Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Cell Physiol ; 237(7): 3057-3068, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35561261

RESUMO

Bisphenol S (BPS) is a common substitute of bisphenol A (BPA). Recent data suggest that BPS acts as an obesogenic endocrine disruptor with emerging implications in the physiopathology of metabolic syndrome. However, the effects of BPS on monocarboxylate transporters (acting as carriers for lactate, pyruvate, and ketone bodies) and the mitochondrial respiratory system in the liver remain limited. For this purpose, male Swiss mice were treated with BPS at 100 µg/kg/day for 10 weeks, in drinking water. An increase in body weight and food intake was observed with no increase in locomotor activity. Moreover, data show that BPS increases hepatic MCT1 (a key energetic fuel transporter) mRNA expression accompanied by hepatic steatosis initiation and lipid accumulation, while disrupting mitochondrial function and oxidative stress parameters. Furthermore, BPS produced a significant increase in lactate dehydrogenase and creatine kinase activities. We can suggest that BPS contributes to hepatic steatosis in mice by upregulating monocarboxylate transporters and affecting the bioenergetic status characterized by an impaired mitochondrial respiratory system. Thus, our data highlight a new mechanism putatively implicated in hepatic steatosis development during BPS-induced obesity involving lactate metabolism.


Assuntos
Compostos Benzidrílicos , Fígado Gorduroso , Animais , Compostos Benzidrílicos/toxicidade , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/genética , Lactatos , Masculino , Camundongos , Mitocôndrias , Sistema Respiratório , Regulação para Cima
2.
Int J Mol Sci ; 22(4)2021 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-33557247

RESUMO

The effect of a cellular prion protein (PrPc) deficiency on neuroenergetics was primarily analyzed via surveying the expression of genes specifically involved in lactate/pyruvate metabolism, such as monocarboxylate transporters (MCT1, MCT2, MCT4). The aim of the present study was to elucidate a potential involvement of PrPc in the regulation of energy metabolism in different brain regions. By using quantitative real-time polymerase chain reaction (qRT-PCR), we observed a marked reduction in MCT1 mRNA expression in the cortex of symptomatic Zürich I Prnp-/- mice, as compared to their wild-type (WT) counterparts. MCT1 downregulation in the cortex was accompanied with significantly decreased expression of the MCT1 functional interplayer, the Na+/K+ ATPase α2 subunit. Conversely, the MCT1 mRNA level was significantly raised in the cerebellum of Prnp-/- vs. WT control group, without a substantial change in the Na+/K+ ATPase α2 subunit expression. To validate the observed mRNA findings, we confirmed the observed change in MCT1 mRNA expression level in the cortex at the protein level. MCT4, highly expressed in tissues that rely on glycolysis as an energy source, exhibited a significant reduction in the hippocampus of Prnp-/- vs. WT mice. The present study demonstrates that a lack of PrPc leads to altered MCT1 and MCT4 mRNA/protein expression in different brain regions of Prnp-/- vs. WT mice. Our findings provide evidence that PrPc might affect the monocarboxylate intercellular transport, which needs to be confirmed in further studies.


Assuntos
Cerebelo/metabolismo , Córtex Cerebral/metabolismo , Ácido Láctico/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Priônicas/fisiologia , RNA Mensageiro/metabolismo , Simportadores/metabolismo , Animais , Transporte Biológico , Glicólise , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transportadores de Ácidos Monocarboxílicos/genética , RNA Mensageiro/genética , Simportadores/genética
3.
Int J Mol Sci ; 22(17)2021 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-34502381

