levels of TTP mRNA, but not HuR mRNA. Transcripts of both HuR and TTP underwent stabilization upon lipopolysaccharide (LPS) treatment, measured with the actinomycin D protocol. This effect was abolished by treatment with SB203580, an inhibitor of р38 МАРК. Both TTP and HuR transcripts were sensitive to modulation by anisomycin and cycloheximide. LPS induced translocation of HuR protein from nucleus to cytoplasm. TTP is localized in the cytosolic fraction and localization is not sensitive to LPS treatment. Our data for the first time reveal specificity of regulation of ARE-binding proteins in astrocytes. We propose possibilities to manipulate brain inflammatory processes via post-transcription regulatory steps in astrocytes.


Asunto(s)
Astrocitos/metabolismo , Proteínas Portadoras/metabolismo , Proteína 1 Similar a ELAV/metabolismo , Tristetraprolina/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Células Cultivadas , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , Lipopolisacáridos/toxicidad , Ratones , Células RAW 264.7 , Ratas Wistar
4.
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
5.
Neurochem Int ; 115: 69-79, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29425965

RESUMEN

Small heat shock proteins (sHsps) are a group of proteins with molecular mass between 12 and 43 kDa. Currently, 11 members of this family have been classified, namely HspB1 to HspB11. HspB1, HspB2, HspB5, HspB6, HspB7, and HspB8, which are expressed in brain have been observed to be related to the pathology of neurodegenerative diseases, including Parkinson's, Alzheimer's, Alexander's disease, multiple sclerosis, and human immunodeficiency virus-associated dementia. Specifically, sHsps interact with misfolding and damaging protein aggregates, like Glial fibrillary acidic protein in AxD, ß-amyloid peptides aggregates in Alzheimer's disease, Superoxide dismutase 1 in Amyotrophic lateral sclerosis and cytosine-adenine-guanine/polyglutamine (CAG/PolyQ) in Huntington's disease, Spinocerebellar ataxia type 3, Spinal-bulbar muscular atrophy, to reduce the toxicity or increase the clearance of these protein aggregates. The degree of HspB4 expression in brain is still debated. For neuroprotective mechanisms, sHsps attenuate mitochondrial dysfunctions, reduce accumulation of misfolded proteins, block oxidative/nitrosative stress, and minimize neuronal apoptosis and neuroinflammation, which are molecular mechanisms commonly accepted to mirror the progression and development of neurodegenerative diseases. The increasing incidence of the neurodegenerative diseases enhanced search for effective approaches to rescue neural tissue from degeneration with minimal side effects. sHsps have been found to exert neuroprotective functions. HspB5 has been emphasized to reduce the paralysis in a mouse model of experimental autoimmune encephalomyelitis, providing a therapeutic basis for the disease. In this review, we discuss the current understanding of the properties and the mechanisms of protection orchestrated by sHsps in the nervous system, highlighting the promising therapeutic role of sHsps in neurodegenerative diseases.


Asunto(s)
Proteínas de Choque Térmico/efectos de los fármacos , Enfermedades Neurodegenerativas/fisiopatología , Cadena B de alfa-Cristalina/genética , Animales , Encéfalo/metabolismo , Proteínas de Choque Térmico HSP27 , Proteínas de Choque Térmico/metabolismo , Proteínas de Choque Térmico Pequeñas/metabolismo , Ratones , Péptidos/farmacología
6.
Neurochem Int ; 109: 68-77, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28366720

