232 resultados para Glucose transporter proteins
Resumo:
To report the case of a child with short absences and occasional myoclonias since infancy who was first diagnosed with an idiopathic generalized epilepsy, but was documented at follow-up to have a mild phenotype of glucose transporter type 1 deficiency syndrome. Unlike other reported cases of Glut-1 DS and epilepsy, this child had a normal development as well as a normal head growth and neurological examination. Early onset of seizures and later recognized episodes of mild confusion before meals together with persistent atypical EEG features and unexpected learning difficulties led to the diagnosis. Seizure control and neuropsychological improvements were obtained with a ketogenic diet.
Resumo:
Phosphoinositides, synthesized from myo-inositol, play a critical role in the development of growth cones and in synaptic activity. As neurons cannot synthesize inositol, they take it up from the extracellular milieu. Here, we demonstrate that, in brain and PC12 cells, the recently identified H(+)/myo-inositol symporter HMIT is present in intracellular vesicles that are distinct from synaptic and dense-core vesicles. We further show that HMIT can be triggered to appear on the cell surface following cell depolarization, activation of protein kinase C or increased intracellular calcium concentrations. HMIT cell surface expression takes place preferentially in regions of nerve growth and at varicosities and leads to increased myo-inositol uptake. The symporter is then endocytosed in a dynamin-dependent manner and becomes available for a subsequent cycle of stimulated exocytosis. HMIT is thus expressed in a vesicular compartment involved in activity-dependent regulation of myo-inositol uptake in neurons. This may be essential for sustained signaling and vesicular traffic activities in growth cones and at synapses.
Resumo:
The aim of our work was to show how a chosen normal-isation strategy can affect the outcome of quantitative gene expression studies. As an example, we analysed the expression of three genes known to be upregulated under hypoxic conditions: HIF1A, VEGF and SLC2A1 (GLUT1). Raw RT-qPCR data were normalised using two different strategies: a straightforward normalisation against a single reference gene, GAPDH, using the 2(-ΔΔCt) algorithm and a more complex normalisation against a normalisation factor calculated from the quantitative raw data from four previously validated reference genes. We found that the two different normalisation strategies revealed contradicting results: normalising against a validated set of reference genes revealed an upregulation of the three genes of interest in three post-mortem tissue samples (cardiac muscle, skeletal muscle and brain) under hypoxic conditions. Interestingly, we found a statistically significant difference in the relative transcript abundance of VEGF in cardiac muscle between donors who died of asphyxia versus donors who died from cardiac death. Normalisation against GAPDH alone revealed no upregulation but, in some instances, a downregulation of the genes of interest. To further analyse this discrepancy, the stability of all reference genes used were reassessed and the very low expression stability of GAPDH was found to originate from the co-regulation of this gene under hypoxic conditions. We concluded that GAPDH is not a suitable reference gene for the quantitative analysis of gene expression in hypoxia and that validation of reference genes is a crucial step for generating biologically meaningful data.