RESUMO

Myelin is of vital importance to the central nervous system and its disruption is related to a large number of both neurodevelopmental and neurodegenerative diseases. The differences observed between human and rodent oligodendrocytes make animals inadequate for modeling these diseases. Although developing human in vitro models for oligodendrocytes and myelinated axons has been a great challenge, 3D cell cultures derived from iPSC are now available and able to partially reproduce the myelination process. We have previously developed a human iPSC-derived 3D brain organoid model (also called BrainSpheres) that contains a high percentage of myelinated axons and is highly reproducible. Here, we have further refined this technology by applying multiple readouts to study myelination disruption. Myelin was assessed by quantifying immunostaining/confocal microscopy of co-localized myelin basic protein (MBP) with neurofilament proteins as well as proteolipid protein 1 (PLP1). Levels of PLP1 were also assessed by Western blot. We identified compounds capable of inducing developmental neurotoxicity by disrupting myelin in a systematic review to evaluate the relevance of our BrainSphere model for the study of the myelination/demyelination processes. Results demonstrated that the positive reference compound (cuprizone) and two of the three potential myelin disruptors tested (Bisphenol A, Tris(1,3-dichloro-2-propyl) phosphate, but not methyl mercury) decreased myelination, while ibuprofen (negative control) had no effect. Here, we define a methodology that allows quantification of myelin disruption and provides reference compounds for chemical-induced myelin disruption.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Bainha de Mielina/metabolismo , Bainha de Mielina/fisiologia , Axônios/metabolismo , Encéfalo/metabolismo , Técnicas de Cultura de Células/métodos , Sistema Nervoso Central/metabolismo , Humanos , Modelos Biológicos , Proteína Básica da Mielina/análise , Proteína Básica da Mielina/metabolismo , Proteína Proteolipídica de Mielina/análise , Proteína Proteolipídica de Mielina/metabolismo , Fibras Nervosas Mielinizadas/metabolismo , Fibras Nervosas Mielinizadas/patologia , Síndromes Neurotóxicas/metabolismo , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Organoides/metabolismo
4.
Proc Natl Acad Sci U S A ; 113(39): 11004-9, 2016 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-27621431

RESUMO

The multifunctional protein E4 transcription factor 1 (E4F1) is an essential regulator of epidermal stem cell (ESC) maintenance. Here, we found that E4F1 transcriptionally regulates a metabolic program involved in pyruvate metabolism that is required to maintain skin homeostasis. E4F1 deficiency in basal keratinocytes resulted in deregulated expression of dihydrolipoamide acetyltransferase (Dlat), a gene encoding the E2 subunit of the mitochondrial pyruvate dehydrogenase (PDH) complex. Accordingly, E4f1 knock-out (KO) keratinocytes exhibited impaired PDH activity and a redirection of the glycolytic flux toward lactate production. The metabolic reprogramming of E4f1 KO keratinocytes associated with remodeling of their microenvironment and alterations of the basement membrane, led to ESC mislocalization and exhaustion of the ESC pool. ShRNA-mediated depletion of Dlat in primary keratinocytes recapitulated defects observed upon E4f1 inactivation, including increased lactate secretion, enhanced activity of extracellular matrix remodeling enzymes, and impaired clonogenic potential. Altogether, our data reveal a central role for Dlat in the metabolic program regulated by E4F1 in basal keratinocytes and illustrate the importance of PDH activity in skin homeostasis.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Di-Hidrolipoil-Lisina-Resíduo Acetiltransferase/metabolismo , Homeostase , Proteínas Mitocondriais/metabolismo , Pele/metabolismo , Fatores de Transcrição/metabolismo , Animais , Animais Recém-Nascidos , Membrana Basal/metabolismo , Adesão Celular , Células Cultivadas , Microambiente Celular , Proteínas de Ligação a DNA/deficiência , Di-Hidrolipoil-Lisina-Resíduo Acetiltransferase/genética , Células Epidérmicas , Epiderme/metabolismo , Regulação da Expressão Gênica , Queratinócitos/citologia , Queratinócitos/metabolismo , Camundongos Knockout , Proteínas Mitocondriais/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Musculares/metabolismo , Piruvatos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Repressoras , Células-Tronco/metabolismo , Fatores de Transcrição/deficiência , Ubiquitina-Proteína Ligases
5.
J Neurosci ; 35(10): 4151-6, 2015 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-25762662

RESUMO

Lactate, a product of glycolysis, has been shown to play a key role in the metabolic support of neurons/axons in the CNS by both astrocytes and oligodendrocytes through monocarboxylate transporters (MCTs). Despite such importance in the CNS, little is known about MCT expression and lactate function in the PNS. Here we show that mouse MCT1, MCT2, and MCT4 are expressed in the PNS. While DRG neurons express MCT1, myelinating Schwann cells (SCs) coexpress MCT1 and MCT4 in a domain-specific fashion, mainly in regions of noncompact myelin. Interestingly, SC-specific downregulation of MCT1 expression in rat neuron/SC cocultures led to increased myelination, while its downregulation in neurons resulted in a decreased amount of neurofilament. Finally, pure rat SCs grown in the presence of lactate exhibited an increase in the level of expression of the main myelin regulator gene Krox20/Egr2 and the myelin gene P0. These data indicate that lactate homeostasis participates in the regulation of the SC myelination program and reveal that similar to CNS, PNS axon-glial metabolic interactions are most likely mediated by MCTs.