RESUMEN

The brain uses long-chain fatty acids (LCFAs) to a negligible extent as fuel for the mitochondrial energy generation, in contrast to other tissues that also demand high energy. Besides this generally accepted view, some studies using cultured neural cells or whole brain indicate a moderately active mitochondrial ß-oxidation. Here, we corroborate the conclusion that brain mitochondria are unable to oxidize fatty acids. In contrast, the combustion of liver-derived ketone bodies by neural cells is long-known. Furthermore, new insights indicate the use of odd-numbered medium-chain fatty acids as valuable source for maintaining the level of intermediates of the citric acid cycle in brain mitochondria. Non-esterified LCFAs or their activated forms exert a large variety of harmful side-effects on mitochondria, such as enhancing the mitochondrial ROS generation in distinct steps of the ß-oxidation and therefore potentially increasing oxidative stress. Hence, the question arises: Why do in brain energy metabolism mitochondria selectively spurn LCFAs as energy source? The most likely answer are the relatively higher content of peroxidation-sensitive polyunsaturated fatty acids and the low antioxidative defense in brain tissue. There are two remarkable peroxisomal defects, one relating to α-oxidation of phytanic acid and the other to uptake of very long-chain fatty acids (VLCFAs) which lead to pathologically high tissue levels of such fatty acids. Both, the accumulation of phytanic acid and that of VLCFAs give an enlightening insight into harmful activities of fatty acids on neural cells, which possibly explain why evolution has prevented brain mitochondria from the equipment with significant ß-oxidation enzymatic capacity.


Asunto(s)
Encéfalo/metabolismo , Metabolismo Energético/fisiología , Ácidos Grasos/metabolismo , Mitocondrias/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Estrés Oxidativo/fisiología , Animales , Humanos , Especies Reactivas de Oxígeno/metabolismo
7.
Neurochem Int ; 108: 121-132, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28279750

RESUMEN

INAD (infantile neuroaxonal dystrophy, OMIM#256600), an autosomal recessive inherited degenerative disease, is associated with PLA2G6 mutations. PLA2G6 encodes Ca2+-independent phospholipase A2 (VIA iPLA2). However, it is unclear how the PLA2G6-mutations lead to disease. Non-canonical functions, which were suggested for VIA iPLA2, such as regulation of cellular and mitochondrial Ca2+ are promising candidates. Therefore, we investigate glutamate (Glu)-evoked Ca2+ signals in neurons and astrocytes in co-culture obtained from three INAD mouse model strains with Pla2g6 mutations, (i) hypomorphic Pla2g6 allele with reduced transcript levels, (ii) knocked-out Pla2g6, and (iii) (G373R)-point mutation with inactive VIA iPLA2 enzyme. Homozygous offspring from these strains develop pathology similar to that observed in INAD patients. We found that in mouse neurons the Pla2g6 mutation disrupted the dependency of Glu-induced extracellular Ca2+ influx on mitochondrial Ca2+ uptake. Thus, in neurons with Pla2g6 mutation we did not detect the characteristic reduction in Glu-induced Ca2+ influx upon treatment with Ru360, a blocker of mitochondrial Ca2+ uniporter, or with rotenone. In contrast to neurons, in astrocytes, both with Pla2g6 mutation or wild-type cells, the treatment with Ru360 or rotenone reduced the rate of Glu-induced Ca2+ influx ∼2-fold. This Ca2+ influx in astrocytes represents capacitative Ca2+ entry. In astrocytes with Pla2g6 mutation, the Glu-induced Ca2+ influx was ∼2-fold lower than in wild-type controls. We suggest that this is the mechanism for strongly decreased durations of Glu-induced Ca2+ responses in astrocytes with Pla2g6 mutation. We could mimic the mutation by pharmacological inhibition of iPLA2 using S-BEL. Thus, lack of VIA iPLA2 activity caused effects in astrocytes. In summary, three INAD mouse models show comparable changes in Glu-induced Ca2+ signaling, but specific for neurons or astrocytes. This finding helps to identify pathways altered during INAD and highlights non-canonical VIA iPLA2 functions, like regulation of cellular Ca2+ fluxes by mitochondria or capacitative Ca2+-entry.