Resumo:
AbstractType 2 diabetes (T2D) is a metabolic disease which affects more than 200 millions people worldwide. The progression of this affection reaches nowadays epidemic proportions, owing to the constant augmentation in the frequency of overweight, obesity and sedentary. The pathogenesis of T2D is characterized by reduction in the action of insulin on its target tissues - an alteration referred as insulin resistance - and pancreatic β-cell dysfunction. This latter deterioration is defined by impairment in insulin biosynthesis and secretion, and a loss of β-cell mass by apoptosis. Environmental factors related to T2D, such as chronic elevation in glucose and free fatty acids levels, inflammatory cytokines and pro-atherogenic oxidized low- density lipoproteins (LDL), contribute to the loss of pancreatic β-cell function.In this study, we have demonstrated that the transcription factor Inducible Cyclic AMP Early Repressor (ICER) participates to the progression of both β-cell dysfunction and insulin resistance. The expression of this factor is driven by an alternative promoter and ICER protein represents therefore a truncated product of the Cyclic AMP Response Element Modulator (CREM) family which lacks transactivation domain. Consequently, the transcription factor ICER acts as a passive repressor which reduces expression of genes controlled by the cyclic AMP and Cyclic AMP Response Element Binding protein (CREB) pathway.In insulin-secreting cells, the accumulation of reactive oxygen species caused by environmental factors and notably oxidized LDL - a process known as oxidative stress - induces the transcription factor ICER. This transcriptional repressor hampers the secretory capacity of β-cells by silencing key genes of the exocytotic machinery. In addition, the factor ICER reduces the expression of the scaffold protein Islet Brain 1 (IB 1 ), thereby favouring the activation of the c-Jun N-terminal Kinase (JNK) pathway. This triggering alters in turn insulin biosynthesis and survival capacities of pancreatic β-cells.In the adipose tissue of mice and human subjects suffering from obesity, the transcription factor ICER contributes to the alteration in insulin action. The loss in ICER protein in these tissues induces a constant activation of the CREB pathway and the subsequent expression of the Activating Transcription Factor 3 (ATF3). In turn, this repressor reduces the transcript levels of the glucose transporter GLUT4 and the insulin-sensitizer peptide adiponectin, thereby contributing to the diminution in insulin action.In conclusion, these data shed light on the important role of the transcriptional repressor ICER in the pathogenesis of T2D, which contributes to both alteration in β-cell function and aggravation of insulin resistance. Consequently, a better understanding of the molecular mechanisms responsible for the alterations in ICER levels is required and could lead to develop new therapeutic strategies for the treatment of T2D.RésuméLe diabète de type 2 (DT2) est une maladie métabolique qui affecte plus de 200 millions de personnes dans le monde. La progression de cette affection atteint aujourd'hui des proportions épidémiques imputables à l'augmentation rapide dans les fréquences du surpoids, de l'obésité et de la sédentarité. La pathogenèse du DT2 se caractérise par une diminution de l'action de l'insuline sur ses tissus cibles - un processus nommé insulino-résistance - ainsi qu'une dysfonction des cellules β pancréatiques sécrétrices d'insuline. Cette dernière détérioration se définit par une réduction de la capacité de synthèse et de sécrétion de l'insuline et mène finalement à une perte de la masse de cellules β par apoptose. Des facteurs environnementaux fréquemment associés au DT2, tels l'élévation chronique des taux plasmatiques de glucose et d'acides gras libres, les cytokines pro-inflammatoires et les lipoprotéines de faible densité (LDL) oxydées, contribuent à la perte de fonction des cellules β pancréatiques.Dans cette étude, nous avons démontré que le facteur de transcription « Inducible Cyclic AMP Early Repressor » (ICER) participe à la progression de la dysfonction des cellules β pancréatiques et au développement de Pinsulino-résistance. Son expression étant gouvernée par un promoteur alternatif, la protéine d'ICER représente un produit tronqué de la famille des «Cyclic AMP Response Element Modulator » (CREM), sans domaine de transactivation. Par conséquent, le facteur ICER agit comme un répresseur passif qui réduit l'expression des gènes contrôlés par la voie de l'AMP cyclique et des « Cyclic