Assuntos
Regulação da Expressão Gênica/fisiologia , Ácido Láctico/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Bainha de Mielina/metabolismo , Nervos Periféricos/metabolismo , Células Receptoras Sensoriais/metabolismo , Actinas/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Moléculas de Adesão Celular Neuronais/metabolismo , Células Cultivadas , Técnicas de Cocultura , Proteína 2 de Resposta de Crescimento Precoce/genética , Embrião de Mamíferos , Gânglios Espinais/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Técnicas In Vitro , Ácido Láctico/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transportadores de Ácidos Monocarboxílicos/classificação , Transportadores de Ácidos Monocarboxílicos/genética , Proteína Básica da Mielina/metabolismo , Proteína P0 da Mielina/genética , Proteínas de Neurofilamentos/metabolismo , Nervos Periféricos/citologia , Ratos , Ratos Sprague-Dawley , Células de Schwann/efeitos dos fármacos , Células de Schwann/metabolismo , Células Receptoras Sensoriais/efeitos dos fármacos
6.
Am J Physiol Endocrinol Metab ; 310(2): E103-15, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26530151

RESUMO

Monocarboxylates have been implicated in the control of energy homeostasis. Among them, the putative role of ketone bodies produced notably during high-fat diet (HFD) has not been thoroughly explored. In this study, we aimed to determine the impact of a specific rise in cerebral ketone bodies on food intake and energy homeostasis regulation. A carotid infusion of ketone bodies was performed on mice to stimulate sensitive brain areas for 6 or 12 h. At each time point, food intake and different markers of energy homeostasis were analyzed to reveal the consequences of cerebral increase in ketone body level detection. First, an increase in food intake appeared over a 12-h period of brain ketone body perfusion. This stimulated food intake was associated with an increased expression of the hypothalamic neuropeptides NPY and AgRP as well as phosphorylated AMPK and is due to ketone bodies sensed by the brain, as blood ketone body levels did not change at that time. In parallel, gluconeogenesis and insulin sensitivity were transiently altered. Indeed, a dysregulation of glucose production and insulin secretion was observed after 6 h of ketone body perfusion, which reversed to normal at 12 h of perfusion. Altogether, these results suggest that an increase in brain ketone body concentration leads to hyperphagia and a transient perturbation of peripheral metabolic homeostasis.


Assuntos
Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Corpos Cetônicos/farmacologia , Adenilato Quinase/metabolismo , Proteína Relacionada com Agouti/metabolismo , Animais , Glicemia , Dieta Hiperlipídica , Ingestão de Alimentos/fisiologia , Metabolismo Energético/fisiologia , Gluconeogênese/efeitos dos fármacos , Gluconeogênese/fisiologia , Homeostase , Hipotálamo/metabolismo , Resistência à Insulina/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuropeptídeo Y/metabolismo , Fosforilação/efeitos dos fármacos
7.
Chem Senses ; 40(8): 537-56, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26209545

RESUMO

Obesity is associated with chronic food intake disorders and binge eating. Food intake relies on the interaction between homeostatic regulation and hedonic signals among which, olfaction is a major sensory determinant. However, its potential modulation at the peripheral level by a chronic energy imbalance associated to obese status remains a matter of debate. We further investigated the olfactory function in a rodent model relevant to the situation encountered in obese humans, where genetic susceptibility is juxtaposed on chronic eating disorders. Using several olfactory-driven tests, we compared the behaviors of obesity-prone Sprague-Dawley rats (OP) fed with a high-fat/high-sugar diet with those of obese-resistant ones fed with normal chow. In OP rats, we reported 1) decreased odor threshold, but 2) poor olfactory performances, associated with learning/memory deficits, 3) decreased influence of fasting, and 4) impaired insulin control on food seeking behavior. Associated with these behavioral modifications, we found a modulation of metabolism-related factors implicated in 1) electrical olfactory signal regulation (insulin receptor), 2) cellular dynamics (glucorticoids receptors, pro- and antiapoptotic factors), and 3) homeostasis of the olfactory mucosa and bulb (monocarboxylate and glucose transporters). Such impairments might participate to the perturbed daily food intake pattern that we observed in obese animals.