Asunto(s)
Astrocitos/metabolismo , Señalización del Calcio/fisiología , Modelos Animales de Enfermedad , Ácido Glutámico/toxicidad , Distrofias Neuroaxonales/metabolismo , Neuronas/metabolismo , Animales , Astrocitos/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Técnicas de Cocultivo , Femenino , Fosfolipasas A2 Grupo VI/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Distrofias Neuroaxonales/inducido químicamente , Distrofias Neuroaxonales/genética , Neuronas/efectos de los fármacos
8.
Restor Neurol Neurosci ; 35(1): 87-103, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28059802

RESUMEN

The already established and widely used intravenous application of recombinant tissue plasminogen activator as a re-opening strategy for acute vessel occlusion in ischemic stroke was recently added by mechanical thrombectomy, representing a fundamental progress in evidence-based medicine to improve the patient's outcome. This has been paralleled by a swift increase in our understanding of pathomechanisms underlying many neurovascular diseases and most prevalent forms of dementia. Taken together, these current advances offer the potential to overcome almost two decades of marginally successful translational research on stroke and dementia, thereby spurring the entire field of translational neuroscience. Moreover, they may also pave the way for the renaissance of classical neuroprotective paradigms.This review reports and summarizes some of the most interesting and promising recent achievements in neurovascular and dementia research. It highlights sessions from the 9th International Symposium on Neuroprotection and Neurorepair that have been discussed from April 19th to 22nd in Leipzig, Germany. To acknowledge the emerging culture of interdisciplinary collaboration and research, special emphasis is given on translational stories ranging from fundamental research on neurode- and -regeneration to late stage translational or early stage clinical investigations.


Asunto(s)
Trastornos Cerebrovasculares/diagnóstico , Trastornos Cerebrovasculares/terapia , Enfermedades Neurodegenerativas/diagnóstico , Enfermedades Neurodegenerativas/terapia , Investigación Biomédica Traslacional , Animales , Humanos
9.
J Cereb Blood Flow Metab ; 37(3): 848-854, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27055779

RESUMEN

According to recent reports, systemic treatment of rats with methylpalmoxirate (carnitine palmitoyltransferase-1 inhibitor) decreased peroxidation of polyunsaturated fatty acids in brain tissue. This was taken as evidence of mitochondrial ß-oxidation in brain, thereby contradicting long-standing paradigms of cerebral metabolism, which claim that ß-oxidation of activated fatty acids has minor importance for brain energy homeostasis. We addressed this controversy. Our experiments are the first direct experimental analysis of this question. We fueled isolated brain mitochondria or rat brain astrocytes with octanoic acid, but octanoic acid does not enhance formation of reactive oxygen species, neither in isolated brain mitochondria nor in astrocytes, even at limited hydrogen delivery to mitochondria. Thus, octanoic acid or l-octanoylcarnitine does not stimulate H2O2 release from brain mitochondria fueled with malate, in contrast to liver mitochondria (2.25-fold rise). This does obviously not support the possible occurrence of ß-oxidation of the fatty acid octanoate in the brain. We conclude that a proposed inhibition of ß-oxidation does not seem to be a helpful strategy for therapies aiming at lowering oxidative stress in cerebral tissue. This question is important, since oxidative stress is the cause of neurodegeneration in numerous neurodegenerative or inflammatory disease situations.


Asunto(s)
Encéfalo/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Animales , Caprilatos/metabolismo , Carnitina/análogos & derivados , Carnitina/metabolismo , Metabolismo Energético , Homeostasis , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Ratas
10.
Neurochem Int ; 99: 187-193, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27395788

RESUMEN

Mutations in the PLA2G6 gene which encodes Ca(2+)-independent phospholipase A2 (VIA iPLA2) were detected in 85% of cases of the inherited degenerative nervous system disorder INAD (infantile neuroaxonal dystrophy, OMIM #256600). However, molecular mechanisms linking these mutations to the disease progression are unclear. VIA iPLA2 is expressed also in mitochondria. Here, we investigate Ca(2+) handling by brain mitochondria derived from mice with hypomorph Pla2g6 allele. These animals with reduced transcript levels (5% of wild type) represent a suitable model for INAD. We demonstrated significant reduction of Ca(2+) uptake rate and Ca(2+) retention capacity in brain mitochondria isolated from this mutant. This phenotype could be mimicked when in wild-type controls VIA iPLA2 was inhibited by S-BEL. Importantly, the reduction could be ameliorated partly by addition of the VIA iPLA2 product, sn-2 lysophosphatidyl-choline. Furthermore, we demonstrated in situ a reduced mitochondrial potential in neurons from mice deficient in VIA iPLA2, which could cause the reduced Ca(2+) uptake rate via the potential-dependent mitochondrial Ca(2+) uniporter. Thus, the disturbances in mitochondrial potential and the changes in Ca(2+) handling were dependent on VIA iPLA2 activity. Reduced mitochondrial Ca(2+) uptake rate and Ca(2+) retention capacity might result in increased vulnerability of mitochondria to the Ca(2+) overload and in disturbed cellular Ca(2+) signaling during INAD. For VIA iPLA2, non-canonical functions beyond sole phospholipid turnover seem to be important, such as regulation of store-operated Ca(2+) entry in cells. Thus, our findings bring new insight into molecular mechanism affected in INAD and highlight the non-canonical function of VIA iPLA2 in regulation of mitochondrial Ca(2+) handling.