AMP Response Element Binding protein » (CREB).Dans les cellules sécrétrices d'insuline, l'accumulation de radicaux d'oxygène libres, soutenue par les facteurs environnementaux et notamment les LDL oxydées - un processus appelé stress oxydatif- induit de manière ininterrompue le facteur de transcription ICER. Ainsi activé, ce répresseur transcriptionnel altère la capacité sécrétoire des cellules β en bloquant l'expression de gènes clés de la machinerie d'exocytose. En outre, le facteur ICER favorise l'activation de la cascade de signalisation « c-Jun N- terminal Kinase » (JNK) en réduisant l'expression de la protéine « Islet Brain 1 » (IB1), altérant ainsi les fonctions de biosynthèse de l'insuline et de survie des cellules β pancréatiques.Dans le tissu adipeux des souris et des sujets humains souffrant d'obésité, le facteur de transcription ICER contribue à l'altération de la réponse à l'insuline. La disparition de la protéine ICER dans ces tissus entraîne une activation persistante de la voie de signalisation des CREB et une induction du facteur de transcription « Activating Transcription Factor 3 » (ATF3). A son tour, le répresseur ATF3 inhibe l'expression du transporteur de glucose GLUT4 et du peptide adipocytaire insulino-sensibilisateur adiponectine, contribuant ainsi à la diminution de l'action de l'insuline en conditions d'obésité.En conclusion, à la lumière de ces résultats, le répresseur transcriptionnel ICER apparaît comme un facteur important dans la pathogenèse du DT2, en participant à la perte de fonction des cellules β pancréatiques et à l'aggravation de l'insulino-résistance. Par conséquent, l'étude des mécanismes moléculaires responsables de l'altération des niveaux du facteur ICER pourrait permettre le développement de nouvelles stratégies de traitement du DT2.Résumé didactiqueL'énergie nécessaire au bon fonctionnement de l'organisme est fournie par l'alimentation, notamment sous forme de sucres (glucides). Ceux-ci sont dégradés en glucose, lequel sera distribué aux différents organes par la circulation sanguine. Après un repas, le niveau de glucose sanguin, nommé glycémie, s'élève et favorise la sécrétion d'une hormone appelée insuline par les cellules β du pancréas. L'insuline permet, à son tour, aux organes, tels le foie, les muscles et le tissu adipeux de capter et d'utiliser le glucose ; la glycémie retrouve ainsi son niveau basai.Le diabète de type 2 (DT2) est une maladie métabolique qui affecte plus de 200 millions de personnes dans le monde. Le développement de cette affection est causée par deux processus pathologiques. D'une part, les quantités d'insuline secrétée par les cellules β pancréatiques, ainsi que la survie de ces cellules sont réduites, un phénomène connu sous le nom de dysfonction des cellules β. D'autre part, la sensibilité des tissus à l'insuline se trouve diminuée. Cette dernière altération, l'insulino-résistance, empêche le transport et l'utilisation du glucose par les tissus et mène à une accumulation de ce sucre dans le sang. Cette stagnation de glucose dans le compartiment sanguin est appelée hyperglycémie et favorise l'apparition des complications secondaires du diabète, telles que les maladies cardiovasculaires, l'insuffisance rénale, la cécité et la perte de sensibilité des extrémités.Dans cette étude, nous avons démontré que le facteur ICER qui contrôle spécifiquement l'expression de certains gènes, contribue non seulement à la dysfonction des cellules β, mais aussi au développement de l'insulino-résistance. En effet, dans les cellules β pancréatiques en conditions diabétiques, l'activation du facteur ICER altère la capacité de synthèse et de sécrétion d'insuline et réduit la survie ces cellules.Dans le tissu adipeux des souris et des sujets humains souffrant d'obésité, le facteur ICER contribue à la perte de sensibilité à l'insuline. La disparition d'ICER altère l'expression de la protéine qui capte le glucose, le transoprteur GLUT4, et l'hormone adipocytaire favorisant la sensibilité à l'insuline, nommée adiponectine. Ainsi, la perte d'ICER participe à la réduction de la captation de glucose par le tissue adipeux et au développement de l'insulino-résistance au cours de l'obésité.En conclusion, à la lumière de ces résultats, le facteur ICER apparaît comme un contributeur important à la progression du DT2, en soutenant la dysfonction des cellules β pancréatiques et l'aggravation de l'insulino-résistance. Par conséquent, l'étude des mécanismes responsables de la dérégulation du facteur ICER pourrait permettre le développement de nouvelles stratégies de traitement du DT2.