Assuntos
Obesidade/etiologia , Olfato/fisiologia , Animais , Comportamento Animal , Peso Corporal , Dieta Hiperlipídica , Ingestão de Alimentos , Metabolismo Energético , Proteínas Facilitadoras de Transporte de Glucose/genética , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Insulina/metabolismo , Masculino , Modelos Animais , Obesidade/metabolismo , Odorantes , Bulbo Olfatório/metabolismo , Mucosa Olfatória/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Receptores para Leptina/genética , Receptores para Leptina/metabolismo
8.
Mol Metab ; 83: 101926, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38553002

RESUMO

OBJECTIVE: Ketone bodies (such as ß-hydroxybutyrate or BHB) have been recently proposed as signals involved in brain regulation of energy homeostasis and obesity development. However, the precise role of ketone bodies sensing by the brain, and its impact on metabolic disorder development remains unclear. Nevertheless, partial deletion of the ubiquitous ketone bodies transporter MCT1 in mice (HE mice) results in diet-induced obesity resistance, while there is no alteration under normal chow diet. These results suggest that ketone bodies produced during the high fat diet would be important signals involved in obesity onset. METHODS: In the present study we used a specific BHB infusion of the hypothalamus and analyzed the energy homeostasis of WT or HE mice fed a normal chow diet. RESULTS: Our results indicate that high BHB levels sensed by the hypothalamus disrupt the brain regulation of energy homeostasis. This brain control dysregulation leads to peripheral alterations of energy expenditure mechanisms. CONCLUSIONS: Altogether, the changes induced by high ketone bodies levels sensed by the brain increase the risk of obesity onset in mice.


Assuntos
Ácido 3-Hidroxibutírico , Metabolismo Energético , Hipotálamo , Corpos Cetônicos , Camundongos Endogâmicos C57BL , Obesidade , Animais , Hipotálamo/metabolismo , Camundongos , Corpos Cetônicos/metabolismo , Masculino , Obesidade/metabolismo , Ácido 3-Hidroxibutírico/metabolismo , Dieta Hiperlipídica/efeitos adversos , Doenças Metabólicas/metabolismo , Doenças Metabólicas/etiologia , Homeostase , Transportadores de Ácidos Monocarboxílicos/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Simportadores/metabolismo , Simportadores/genética
9.
Mol Neurobiol ; 60(1): 84-97, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36223047

RESUMO

Astrocyte reaction is a complex cellular process involving astrocytes in response to various types of CNS injury and a marker of neurotoxicity. It has been abundantly studied in rodents but relatively poorly in human cells due to limited access to the brain. Astrocytes play important roles in cerebral energy metabolism and are also key players in neuroinflammation. Astroglial metabolic and inflammatory changes have been reported with age, leading to the hypothesis that mitochondrial metabolism and inflammatory responses are interconnected. However, the relationship between energy metabolism and astrocyte reactivity in the context of neurotoxicity is not known. We hypothesized that changes in energy metabolism of astrocytes will be coupled to their activation by xenobiotics. Astrocyte reaction and associated energy metabolic changes were assessed by immunostaining, gene expression, proteomics, metabolomics, and extracellular flux analyses after 24 h of exposure of human ReN-derived astrocytes to digoxin (1-10 µM) or TNFα (30 ng/ml) used as a positive control. Strong astrocytic reaction was observed, accompanied by increased glycolysis at low concentrations of digoxin (0.1 and 0.5 µM) and after TNFα exposure, suggesting that increased glycolysis may be a common feature of reactive astrocytes, independent of the triggering molecule. In conclusion, whether astrocyte activation is triggered by cytokines or a xenobiotic, it is strongly tied to energy metabolism in human ReN-derived astrocytes. Increased glycolysis might be considered as an endpoint to detect astrocyte activation by potentially neurotoxic compounds in vitro. Finally, ReN-derived astrocytes may help to decipher mechanisms of neurotoxicity in ascertaining the ability of chemicals to directly target astrocytes.