Asunto(s)
Señalización del Calcio/fisiología , Modelos Animales de Enfermedad , Fosfolipasas A2 Grupo VI/metabolismo , Mitocondrias/metabolismo , Distrofias Neuroaxonales/metabolismo , Animales , Fosfolipasas A2 Grupo VI/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/genética , Distrofias Neuroaxonales/genética , Ratas , Ratas Wistar
12.
Aging Dis ; 7(2): 136-49, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27114847

RESUMEN

It is increasingly understood that in the aging brain, especially in the case of patients suffering from neurodegenerative diseases, some fatty acids at pathologically high concentrations exert detrimental activities. To study such activities, we here analyze genetic diseases, which are due to compromised metabolism of specific fatty acids, either the branched-chain phytanic acid or very long-chain fatty acids (VLCFAs). Micromolar concentrations of phytanic acid or of VLCFAs disturb the integrity of neural cells by impairing Ca(2+) homeostasis, enhancing oxidative stress or de-energizing mitochondria. Finally, these combined harmful activities accelerate cell death. Mitochondria are more severely targeted by phytanic acid than by VLCFAs. The insertion of VLCFAs into the inner membrane distorts the arrangement of membrane constituents and their functional interactions. Phytanic acid exerts specific protonophoric activity, induces reactive oxygen species (ROS) generation, and reduces ATP generation. A clear inhibition of the Na(+), K(+)-ATPase activity by phytanic acid has also been reported. In addition to the instantaneous effects, a chronic exposure of brain cells to low micromolar concentrations of phytanic acid may produce neuronal damage in Refsum disease by altering epigenetic transcriptional regulation. Myelin-producing oligodendrocytes respond with particular sensitivity to VLCFAs. Deleterious activity of VLCFAs on energy-dependent mitochondrial functions declines with increasing the hydrocarbon chain length (C22:0 > C24:0 > C26:0). In contrast, the reverse sequence holds true for cell death induction by VLCFAs (C22:0 < C24:0 < C26:0). In adrenoleukodystrophy, the uptake of VLCFAs by peroxisomes is impaired by defects of the ABCD1 transporter. Studying mitochondria from ABCD1-deficient and wild-type mice proves that the energy-dependent functions are not altered in the disease model. Thus, a defective ABCD1 apparently exerts no obvious adaptive pressure on mitochondria. Further research has to elucidate the detailed mechanistic basis for the failures causing fatty acid-mediated neurodegeneration and should help to provide possible therapeutic interventions.

13.
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
14.
Neurochem Int ; 94: 57-66, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26898403