Resumo:
Plasma urate levels are higher in humans than rodents (240-360 vs. â^¼30 μM) because humans lack the liver enzyme uricase. High uricemia in humans may protect against oxidative stress, but hyperuricemia also associates with the metabolic syndrome, and urate and uric acid can crystallize to cause gout and renal dysfunctions. Thus, hyperuricemic animal models to study urate-induced pathologies are needed. We recently generated mice with liver-specific ablation of Glut9, a urate transporter providing access of urate to uricase (LG9KO mice). LG9KO mice had moderately high uricemia (â^¼120 μM). To further increase their uricemia, here we gavaged LG9KO mice for 3 days with inosine, a urate precursor; this treatment was applied in both chow- and high-fat-fed mice. In chow-fed LG9KO mice, uricemia peaked at 300 μM 2 h after the first gavage and normalized 24 h after the last gavage. In contrast, in high-fat-fed LG9KO mice, uricemia further rose to 500 μM. Plasma creatinine strongly increased, indicating acute renal failure. Kidneys showed tubule dilation, macrophage infiltration, and urate and uric acid crystals, associated with a more acidic urine. Six weeks after inosine gavage, plasma urate and creatinine had normalized. However, renal inflammation, fibrosis, and organ remodeling had developed despite the disappearance of urate and uric acid crystals. Thus, hyperuricemia and high-fat diet feeding combined to induce acute renal failure. Furthermore, a sterile inflammation caused by the initial crystal-induced lesions developed despite the disappearance of urate and uric acid crystals.
Resumo:
Brain-derived neurotrophic factor (BDNF) promotes synaptic plasticity via an enhancement in expression of specific synaptic proteins. Recent results suggest that the neuronal monocarboxylate transporter MCT2 is a postsynaptic protein critically involved in synaptic plasticity and long-term memory. To investigate in vivo whether BDNF can modulate the expression of MCT2 as well as other proteins involved in synaptic plasticity, acute injection of BDNF was performed in mouse dorsal hippocampal CA1 area. Using immunohistochemistry, it was found that MCT2 expression was enhanced in part of the CA1 area and in the dentate gyrus 6 h after a single intrahippocampal injection of BDNF. Similarly, expression of the immediate early genes Arc and Zif268 was enhanced in the same hippocampal areas, in accordance with their role in synaptic plasticity. Immunoblot analysis confirmed the significant enhancement in MCT2 protein expression. In contrast, no changes were observed for the glial monocarboxylate transporters MCT1 and MCT4. When other synaptic proteins were investigated, it was found that postsynaptic density 95 (PSD95) and glutamate receptor 2 (GluR2) protein levels were significantly enhanced while no effect could be detected for synaptophysin, synaptosomal-associated protein 25 (SNAP25), αCaMKII and GluR1. These results demonstrate that MCT2 expression can be upregulated together with other key postsynaptic proteins in vivo under conditions related to synaptic plasticity, further suggesting the importance of energetics for memory formation.
Resumo:
The molecular cloning of facilitated sugar transporters has led to the identification of a family of transport molecules having similar functions, but possessing specific kinetic and regulatory properties. These transporter isoforms are characterized by different primary structures, specific tissue localization, and polarized expression within the same epithelial cells. The use of Xenopus oocytes for the functional expression of different members of this transporter family has been of considerable value in defining the kinetic properties and sugar specificities of the different isoforms. The expression of chimeric or variously mutated transporters should, in the near future, permit the determination of the structural basis for their kinetic properties and sugar specificities. cDNA probes and antipeptide antibodies specific for each isoform are now being used to determine their specific regulation during development and in different states of altered glucose homeostasis. The variety of molecular forms implicated in the apparently simple task of sugar uptake or transepithelial transport has been surprising. With the available molecular tools now in hand, it will be possible to study these mechanisms in much greater detail.