Assuntos
Astrócitos , Digoxina , Humanos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Sistema Nervoso Central/metabolismo , Digoxina/farmacologia , Metabolismo Energético , Fator de Necrose Tumoral alfa/farmacologia , Células Cultivadas
10.
Glia ; 59(12): 1987-95, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21901758

RESUMO

The monocarboxylate transporter MCT4 is a proton-linked carrier particularly important for lactate release from highly glycolytic cells. In the central nervous system, MCT4 is exclusively expressed by astrocytes. Surprisingly, MCT4 expression in primary cultures of mouse cortical astrocytes is conspicuously low, suggesting that an external, nonastrocytic signal is necessary to obtain the observed pattern of expression in vivo. Here, we demonstrate that nitric oxide (NO), delivered by various NO donors, time- and dose-dependently induces MCT4 expression in cultured cortical astrocytes both at the mRNA and protein levels. In contrast, NO does not enhance the expression of MCT1, the other astrocytic monocarboxylate transporter. The transcriptional effect of NO is not mediated by a cGMP-dependent mechanism as shown by the absence of effect of a cGMP analog or of a selective guanylate cyclase inhibitor. NO causes an increase in astrocytic lactate transport capacity which requires the enhancement of MCT4 expression as both are prevented by the use of a specific siRNA against MCT4. In addition, cumulated lactate release by astrocytes over a period of 24 h was also enhanced by NO treatment. Our data suggest that NO represents a putative intercellular signal to control MCT4 expression in astrocytes and in doing so, to facilitate lactate transfer to other surrounding cell types in the central nervous system. © 2011 Wiley-Liss, Inc.


Assuntos
Astrócitos/metabolismo , GMP Cíclico/fisiologia , Transportadores de Ácidos Monocarboxílicos/agonistas , Transportadores de Ácidos Monocarboxílicos/genética , Proteínas Musculares/agonistas , Proteínas Musculares/genética , Óxido Nítrico/fisiologia , Ativação Transcricional/fisiologia , Animais , Animais Recém-Nascidos , Astrócitos/citologia , Comunicação Celular/fisiologia , Líquido Extracelular/química , Líquido Extracelular/metabolismo , Regulação da Expressão Gênica/genética , Camundongos , Transportadores de Ácidos Monocarboxílicos/biossíntese , Proteínas Musculares/biossíntese , Cultura Primária de Células , Transdução de Sinais/fisiologia
11.
Front Neurosci ; 14: 616824, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33519368

RESUMO

Neonatal hypoxia-ischemia (nHI) is a major cause of death or subsequent disabilities in infants. Hypoxia-ischemia causes brain lesions, which are induced by a strong reduction in oxygen and nutrient supply. Hypothermia is the only validated beneficial intervention, but not all newborns respond to it and today no pharmacological treatment exists. Among possible therapeutic agents to test, trans-resveratrol is an interesting candidate as it has been reported to exhibit neuroprotective effects in some neurodegenerative diseases. This experimental study aimed to investigate a possible neuroprotection by resveratrol in rat nHI, when administered to the pregnant rat female, at a nutritional dose. Several groups of pregnant female rats were studied in which resveratrol was added to drinking water either during the last week of pregnancy, the first week of lactation, or both. Then, 7-day old pups underwent a hypoxic-ischemic event. Pups were followed longitudinally, using both MRI and behavioral testing. Finally, a last group was studied in which breastfeeding females were supplemented 1 week with resveratrol just after the hypoxic-ischemic event of the pups (to test the curative rather than the preventive effect). To decipher the molecular mechanisms of this neuroprotection, RT-qPCR and Western blots were also performed on pup brain samples. Data clearly indicated that when pregnant and/or breastfeeding females were supplemented with resveratrol, hypoxic-ischemic offspring brain lesions were significantly reduced. Moreover, maternal resveratrol supplementation allowed to reverse sensorimotor and cognitive deficits caused by the insult. The best recoveries were observed when resveratrol was administered during both gestation and lactation (2 weeks before the hypoxic-ischemic event in pups). Furthermore, neuroprotection was also observed in the curative group, but only at the latest stages examined. Our hypothesis is that resveratrol, in addition to the well-known neuroprotective benefits via the sirtuin's pathway (antioxidant properties, inhibition of apoptosis), has an impact on brain metabolism, and more specifically on the astrocyte-neuron lactate shuttle (ANLS) as suggested by RT-qPCR and Western blot data, that contributes to the neuroprotective effects.