RESUMEN

Consequences of neurodegenerative diseases or stroke also depend on astroglial survival during oxidative stress. P2Y receptors that are widely distributed in the central nervous system are suggested to be involved in cytoprotection. However, knowledge about the efficacy of protection by P2Y receptors and their involvement in antioxidant protective pathways is scarce. Here, we investigate the viability and reactive oxygen species (ROS) production after exposure of rat astrocytes to hydrogen peroxide. We determined the influence of treatment with the P2Y1 receptor-specific agonist 2-methyl-thio-ADP (2MeSADP) and the broad range P2Y receptor agonist adenosine 5'-(3-thiotriphosphate) (ATPγS). Preincubation (24-h before hydrogen peroxide application) and incubation with ATPγS and 2MeSADP protected astrocytes. The ROS production in hydrogen peroxide-treated astrocytes was reduced by pre- and co-incubation with ATPγS or 2MeSADP. Changes of levels of expression of antioxidant defense systems in astrocytes by treatment with P2Y receptor agonists were analyzed. Incubation with ATPγS and 2MeSADP increased mRNA levels of CAT encoding catalase and SOD2, encoding mitochondrial manganese dependent superoxide dismutase. ATPγS additionally increased mRNA levels of SOD3, encoding extracellular superoxide dismutase (ECSOD). Levels of total glutathione (GSH) increased in ATPγS/2MeSADP-treated astrocytes. mRNA levels of genes involved in GSH synthesis and in import of GSH precursors were analyzed after treatment with ATPγS and 2MeSADP. Both agonists significantly increased mRNA levels of a subunit of glutamate cysteine ligase, and a subunit of antiporter system xc(-). Changes in mRNA levels of antioxidant enzymes and genes of GSH metabolism depend on rise of intracellular Ca(2+) by P2Y receptor and basal activity of protein kinase A (PKA). SOD3 induction is suggested to depend on increased intracellular Ca(2+), increased cyclic AMP levels and PKA activity. Thus, we confirm a role of purinergic signaling in astrocytic survival during oxidative stress by maintaining antioxidant defense, highlighting P2Y receptors as potential targets for cytoprotection.


Asunto(s)
Adenosina Difosfato/análogos & derivados , Adenosina Trifosfato/análogos & derivados , Antioxidantes/metabolismo , Peróxido de Hidrógeno/toxicidad , Fármacos Neuroprotectores/farmacología , Receptores Purinérgicos P2Y/biosíntesis , Tionucleótidos/farmacología , Adenosina Difosfato/farmacología , Adenosina Trifosfato/farmacología , Animales , Animales Recién Nacidos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Citoprotección/efectos de los fármacos , Citoprotección/fisiología , Nucleótidos/farmacología , Ratas
15.
J Med Chem ; 58(21): 8427-43, 2015 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-26447940

RESUMEN

With a view to identify novel and biocompatible neuroprotectants, we designed nucleoside 5'-thiophosphate analogues, 6-11. We identified 2-SMe-ADP(α-S), 7A, as a most promising neuroprotectant. 7A reduced ROS production in PC12 cells under oxidizing conditions, IC50 of 0.08 vs 21 µM for ADP. Furthermore, 7A rescued primary neurons subjected to oxidation, EC50 of 0.04 vs 19 µM for ADP. 7A is a most potent P2Y1-R agonist, EC50 of 0.0026 µM. Activity of 7A in cells involved P2Y1/12-R as indicated by blocking P2Y12-R or P2Y1-R. Compound 7A inhibited Fenton reaction better than EDTA, IC50 of 37 vs 54 µM, due to radical scavenging, IC50 of 12.5 vs 30 µM for ADP, and Fe(II)-chelation, IC50 of 80 vs >200 µM for ADP (ferrozine assay). In addition, 7A was stable in human blood serum, t1/2 of 15 vs 1.5 h for ADP, and resisted hydrolysis by NPP1/3, 2-fold vs ADP. Hence, we propose 7A as a highly promising neuroprotectant.


Asunto(s)
Antioxidantes/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Nucleósidos/farmacología , Nucleótidos/farmacología , Oligonucleótidos Fosforotioatos/farmacología , Animales , Antioxidantes/química , Células Cultivadas , Descubrimiento de Drogas , Humanos , Peróxido de Hidrógeno/antagonistas & inhibidores , Hidrólisis , Hierro , Modelos Moleculares , Neuronas/citología , Neuronas/metabolismo , Fármacos Neuroprotectores/química , Nucleósidos/química , Nucleótidos/química , Células PC12 , Oligonucleótidos Fosforotioatos/química , Agonistas del Receptor Purinérgico P2Y/química , Agonistas del Receptor Purinérgico P2Y/farmacología , Ratas , Especies Reactivas de Oxígeno/metabolismo , Receptores Purinérgicos P2Y1/metabolismo
16.
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
17.
Neurochem Int ; 90: 46-55, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26188334