Resumo:
Lactate release by astrocytes is postulated to be of importance for neuroenergetics but its regulation is poorly understood. Basigin, a chaperone protein for specific monocarboxylate transporters (MCTs), represents a putatively important regulatory element for lactate fluxes. Indeed, basigin knockdown by RNA interference in primary cultures of astrocytes partially reduced both proton-driven lactate influx and efflux. But more strikingly, enhancement of lactate efflux induced by glutamate was prevented while the effect of sodium azide was significantly reduced by treatment of cultured astrocytes with anti-basigin small interfering RNA. Enhancement of glucose utilization was unaffected under the same conditions. Basal lactate uptake and release were significantly reduced by MCT1 knockdown, even more so than with basigin knockdown, whereas glutamate-driven or sodium azide-induced enhancement of lactate release was not inhibited by either MCT1, 2, or 4 small interfering RNAs. In conclusion, MCT1 plays a pivotal role in the control of basal proton-driven lactate flux in astrocytes while basigin is only partly involved, most likely via its interaction with MCT1. In contrast, basigin appears to critically regulate the enhancement of lactate release caused by glutamate (or sodium azide) but via an effect on another unidentified transporter at least present in astrocytes in vitro.
Resumo:
Bordetella pertussis is the bacterial agent of whooping cough in humans. Under iron-limiting conditions, it produces the siderophore alcaligin. Released to the extracellular environment, alcaligin chelates iron, which is then taken up as a ferric alcaligin complex via the FauA outer membrane transporter. FauA belongs to a family of TonB-dependent outer membrane transporters that function using energy derived from the proton motive force. Using an in-house protocol for membrane-protein expression, purification and crystallization, FauA was crystallized in its apo form together with three other TonB-dependent transporters from different organisms. Here, the protocol used to study FauA is described and its three-dimensional structure determined at 2.3 A resolution is discussed.
Resumo:
Five functional mammalian facilitated hexose carriers (GLUTs) have been characterized by molecular cloning. By functional expression in heterologous systems, their specificity and affinity for different hexoses have been defined. There are three high-affinity transporters (GLUT-1, GLUT-3 and GLUT-4) and one low-affinity transporter (GLUT-2), and GLUT-5 is primarily a fructose carrier. Because their Michaelis constants (Km) are below the normal blood glucose concentration, the high-affinity transporters function at rates close to maximal velocity. Thus their level of cell surface expression greatly influences the rate of glucose uptake into the cells. In contrast, the rate of glucose uptake by GLUT-2 (Km = 17 mM) increases in parallel with the rise in blood glucose over the physiological concentration range. High-affinity transporters are found in almost every tissue, but their expression is higher in cells with high glycolytic activity. Glut-2, however, is found in tissues carrying large glucose fluxes, such as intestine, kidney, and liver. As an adaptive response to variations in metabolic conditions, the expression of these transporters is regulated by glucose and different hormones. Thus, because of their specific characteristics and regulated expression, the facilitated glucose transporters control fundamental aspects of glucose homeostasis. I review data pertaining to the structure and regulated expression of the glucose carriers present in intestine, kidney, and liver and discuss their role in the control of glucose flux into or out of these different tissues.