12.
Eur J Neurosci ; 29(10): 1951-63, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19453627

RESUMO

MCT2 is the major neuronal monocarboxylate transporter (MCT) that allows the supply of alternative energy substrates such as lactate to neurons. Recent evidence obtained by electron microscopy has demonstrated that MCT2, like alpha-amino-3-hydroxyl-5-methyl-4-isoxazole-propionic acid (AMPA) receptors, is localized in dendritic spines of glutamatergic synapses. Using immunofluorescence, we show in this study that MCT2 colocalizes extensively with GluR2/3 subunits of AMPA receptors in neurons from various mouse brain regions as well as in cultured neurons. It also colocalizes with GluR2/3-interacting proteins, such as C-kinase-interacting protein 1, glutamate receptor-interacting protein 1 and clathrin adaptor protein. Coimmunoprecipitation of MCT2 with GluR2/3 and C-kinase-interacting protein 1 suggests their close interaction within spines. Parallel changes in the localization of both MCT2 and GluR2/3 subunits at and beneath the plasma membrane upon various stimulation paradigms were unraveled using an original immunocytochemical and transfection approach combined with three-dimensional image reconstruction. Cell culture incubation with AMPA or insulin triggered a marked intracellular accumulation of both MCT2 and GluR2/3, whereas both tumor necrosis factor alpha and glycine (with glutamate) increased their cell surface immunolabeling. Similar results were obtained using Western blots performed on membrane or cytoplasm-enriched cell fractions. Finally, an enhanced lactate flux into neurons was demonstrated after MCT2 translocation on the cell surface. These observations provide unequivocal evidence that MCT2 is linked to AMPA receptor GluR2/3 subunits and undergoes a similar translocation process in neurons upon activation. MCT2 emerges as a novel component of the synaptic machinery putatively linking neuroenergetics to synaptic transmission.


Assuntos
Transportadores de Ácidos Monocarboxílicos/metabolismo , Neurônios/metabolismo , Receptores de AMPA/metabolismo , Animais , Western Blotting , Células Cultivadas , Imunofluorescência , Imuno-Histoquímica , Imunoprecipitação , Masculino , Camundongos , Transporte Proteico/fisiologia
13.
Food Chem Toxicol ; 132: 110670, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31301325

RESUMO

Bisphenol A has been restricted in a large variety of products. Bisphenol S (BPS) is its major substitute. Yet, the impacts of BPS on the central nervous system are unknown, especially in vulnerable populations like children. The aim of this study was to investigate the effects of BPS on behavioral performances and the expression of cerebral monocarboxylate transporters (MCTs). Male Swiss mice were exposed to BPS at 100 µg/kg in drinking water for 10 weeks. The protocol started after the lactation period, which is a sensitive period of early social-emotional development. Elevated T-maze and open field tests were used to measure respectively, anxiety-related and activity-related behaviors. Molecular expressions of MCTs isoforms (MCT1, MCT2, MCT4) were determined in the frontal cortex. Data showed that BPS does not affect mRNA expression of MCTs. However, BPS decreases the number of entries into the open arms and the time spent on them for BPS-treated mice. These data reveal an anxiogenic effect of BPS. For locomotor activity and exploratory behavior levels, differences did not reach a statistically significant level in the BPS-exposed group. The effect of BPS on behavioral performances unravels a putative risk for psychopathology development in early childhood and calls for more attention.


Assuntos
Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Transportadores de Ácidos Monocarboxílicos/metabolismo , Fenóis/farmacologia , Sulfonas/farmacologia , Animais , Encéfalo/metabolismo , Masculino , Camundongos , Transportadores de Ácidos Monocarboxílicos/genética , RNA Mensageiro/genética
15.
Mol Metab ; 6(12): 1625-1633, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29092796