RESUMEN

We previously reported that 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNP) is present in rat brain and liver mitochondria, in the outer membrane and mitoplasts. Substrates of CNP, 2',3'-cAMP and 2',3'-cNADP, were found to accelerate opening of mitochondrial permeability transition pore (mPTP). In purified non-synaptic mitochondria, CNP was observed to co-immunoprecipitate with main modulators of mPTP, i.e. VDAC, ANT, and cyclophilin D, as well as with tubulin and COX IV. Using Blue Native Electrophoresis, with following Western blot, CNP was revealed to associate with functional inner membrane mitochondrial complexes I-V. In Ca(2+) -overloaded mitochondria, association of CNP with complexes I-V was decreased. Cyclosporine A increased the association of CNP with complexes I and III, supporting the idea of the involvement of these complexes in mPTP function. 2',3'-cAMP enhanced CNP dissociation from complexes I, III, IV and V in Ca(2+)-overloaded mitochondria (i.e. when pore is opened). Association of CNP with complexes I, III, IV, and V was shown in mitochondria isolated from brain, liver and heart. Stimulation of the opening of the non-selective pore in mitochondria correlated with CNP release from mitochondria in parallel with release of cytochrome c, AIF and Endo G. In Ca(2+)-overloaded mitochondria, 2',3'-cAMP further accelerated the release of AIF, Endo G and CNP, but did not alter cytochrome c release. These results provide strong evidence that CNP, one of the possible regulators of mPTP complex, might be involved in the control of respiration and energy production in mitochondria. This reveals a new function of CNP outside the myelin structure.


Asunto(s)
2',3'-Nucleótido Cíclico 3'-Fosfodiesterasa/metabolismo , Apoptosis/fisiología , Calcio/metabolismo , Citocromos c/metabolismo , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Animales , Encéfalo/metabolismo , Mitocondrias Hepáticas/metabolismo , Membranas Mitocondriales/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Ratas
18.
Brain Res ; 1620: 17-28, 2015 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-25998538

RESUMEN

α-Crystallin with two isoforms, αA-crystallin (HSPB4) and αB-crystallin (HSPB5), is found in eye lens, spleen, lung, kidney, cornea, skin, but also in brain. Several studies revealed roles of αA/αB-crystallin in regulating cell viability and protection in the central nervous system. We previously demonstrated that α-crystallin serves as an intracellular protectant in astrocytes. Compared to well-studied intracellular functions of α-crystallin, there is limited proof for the role of α-crystallin as extracellular protectant. In order to clarify protective effects of extracellular αA/αB-crystallin, we exposed astrocytes to the toxic agents, staurosporine or C2-ceramide, or serum-starvation in the presence of αA/αB-crystallin. Extracellular αA/αB-crystallin protected astrocytes from staurosporine- and C2-ceramide-induced cell death. In addition, extracellular αB-crystallin/HSPB5 effectively promoted astrocytes viability through phosphatidylinositol 3 kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) and extracellular signal-regulated kinase 1/2 (ERK1/2), p38 mitogen-activated protein kinases (p38) and c-Jun N-terminal kinases (JNK) signaling pathways under serum-deprivation. Furthermore, αB-crystallin/HSPB5 decreases the staurosporine-mediated cleavage of caspase 3 through PI3K/Akt signaling preventing apoptosis of astrocytes. Thus, the current study indicates that extracellular αA/αB-crystallin protects astrocytes exposed to various harmful stimuli. Furthermore, application of αB-crystallin/HSPB5 to isolated rat brain mitochondria inhibits ROS generation induced by complex III inhibition with Antimycin A.


Asunto(s)
Astrocitos/fisiología , Muerte Celular/fisiología , Espacio Extracelular/metabolismo , Mitocondrias/metabolismo , alfa-Cristalinas/metabolismo , Animales , Antimicina A/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Escherichia coli , Espacio Extracelular/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Mitocondrias/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Esfingosina/análogos & derivados , Esfingosina/toxicidad , Estaurosporina/toxicidad , Serina-Treonina Quinasas TOR/metabolismo , alfa-Cristalinas/administración & dosificación
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...