Resumo:
Glucose has been considered the major, if not the exclusive, energy substrate for the brain. But under certain physiological and pathological conditions other substrates, namely monocarboxylates (lactate, pyruvate and ketone bodies), can contribute significantly to satisfy brain energy demands. These monocarboxylates need to be transported across the blood-brain barrier or out of astrocytes into the extracellular space and taken up into neurons. It has been shown that monocarboxylates are transported by a family of proton-linked transporters called monocarboxylate transporters (MCTs). In the central nervous system, MCT2 is the predominant neuronal isoform and little is known about the regulation of its expression. Noradrenaline (NA), insulin and IGF-1 were previously shown to enhance the expression of MCT2 in cultured cortical neurons via a translational mechanism. Here we demonstrate that the well known brain neurotrophic factor BDNF enhances MCT2 protein expression in cultured cortical neurons and in synaptoneurosome preparations in a time- and concentrationdependent manner without affecting MCT2 mRNA levels. We observed that BDNF induced MCT2 expression by activation of MAPK as well as PI3K/Akt/mTOR signaling pathways. Furthermore, we investigated the possible post-transcriptional regulation of MCT2 expression by a neuronal miRNA. Then, we demonstrated that BDNF enhanced MCT2 expression in the hippocampus in vivo, in parallel with some post-synaptic proteins such as PSD95 and AMPA receptor GluR2/3 subunits, and two immediate early genes Arc and Zif268 known to be expressed in conditions related to synaptic plasticity. In the last part, we demonstrated in vivo that a downregulation of hippocampal MCT2 via silencing with an appropriate lentiviral vector in mice caused an impairment of working memory without reference memory deficit. In conclusion, these results suggest that regulation of neuronal monocarboxylate transporter MCT2 expression could be a key event in the context of synaptic plasticity, allowing an adequate energy substrate supply in situations of altered synaptic efficacy. - Le glucose représente le substrat énergétique majeur pour le cerveau. Cependant, dans certaines conditions physiologiques ou pathologiques, le cerveau a la capacité d'utiliser des substrats énergéiques appartenant à la classe des monocarboxylates (lactate, pyruvate et corps cétoniques) afin de satisfaire ses besoins énergétiques. Ces monocarboxylates doivent être transportés à travers la barrière hématoencéphalique mais aussi hors des astrocytes vers l'espace extracellulaire puis re-captés par les neurones. Leur transport est assuré par une famillle de transporteurs aux monocarboxylates (MCTs). Dans le système nerveux central, les neurones expriment principalement l'isoforme MCT2 mais peu d'informations sont disponibles concernant la régulation de son expression. Il a été montré que la noradrénaline, l'insuline et l'IGF-1 induisent l'expression de MCT2 dans des cultures de neurones corticaux par un mécanisme traductionnel. Dans cette étude nous démontrons dans un premier temps que le facteur neurotrophique BDNF augmente l'expression de MCT2 à la fois dans des cultures de neurones corticaux et dans les préparations synaptoneurosomales selon un décours temporel et une gamme de concentrations propre. Aucun changement n'a été observé concernant les niveaux d'ARNm de MCT2. Nous avons observé que le BDNF induisait l'expression de MCT2 par l'activation simultanée des voies de signalisation MAPK et PI3K/Akt/mTOR. De plus, nous nous sommes intéressés à une potentielle régulation par les micro-ARNs de la synthèse de MCT2. Ensuite, nous avons démontré que le BDNF induit aussi l'expression de MCT2 dans l'hippocampe de la souris en parallèle avec d'autres protéines post-synaptiques telles que PSD95 et GluR2/3 et avec deux « immediate early genes » tels que Arc et Zif268 connus pour être exprimés dans des conditions de plasticité synaptique. Dans un dernier temps, nous avons démontré qu'une diminution d'expression de MCT2 induite par le biais d'un siRNA exprimé via un vecteur lentiviral dans l'hippocampe de souris générait des déficits de mémoire de travail sans affecter la mémoire de référence. En conclusion, ces résultats nous suggèrent que le transporteur aux monocarboxylates neuronal MCT2 serait essentiel pour l'apport énergétique du lactate pour les neurones dans des conditions de haute activité neuronale comme c'est le cas pendant les processus de plasticité synaptique.
Resumo:
The excitatory neurotransmitter glutamate has been reported to have a major impact on brain energy metabolism. Using primary cultures of rat hippocampal neurons, we observed that glutamate reduces glucose utilization in this cell type, suggesting alteration in mitochondrial oxidative metabolism. The aquaglyceroporin AQP9 and the monocarboxylate transporter MCT2, two transporters for oxidative energy substrates, appear to be present in mitochondria of these neurons. Moreover, they not only co-localize but they interact with each other as they were found to co-immunoprecipitate from hippocampal neuron homogenates. Exposure of cultured hippocampal neurons to glutamate 100 μM for 1 h led to enhanced expression of both AQP9 and MCT2 at the protein level without any significant change at the mRNA level. In parallel, a similar increase in the protein expression of LDHA was evidenced without an effect on the mRNA level. These data suggest that glutamate exerts an influence on neuronal energy metabolism likely through a regulation of the expression of some key mitochondrial proteins.