RESUMO

OBJECTIVE: Hepatic steatosis is the first step leading to non-alcoholic fatty liver disease, which represents a major complication of obesity. Here, we show that MCT1 haploinsufficient mice resist to hepatic steatosis development when fed a high fat diet. They exhibit a reduced hepatic capacity to metabolize monocarboxylates such as lactate compared to wildtype mice. METHODS: To understand how this resistance to steatosis develops, we used HFD fed wildtype mice with hepatic steatosis and MCT1 haploinsufficient mice to study hepatic metabolism. RESULTS: AMPK is constitutively activated in the liver of MCT1 haploinsufficient mice, leading to an inactivation of SREBP1. Therefore, expression of key transcription factors for lipid metabolism, such as PPARα and γ, CHREB, or SREBP1 itself, as well as several enzymes including FAS and CPT1, was not upregulated in these mice when fed a high fat diet. It is proposed that reduced hepatic lactate metabolism is responsible for the protection against hepatic steatosis in MCT1 haploinsufficient mice via a constitutive activation of AMPK and repression of several major elements involved in hepatic lipid metabolism. CONCLUSION: Our results support a role of increased lactate uptake in hepatocytes during HFD that, in turn, induce a metabolic shift stimulating SREBP1 activity and lipid accumulation.


Assuntos
Fígado Gorduroso/metabolismo , Ácido Láctico/metabolismo , Fígado/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Proteínas Quinases/metabolismo , Simportadores/genética , Quinases Proteína-Quinases Ativadas por AMP , Animais , Fígado Gorduroso/genética , Haploinsuficiência , Metabolismo dos Lipídeos , Masculino , Camundongos , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Simportadores/metabolismo
16.
PLoS One ; 12(4): e0174990, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28388627

RESUMO

Although several in vitro and ex vivo evidence support the existence of lactate exchange between astrocytes and neurons, a direct demonstration in vivo is still lacking. In the present study, a lentiviral vector carrying a short hairpin RNA (shRNA) was used to downregulate the expression of the monocarboxylate transporter type 2 (MCT2) in neurons of the rat somatosensory cortex (called S1BF) by ~ 25%. After one hour of whisker stimulation, HRMAS 1H-NMR spectroscopy analysis of S1BF perchloric acid extracts showed that while an increase in lactate content is observed in both uninjected and shRNA-control injected extracts, such an effect was abrogated in shMCT2 injected rats. A 13C-incorporation analysis following [1-13C]glucose infusion during the stimulation confirmed that the elevated lactate observed during activation originates from newly synthesized [3-13C]lactate, with blood-derived [1-13C]glucose being the precursor. Moreover, the analysis of the 13C-labeling of glutamate in position C3 and C4 indicates that upon activation, there is an increase in TCA cycle velocity for control rats while a decrease is observed for MCT2 knockdown animals. Using in vivo localized 1H-NMR spectroscopy, an increase in lactate levels is observed in the S1BF area upon whisker stimulation for shRNA-control injected rats but not for MCT2 knockdown animals. Finally, while a robust BOLD fMRI response was evidenced in control rats, it was absent in MCT2 knockdown rats. These data not only demonstrate that glucose-derived lactate is locally produced following neuronal activation but also suggest that its use by neurons via MCT2 is probably essential to maintain synaptic activity within the barrel cortex.


Assuntos
Técnicas de Silenciamento de Genes , Ácido Láctico/metabolismo , Transportadores de Ácidos Monocarboxílicos/fisiologia , Neurônios/metabolismo , Córtex Somatossensorial/fisiologia , Vibrissas , Animais , Espectroscopia de Ressonância Magnética Nuclear de Carbono-13 , Feminino , Vetores Genéticos , Lentivirus/genética , Imageamento por Ressonância Magnética , Transportadores de Ácidos Monocarboxílicos/genética , Espectroscopia de Prótons por Ressonância Magnética , Ratos , Ratos Wistar , Córtex Somatossensorial/diagnóstico por imagem , Córtex Somatossensorial/metabolismo
17.
Sci Rep ; 6: 34909, 2016 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-27708432

RESUMO

Ketone bodies have been shown to transiently stimulate food intake and modify energy homeostasis regulatory systems following cerebral infusion for a moderate period of time (<6 hours). As ketone bodies are usually enhanced during episodes of fasting, this effect might correspond to a physiological regulation. In contrast, ketone bodies levels remain elevated for prolonged periods during obesity, and thus could play an important role in the development of this pathology. In order to understand this transition, ketone bodies were infused through a catheter inserted in the carotid to directly stimulate the brain for a period of 24 hours. Food ingested and blood circulating parameters involved in metabolic control as well as glucose homeostasis were determined. Results show that ketone bodies infusion for 24 hours increased food intake associated with a stimulation of hypothalamic orexigenic neuropeptides. Moreover, insulinemia was increased and caused a decrease in glucose production despite an increased resistance to insulin. The present study confirms that ketone bodies reaching the brain stimulates food intake. Moreover, we provide evidence that a prolonged hyperketonemia leads to a dysregulation of energy homeostasis control mechanisms. Finally, this study shows that brain exposure to ketone bodies alters insulin signaling and consequently glucose homeostasis.