Resumo:
Land plants have developed a cuticle preventing uncontrolled water loss. Here we report that an ATP-binding cassette (ABC) subfamily G (ABCG) full transporter is required for leaf water conservation in both wild barley and rice. A spontaneous mutation, eibi1.b, in wild barley has a low capacity to retain leaf water, a phenotype associated with reduced cutin deposition and a thin cuticle. Map-based cloning revealed that Eibi1 encodes an HvABCG31 full transporter. The gene was highly expressed in the elongation zone of a growing leaf (the site of cutin synthesis), and its gene product also was localized in developing, but not in mature tissue. A de novo wild barley mutant named "eibi1.c," along with two transposon insertion lines of rice mutated in the ortholog of HvABCG31 also were unable to restrict water loss from detached leaves. HvABCG31 is hypothesized to function as a transporter involved in cutin formation. Homologs of HvABCG31 were found in green algae, moss, and lycopods, indicating that this full transporter is highly conserved in the evolution of land plants.
Resumo:
Insulin-dependent diabetes mellitus is an autoimmune disease in which pancreatic islet beta cells are destroyed by a combination of immunological and inflammatory mechanisms. In particular, cytokine-induced production of nitric oxide has been shown to correlate with beta cell apoptosis and/or inhibition of insulin secretion. In the present study, we investigated whether the interleukin (IL)-1beta intracellular signal transduction pathway could be blocked by overexpression of dominant negative forms of the IL-1 receptor interacting protein MyD88. We show that overexpression of the Toll domain or the lpr mutant of MyD88 in betaTc-Tet cells decreased nuclear factor kappaB (NF-kappaB) activation upon IL-1beta and IL-1beta/interferon (IFN)-gamma stimulation. Inducible nitric oxide synthase mRNA accumulation and nitrite production, which required the simultaneous presence of IL-1beta and IFN-gamma, were also suppressed by approximately 70%, and these cells were more resistant to cytokine-induced apoptosis as compared with parental cells. The decrease in glucose-stimulated insulin secretion induced by IL-1beta and IFN-gamma was however not prevented. This was because these dysfunctions were induced by IFN-gamma alone, which decreased cellular insulin content and stimulated insulin exocytosis. These results demonstrate that IL-1beta is involved in inducible nitric oxide synthase gene expression and induction of apoptosis in mouse beta cells but does not contribute to impaired glucose-stimulated insulin secretion. Furthermore, our data show that IL-1beta cellular actions can be blocked by expression of MyD88 dominant negative proteins and, finally, that cytokine-induced beta cell secretory dysfunctions are due to the action of IFN-gamma.
Resumo:
The monocarboxylate transporter MCT2 belongs to a large family of membrane proteins involved in the transport of lactate, pyruvate and ketone bodies. Although its expression in rodent brain has been well documented, the presence of MCT2 in the human brain has been questioned on the basis of low mRNA abundance. In this study, the distribution of the monocarboxylate transporter MCT2 has been investigated in the cortex of normal adult human brain using an immunohistochemical approach. Widespread neuropil staining in all cortical layers was observed by light microscopy. Such a distribution was very similar in three different cortical areas investigated. At the cellular level, the expression of MCT2 could be observed in a large number of neurons, in fibers both in grey and white matter, as well as in some astrocytes, mostly localized in layer I and in the white matter. Double staining experiments combined with confocal microscopy confirmed the neuronal expression but also suggested a preferential postsynaptic localization of synaptic MCT2 expression. A few astrocytes in the grey matter appeared to exhibit MCT2 labelling but at low levels. Electron microscopy revealed strong MCT2 expression at asymmetric synapses in the postsynaptic density and also within the spine head but not in the presynaptic terminal. These data not only demonstrate neuronal MCT2 expression in human, but since a portion of it exhibits a distinct synaptic localization, it further supports a putative role for MCT2 in adjustment of energy supply to levels of activity.