Assuntos
Ingestão de Alimentos/fisiologia , Hipotálamo/metabolismo , Corpos Cetônicos/metabolismo , Ácido 3-Hidroxibutírico/farmacologia , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Artérias Carótidas , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/genética , Regulação da Expressão Gênica , Homeostase , Hipotálamo/efeitos dos fármacos , Hipotálamo/fisiopatologia , Infusões Intra-Arteriais , Resistência à Insulina , Corpos Cetônicos/genética , Corpos Cetônicos/farmacologia , Fígado/efeitos dos fármacos , Fígado/enzimologia , Camundongos Endogâmicos C57BL , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fosfoenolpiruvato Carboxiquinase (ATP)/metabolismo , Pró-Opiomelanocortina/metabolismo , Simportadores/metabolismo
18.
Front Neurosci ; 10: 509, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27877108

RESUMO

Diabetes mellitus (DM) causes important modifications in the availability and use of different energy substrates in various organs and tissues. Similarly, dietary manipulations such as high fat diets also affect systemic energy metabolism. However, how the brain adapts to these situations remains unclear. To investigate these issues, control and alloxan-induced type I diabetic rats were fed either a standard or a high fat diet enriched with advanced glycation end products (AGEs) (HAGE diet). The HAGE diet increased their levels of blood ketone bodies, and this effect was exacerbated by DM induction. To determine the effects of diet and/or DM induction on key cerebral bioenergetic parameters, both ketone bodies (ß-hydroxybutyric acid) and lactate oxidation were measured. In parallel, the expression of Monocarboxylate Transporter 1 (MCT1) and 2 (MCT2) isoforms in hippocampal and cortical slices from rats submitted to these diets was assessed. Ketone body oxidation increased while lactate oxidation decreased in hippocampal and cortical slices in both control and diabetic rats fed a HAGE diet. In parallel, the expression of both MCT1 and MCT2 increased only in the cerebral cortex in diabetic rats fed a HAGE diet. These results suggest a shift in the preferential cerebral energy substrate utilization in favor of ketone bodies in animals fed a HAGE diet, an effect that, in DM animals, is accompanied by the enhanced expression of the related transporters.

19.
J Cereb Blood Flow Metab ; 29(7): 1273-83, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19401710

RESUMO

Monocarboxylate transporters (MCTs) are essential for the use of lactate, an energy substrate known to be overproduced in brain during an ischemic episode. The expression of MCT1 and MCT2 was investigated at 48 h of reperfusion from focal ischemia induced by unilateral extradural compression in Wistar rats. Increased MCT1 mRNA expression was detected in the injured cortex and hippocampus of compressed animals compared to sham controls. In the contralateral, uncompressed hemisphere, increases in MCT1 mRNA level in the cortex and MCT2 mRNA level in the hippocampus were noted. Interestingly, strong MCT1 and MCT2 protein expression was found in peri-lesional macrophages/microglia and in an isolectin B4+/S100beta+ cell population in the corpus callosum. In vitro, MCT1 and MCT2 protein expression was observed in the N11 microglial cell line, whereas an enhancement of MCT1 expression by tumor necrosis factor-alpha (TNF-alpha) was shown in these cells. Modulation of MCT expression in microglia suggests that these transporters may help sustain microglial functions during recovery from focal brain ischemia. Overall, our study indicates that changes in MCT expression around and also away from the ischemic area, both at the mRNA and protein levels, are a part of the metabolic adaptations taking place in the brain after ischemia.


Assuntos
Encéfalo/patologia , Regulação da Expressão Gênica/fisiologia , Microglia/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Simportadores/genética , Animais , Encéfalo/metabolismo , Química Encefálica , Isquemia Encefálica , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Metabolismo Energético , Hipocampo/metabolismo , Hipocampo/patologia , Microglia/patologia , Transportadores de Ácidos Monocarboxílicos/fisiologia , RNA Mensageiro/análise , Ratos , Ratos Wistar , Simportadores/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA