101
|
Preferential transport and metabolism of glucose in Bergmann glia over Purkinje cells: A multiphoton study of cerebellar slices. Glia 2009; 57:962-70. [DOI: 10.1002/glia.20820] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
102
|
Maekawa F, Tsuboi T, Pellerin L. Regulation of the intracellular distribution, cell surface expression, and protein levels of AMPA receptor GluR2 subunits by the monocarboxylate transporter MCT2 in neuronal cells. J Neurochem 2009; 109:1767-78. [DOI: 10.1111/j.1471-4159.2009.06100.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
103
|
Pierre K, Chatton JY, Parent A, Repond C, Gardoni F, Di Luca M, Pellerin L. Linking supply to demand: the neuronal monocarboxylate transporter MCT2 and the alpha-amino-3-hydroxyl-5-methyl-4-isoxazole-propionic acid receptor GluR2/3 subunit are associated in a common trafficking process. Eur J Neurosci 2009; 29:1951-63. [PMID: 19453627 DOI: 10.1111/j.1460-9568.2009.06756.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
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.
Collapse
Affiliation(s)
- Karin Pierre
- Département de Physiologie, Université de Lausanne, 7 Rue du Bugnon, CH-1005 Lausanne, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
104
|
Castro MA, Beltrán FA, Brauchi S, Concha II. A metabolic switch in brain: glucose and lactate metabolism modulation by ascorbic acid. J Neurochem 2009; 110:423-40. [PMID: 19457103 DOI: 10.1111/j.1471-4159.2009.06151.x] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In this review, we discuss a novel function of ascorbic acid in brain energetics. It has been proposed that during glutamatergic synaptic activity neurons preferably consume lactate released from glia. The key to this energetic coupling is the metabolic activation that occurs in astrocytes by glutamate and an increase in extracellular [K(+)]. Neurons are cells well equipped to consume glucose because they express glucose transporters and glycolytic and tricarboxylic acid cycle enzymes. Moreover, neuronal cells express monocarboxylate transporters and lactate dehydrogenase isoenzyme 1, which is inhibited by pyruvate. As glycolysis produces an increase in pyruvate concentration and a decrease in NAD(+)/NADH, lactate and glucose consumption are not viable at the same time. In this context, we discuss ascorbic acid participation as a metabolic switch modulating neuronal metabolism between rest and activation periods. Ascorbic acid is highly concentrated in CNS. Glutamate stimulates ascorbic acid release from astrocytes. Ascorbic acid entry into neurons and within the cell can inhibit glucose consumption and stimulate lactate transport. For this switch to occur, an ascorbic acid flow is necessary between astrocytes and neurons, which is driven by neural activity and is part of vitamin C recycling. Here, we review the role of glucose and lactate as metabolic substrates and the modulation of neuronal metabolism by ascorbic acid.
Collapse
Affiliation(s)
- Maite A Castro
- Instituto de Bioquímica, Universidad Austral de Chile, Valdivia, Chile.
| | | | | | | |
Collapse
|
105
|
Mangia S, Giove F, Tkác I, Logothetis NK, Henry PG, Olman CA, Maraviglia B, Di Salle F, Uğurbil K. Metabolic and hemodynamic events after changes in neuronal activity: current hypotheses, theoretical predictions and in vivo NMR experimental findings. J Cereb Blood Flow Metab 2009; 29:441-63. [PMID: 19002199 PMCID: PMC2743443 DOI: 10.1038/jcbfm.2008.134] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Unraveling the energy metabolism and the hemodynamic outcomes of excitatory and inhibitory neuronal activity is critical not only for our basic understanding of overall brain function, but also for the understanding of many brain disorders. Methodologies of magnetic resonance spectroscopy (MRS) and magnetic resonance imaging (MRI) are powerful tools for the noninvasive investigation of brain metabolism and physiology. However, the temporal and spatial resolution of in vivo MRS and MRI is not suitable to provide direct evidence for hypotheses that involve metabolic compartmentalization between different cell types, or to untangle the complex neuronal microcircuitry, which results in changes of electrical activity. This review aims at describing how the current models of brain metabolism, mainly built on the basis of in vitro evidence, relate to experimental findings recently obtained in vivo by (1)H MRS, (13)C MRS, and MRI. The hypotheses related to the role of different metabolic substrates, the metabolic neuron-glia interactions, along with the available theoretical predictions of the energy budget of neurotransmission will be discussed. In addition, the cellular and network mechanisms that characterize different types of increased and suppressed neuronal activity will be considered within the sensitivity-constraints of MRS and MRI.
Collapse
Affiliation(s)
- Silvia Mangia
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minnesota 55455, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
106
|
Maekawa F, Minehira K, Kadomatsu K, Pellerin L. Basal and stimulated lactate fluxes in primary cultures of astrocytes are differentially controlled by distinct proteins. J Neurochem 2008; 107:789-98. [PMID: 18761711 DOI: 10.1111/j.1471-4159.2008.05650.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
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.
Collapse
Affiliation(s)
- Fumihiko Maekawa
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | | | | | | |
Collapse
|
107
|
Evolution and functional divergence of monocarboxylate transporter genes in vertebrates. Gene 2008; 423:14-22. [PMID: 18674605 DOI: 10.1016/j.gene.2008.07.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2008] [Revised: 06/28/2008] [Accepted: 07/03/2008] [Indexed: 11/22/2022]
Abstract
Monocarboxylate transporters (MCTs) form a gene family with an ancient past. The identification of MCTs (MCHs) from bacteria, protozoa, fungi, invertebrates, as well as vertebrates, but not from plants and virus, allowed illuminating the phylogenetic and evolutionary history of this gene family. The significant expansion of vertebrate MCT genes should have primarily occurred after the divergence of vertebrates and invertebrates, but before the divergence time between ray-finned fish and mammals. The divergence of insect MCTs should have at least occurred in the common ancestor of fruit fly, beetle, and honeybee. Fungi monocarboxylate transporter homologues (MCHs) might evolve independently from an ancient ancestor. The results of functional divergence analysis provided statistical evidences for shifted evolutionary rate and/or changes of amino acid property after gene duplication. The sliding window analysis of the d(N)/d(S) ratio values showed that strong functional constraints must impose on the N- and C-terminal domains of vertebrate MCTs. These corresponding regions may play crucial roles for functionality of MCT proteins.
Collapse
|
108
|
Chiry O, Fishbein WN, Merezhinskaya N, Clarke S, Galuske R, Magistretti PJ, Pellerin L. Distribution of the monocarboxylate transporter MCT2 in human cerebral cortex: an immunohistochemical study. Brain Res 2008; 1226:61-9. [PMID: 18598673 DOI: 10.1016/j.brainres.2008.06.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2008] [Revised: 05/14/2008] [Accepted: 06/18/2008] [Indexed: 10/21/2022]
Abstract
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.
Collapse
Affiliation(s)
- Oriana Chiry
- Max Planck Institute for Brain Research, Deutschordenstrasse 46, 60528 Frankfurt am Main, Germany
| | | | | | | | | | | | | |
Collapse
|
109
|
Erlichman JS, Hewitt A, Damon TL, Hart M, Kurascz J, Li A, Leiter JC. Inhibition of monocarboxylate transporter 2 in the retrotrapezoid nucleus in rats: a test of the astrocyte-neuron lactate-shuttle hypothesis. J Neurosci 2008; 28:4888-96. [PMID: 18463242 PMCID: PMC2645067 DOI: 10.1523/jneurosci.5430-07.2008] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2007] [Revised: 02/14/2008] [Accepted: 03/18/2008] [Indexed: 01/23/2023] Open
Abstract
The astrocyte-neuronal lactate-shuttle hypothesis posits that lactate released from astrocytes into the extracellular space is metabolized by neurons. The lactate released should alter extracellular pH (pHe), and changes in pH in central chemosensory regions of the brainstem stimulate ventilation. Therefore, we assessed the impact of disrupting the lactate shuttle by administering 100 microM alpha-cyano-4-hydroxy-cinnamate (4-CIN), a dose that blocks the neuronal monocarboxylate transporter (MCT) 2 but not the astrocytic MCTs (MCT1 and MCT4). Administration of 4-CIN focally in the retrotrapezoid nucleus (RTN), a medullary central chemosensory nucleus, increased ventilation and decreased pHe in intact animals. In medullary brain slices, 4-CIN reduced astrocytic intracellular pH (pHi) slightly but alkalinized neuronal pHi. Nonetheless, pHi fell significantly in both cell types when they were treated with exogenous lactate, although 100 microM 4-CIN significantly reduced the magnitude of the acidosis in neurons but not astrocytes. Finally, 4-CIN treatment increased the uptake of a fluorescent 2-deoxy-D-glucose analog in neurons but did not alter the uptake rate of this 2-deoxy-D-glucose analog in astrocytes. These data confirm the existence of an astrocyte to neuron lactate shuttle in intact animals in the RTN, and lactate derived from astrocytes forms part of the central chemosensory stimulus for ventilation in this nucleus. When the lactate shuttle was disrupted by treatment with 4-CIN, neurons increased the uptake of glucose. Therefore, neurons seem to metabolize a combination of glucose and lactate (and other substances such as pyruvate) depending, in part, on the availability of each of these particular substrates.
Collapse
Affiliation(s)
- Joseph S. Erlichman
- Department of Biology, St. Lawrence University, Canton, New York 13617-1475, and
| | - Amy Hewitt
- Department of Biology, St. Lawrence University, Canton, New York 13617-1475, and
| | - Tracey L. Damon
- Department of Biology, St. Lawrence University, Canton, New York 13617-1475, and
| | - Michael Hart
- Department of Biology, St. Lawrence University, Canton, New York 13617-1475, and
| | - Jennifer Kurascz
- Department of Biology, St. Lawrence University, Canton, New York 13617-1475, and
| | - Aihua Li
- Department of Physiology, Dartmouth Medical School, Lebanon, New Hampshire 03756
| | - James C. Leiter
- Department of Physiology, Dartmouth Medical School, Lebanon, New Hampshire 03756
| |
Collapse
|
110
|
Kuluz J, Huang T, Watson B, Vannucci S. Stroke in the immature brain: review of pathophysiology and animal models of pediatric stroke. FUTURE NEUROLOGY 2008. [DOI: 10.2217/14796708.3.2.199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Pediatric stroke research presents many challenges. Relatively low incidence, need for age stratification, diverse etiologies, delays in diagnosis, lack of an established age-based stroke severity scale and outcome measures are only some of the issues that have prevented the implementation of clinical trials in infants and children with stroke. Experimental animal models of pediatric stroke, therefore, are critical to understanding the pathophysiology and management of ischemic brain damage in the immature brain, and provide the necessary platform for future clinical trials. In this review we discuss the pertinent clinical aspects of pediatric stroke, the pathophysiology of stroke in the developing brain and the animal models established to study basic mechanisms as well as translational issues in pediatric stroke.
Collapse
Affiliation(s)
- John Kuluz
- Associate Professor of Pediatrics, University of Miami, Department of Pediatrics (R-131), Miller School of Medicine, PO Box 016960, Miami, FL 33101, USA
| | - Tingting Huang
- Post-Doctoral Research Associate, University of Miami, Department of Pediatrics (R-131), Miller School of Medicine, PO Box 016960 Miami, FL 33101, USA
| | - Brant Watson
- Professor of Neurology, University of Miami, Department of Neurology (D4–5), Miller School of Medicine, PO Box 016960, Miami, FL 33136, USA
| | - Susan Vannucci
- Research Professor of Neuroscience in Pediatrics/Newborn Medicine, Weill Cornell Medical College, 525 East 68th Street, N-506, NY 10065, USA
| |
Collapse
|
111
|
Chenal J, Pierre K, Pellerin L. Insulin and IGF-1 enhance the expression of the neuronal monocarboxylate transporter MCT2 by translational activation via stimulation of the phosphoinositide 3-kinase-Akt-mammalian target of rapamycin pathway. Eur J Neurosci 2007; 27:53-65. [PMID: 18093179 DOI: 10.1111/j.1460-9568.2007.05981.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
MCT2 is the main neuronal monocarboxylate transporter essential for facilitating lactate and ketone body utilization as energy substrates. Our study reveals that treatment of cultured cortical neurons with insulin and IGF-1 led to a striking enhancement of MCT2 immunoreactivity in a time- and concentration-dependent manner. Surprisingly, neither insulin nor IGF-1 affected MCT2 mRNA expression, suggesting that regulation of MCT2 protein expression occurs at the translational rather than the transcriptional level. Investigation of the putative signalling pathways leading to translation activation revealed that insulin and IGF-1 induced p44- and p42 MAPK, Akt and mTOR phosphorylation. S6 ribosomal protein, a component of the translational machinery, was also strongly activated by insulin and IGF-1. Phosphorylation of p44- and p42 MAPK was blocked by the MEK inhibitor PD98058, while Akt phosphorylation was abolished by the PI3K inhibitor LY294002. Phosphorylation of mTOR and S6 was blocked by the mTOR inhibitor rapamycin. In parallel, it was observed that LY294002 and rapamycin almost completely blocked the effects of insulin and IGF-1 on MCT2 protein expression, whereas PD98059 and SB202190 (a p38K inhibitor) had no effect on insulin-induced MCT2 expression and only a slight effect on IGF-1-induced MCT2 expression. At the subcellular level, a significant increase in MCT2 protein expression within an intracellular pool was observed while no change at the cell surface was apparent. As insulin and IGF-1 are involved in synaptic plasticity, their effect on MCT2 protein expression via an activation of the PI3K-Akt-mTOR-S6K pathway might contribute to the preparation of neurons for enhanced use of nonglucose energy substrates following altered synaptic efficacy.
Collapse
Affiliation(s)
- Julie Chenal
- Department of Physiology, Université de Lausanne, 7 Rue du Bugnon, 1005 Lausanne, Switzerland
| | | | | |
Collapse
|
112
|
Simpson IA, Carruthers A, Vannucci SJ. Supply and demand in cerebral energy metabolism: the role of nutrient transporters. J Cereb Blood Flow Metab 2007; 27:1766-91. [PMID: 17579656 PMCID: PMC2094104 DOI: 10.1038/sj.jcbfm.9600521] [Citation(s) in RCA: 602] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Glucose is the obligate energetic fuel for the mammalian brain, and most studies of cerebral energy metabolism assume that the majority of cerebral glucose utilization fuels neuronal activity via oxidative metabolism, both in the basal and activated state. Glucose transporter (GLUT) proteins deliver glucose from the circulation to the brain: GLUT1 in the microvascular endothelial cells of the blood-brain barrier (BBB) and glia; GLUT3 in neurons. Lactate, the glycolytic product of glucose metabolism, is transported into and out of neural cells by the monocarboxylate transporters (MCT): MCT1 in the BBB and astrocytes and MCT2 in neurons. The proposal of the astrocyte-neuron lactate shuttle hypothesis suggested that astrocytes play the primary role in cerebral glucose utilization and generate lactate for neuronal energetics, especially during activation. Since the identification of the GLUTs and MCTs in brain, much has been learned about their transport properties, that is capacity and affinity for substrate, which must be considered in any model of cerebral glucose uptake and utilization. Using concentrations and kinetic parameters of GLUT1 and -3 in BBB endothelial cells, astrocytes, and neurons, along with the corresponding kinetic properties of the MCTs, we have successfully modeled brain glucose and lactate levels as well as lactate transients in response to neuronal stimulation. Simulations based on these parameters suggest that glucose readily diffuses through the basal lamina and interstitium to neurons, which are primarily responsible for glucose uptake, metabolism, and the generation of the lactate transients observed on neuronal activation.
Collapse
Affiliation(s)
- Ian A Simpson
- Department of Neural and Behavioral Sciences College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA.
| | | | | |
Collapse
|
113
|
Obara M, Szeliga M, Albrecht J. Regulation of pH in the mammalian central nervous system under normal and pathological conditions: facts and hypotheses. Neurochem Int 2007; 52:905-19. [PMID: 18061308 DOI: 10.1016/j.neuint.2007.10.015] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2007] [Revised: 10/17/2007] [Accepted: 10/22/2007] [Indexed: 11/27/2022]
Abstract
The maintenance of pH homeostasis in the CNS is of key importance for proper execution and regulation of neurotransmission, and deviations from this homeostasis are a crucial factor in the mechanism underlying a spectrum of pathological conditions. The first few sections of the review are devoted to the brain operating under normal conditions. The article commences with an overview of how extrinsic factors modelling the brain at work: neurotransmitters, depolarising stimuli (potassium and voltage changes) and cyclic nucleotides as major signal transducing vehicles affect pH in the CNS. Further, consequences of pH alterations on the major aspects of CNS function and metabolism are outlined. Next, the major cellular events involved in the transport, sequestration, metabolic production and buffering of protons that are common to all the mammalian cells, including the CNS cells. Since CNS function reflects tight interaction between astrocytes and neurons, the pH regulatory events pertinent to either cell type are discussed: overwhelming evidence implicates astrocytes as a key player in pH homeostasis in the brain. The different classes of membrane proteins involved in proton shuttling are listed and their mechanisms of action are given. These include: the Na+/H+ exchanger, different classes of bicarbonate transporters acting in a sodium-dependent- or -independent mode, monocarboxylic acid transporters and the vacuolar-type proton ATPase. A separate section is devoted to carbonic anhydrase, which is represented by multiple isoenzymes capable of pH buffering both in the cell interior and in the extracellular space. Next, impairment of pH regulation and compensatory responses occurring in brain affected by different pathologies: hypoxia/ischemia, epilepsy, hyperammonemic encephalopathies, cerebral tumours and HIV will be described. The review is limited to facts and plausible hypotheses pertaining to phenomena directly involved in pH regulation: changes in pH that accompany metabolic stress but have no distinct implications for the pH regulatory mechanisms are not dealt with. In most cases, the vast body of knowledge derived from in vitro studies remains to be verified in in vivo settings.
Collapse
Affiliation(s)
- Marta Obara
- Department of Neurotoxicology, Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Street, 02-106 Warsaw, Poland
| | | | | |
Collapse
|
114
|
Zhang SXL, Searcy TR, Wu Y, Gozal D, Wang Y. Alternative promoter usage and alternative splicing contribute to mRNA heterogeneity of mouse monocarboxylate transporter 2. Physiol Genomics 2007; 32:95-104. [PMID: 17911380 DOI: 10.1152/physiolgenomics.00192.2007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Expression patterns of monocarboxylate transporter 2 (MCT2) display mRNA diversity in a tissue-specific fashion. We cloned and characterized multiple mct2 5'-cDNA ends from the mouse and determined the structural organization of the mct2 gene. We found that transcription of this gene was initiated from five independent genomic regions that spanned >80 kb on chromosome 10, resulting in five unique exon 1 variants (exons 1a, 1b, 1c, 1d, and 1e) that were then spliced to the common exon 2. Alternative splicing of four internal exons (exons AS1, AS2, AS3, and exon 3) greatly increased the complexity of mRNA diversity. While exon 1c was relatively commonly used for transcription initiation in various tissues, other exon 1 variants were used in a tissue-specific fashion, especially exons 1b and 1d that were used exclusively for testis-specific expression. Sequence analysis of 5'-flanking regions upstream of exons 1a, 1b, and 1c revealed the presence of numerous potential binding sites for ubiquitous transcription factors in all three regions and for transcription factors implicated in testis-specific or hypoxia-induced gene expression in the 1b region. Transient transfection assays demonstrated that each of the three regions contained a functional promoter and that the in vitro, cell type-specific activities of these promoters were consistent with the tissue-specific expression pattern of the mct2 gene in vivo. These results indicate that tissue-specific expression of the mct2 gene is controlled by multiple alternative promoters and that both alternative promoter usage and alternative splicing contribute to the remarkable mRNA diversity of the gene.
Collapse
Affiliation(s)
- Shelley X L Zhang
- Kosair Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, Kentucky 40202, USA
| | | | | | | | | |
Collapse
|
115
|
Baker KD, Edwards TM. d-Lactate inhibition of memory in a single trial discrimination avoidance task in the young chick. Neurobiol Learn Mem 2007; 88:269-76. [PMID: 17692538 DOI: 10.1016/j.nlm.2007.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2007] [Revised: 06/17/2007] [Accepted: 06/19/2007] [Indexed: 11/26/2022]
Abstract
L-Lactate is a metabolite possibly able to meet some neuronal energy demands. However, a clear role for L-lactate in behaviour remains elusive. Administration of the inactive isomer D-lactate (1.75 mM; ic), immediately post-training, resulted in a persistent retention loss from 40 min post-training when used in conjuction with a single trial discrimination avoidance task designed for the young chick. Furthermore, 1mM noradrenaline (ic) administered 20 min post-training overcame the retention loss induced by D-lactate. Although not directly demonstrated in the current study, it is plausible that D-lactate inhibited memory processing by competing with L-lactate for uptake into neurons. The time of onset of the retention loss induced by D-lactate is in accord with findings where the action of noradrenaline is inhibited. The successful challenge of D-lactate inhibition by a high concentration of noradrenaline may suggest a relationship by some unidentified mechanism.
Collapse
Affiliation(s)
- K D Baker
- School of Psychology, Psychiatry and Psychological Medicine, Monash University, 3800 Vic., Australia
| | | |
Collapse
|
116
|
Atlante A, de Bari L, Bobba A, Marra E, Passarella S. Transport and metabolism of L-lactate occur in mitochondria from cerebellar granule cells and are modified in cells undergoing low potassium dependent apoptosis. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2007; 1767:1285-99. [PMID: 17950241 DOI: 10.1016/j.bbabio.2007.08.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Revised: 07/24/2007] [Accepted: 08/20/2007] [Indexed: 10/22/2022]
Abstract
Having confirmed that externally added L-lactate can enter cerebellar granule cells, we investigated whether and how L-lactate is metabolized by mitochondria from these cells under normal or apoptotic conditions. (1) L-lactate enters mitochondria, perhaps via an L-lactate/H+ symporter, and is oxidized in a manner stimulated by ADP. The existence of an L-lactate dehydrogenase, located in the inner mitochondrial compartment, was shown by immunological analysis. Neither the protein level nor the Km and Vmax values changed en route to apoptosis. (2) In both normal and apoptotic cell homogenates, externally added L-lactate caused reduction of the intramitochondrial pyridine cofactors, inhibited by phenylsuccinate. This process mirrored L-lactate uptake by mitochondria and occurred with a hyperbolic dependence on L-lactate concentrations. Pyruvate appeared outside mitochondria as a result of external addition of L-lactate. The rate of the process depended on L-lactate concentration and showed saturation characteristics. This shows the occurrence of an intracellular L-lactate/pyruvate shuttle, whose activity was limited by the putative L-lactate/pyruvate antiporter. Both the carriers were different from the monocarboxylate carrier. (3) L-lactate transport changed en route to apoptosis. Uptake increased in the early phase of apoptosis, but decreased in the late phase with characteristics of a non-competitive like inhibition. In contrast, the putative L-lactate/pyruvate antiport decreased en route to apoptosis with characteristics of a competitive like inhibition in early apoptosis, and a mixed non-competitive like inhibition in late apoptosis.
Collapse
Affiliation(s)
- Anna Atlante
- Istituto di Biomembrane e Bioenergetica, Consiglio Nazionale delle Ricerche, Via G Amendola, 165/A-70126, Bari, Italy.
| | | | | | | | | |
Collapse
|
117
|
Pierre K, Parent A, Jayet PY, Halestrap AP, Scherrer U, Pellerin L. Enhanced expression of three monocarboxylate transporter isoforms in the brain of obese mice. J Physiol 2007; 583:469-86. [PMID: 17599960 PMCID: PMC2277016 DOI: 10.1113/jphysiol.2007.138594] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Monocarboxylate transporters (MCTs) are membrane carriers for lactate and ketone bodies. Three isoforms, MCT1, MCT2 and MCT4, have been described in the central nervous system but little information is available about the regulation of their expression in relation to altered metabolic and/or nutritional conditions. We show here that brains of mice fed on a high fat diet (HFD) up to 12 weeks as well as brains of genetically obese (ob/ob) or diabetic (db/db) mice exhibit an increase of MCT1, MCT2 and MCT4 expression as compared to brains of control mice fed a standard diet. Enhanced expression of each transporter was visible throughout the brain but most prominently in the cortex and in the hippocampus. Using immunohistochemistry, we observed that neurons (expressing mainly MCT2 but also sometimes low levels of MCT1 under normal conditions) were immunolabelled for all three transporters in HFD mice as well as in ob/ob and db/db mice. At the subcellular level, changes were most remarkable in neuronal cell bodies. Western blotting performed on brain structure extracts allowed us to confirm quantitatively the enhancement of MCT1 and MCT2 expression. Our data demonstrate that the expression of cerebral MCT isoforms can be modulated by alterations of peripheral metabolism, suggesting that the adult brain is sensitive and adapts to new metabolic states. This observation could be relevant in the context of obesity development and its consequences for brain function.
Collapse
Affiliation(s)
- Karin Pierre
- Departement de Physiologie, Universite de Lausanne, Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
118
|
Wang Q, Lu Y, Yuan M, Darling IM, Repasky EA, Morris ME. Characterization of monocarboxylate transport in human kidney HK-2 cells. Mol Pharm 2007; 3:675-85. [PMID: 17140255 DOI: 10.1021/mp060037b] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The objectives of this study were to characterize the expression and function of monocarboxylate transporters (MCTs) in human kidney HK-2 cells and to compare the expression of MCTs in HK-2 cells to that found in human kidney. mRNA and protein expression of MCTs were determined by RT-PCR and Western analyses, respectively, while immunofluorescence staining was used to determine the membrane localization of MCT1. The driving force, transport kinetics, and inhibition of two MCT substrates, D-lactate and butyrate, were characterized in HK-2 cells. mRNA of MCT1, -2, -3, -4 isoforms were present in HK-2 cells and in human kidney cortex. MCT1 was present predominantly on the basal membranes of HK-2 cells. The cellular uptake of D-lactate and butyrate exhibited pH- and concentration-dependence (D-lactate, Km of 26.5 +/- 2.2 mM and Vmax of 72.0 +/- 14.5 nmol mg-1 min-1; butyrate, Km of 0.8 +/- 0.3 mM, Vmax of 29.3 +/- 2.5 nmol mg-1 min-1, and a diffusional clearance of 2.1 microL mg-1 min-1). The uptake of D-lactate and butyrate by HK-2 cells was inhibited by MCT analogues and the classical MCT inhibitors alpha-cyano-4-hydroxycinnamate, pCMB, and phloretin. The uptake of D-lactate and butyrate by HK-2 cells significantly decreased after transfection with small-interference RNA for MCT1. In summary, MCTs were present in both HK-2 cells and human kidney cortex, and HK-2 cells exhibited polarized MCT expression and pH-dependent transport of D-lactate and butyrate. Our results also support the usefulness of HK-2 cells as an in vitro model for studying monocarboxylate transport in renal proximal tubule cells.
Collapse
Affiliation(s)
- Qi Wang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Amherst, New York 14260, USA
| | | | | | | | | | | |
Collapse
|
119
|
Chenal J, Pellerin L. Noradrenaline enhances the expression of the neuronal monocarboxylate transporter MCT2 by translational activation via stimulation of PI3K/Akt and the mTOR/S6K pathway. J Neurochem 2007; 102:389-97. [PMID: 17394554 DOI: 10.1111/j.1471-4159.2007.04495.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Monocarboxylate transporter 2 (MCT2) expression is up-regulated by noradrenaline (NA) in cultured cortical neurons via a putative but undetermined translational mechanism. Western blot analysis showed that p44/p42 mitogen-activated protein kinase (MAPK) was rapidly and strongly phosphorylated by NA treatment. NA also rapidly induced serine/threonine protein kinase from AKT virus (Akt) phosphorylation but to a lesser extent than p44/p42 MAPK. However, Akt activation persisted over a longer period. Similarly, NA induced a rapid and persistent phosphorylation of mammalian target of rapamycin (mTOR), a kinase implicated in the regulation of translation in the central nervous system. Consistent with activation of the mTOR/S6 kinase pathway, phosphorylation of the ribosomal S6 protein, a component of the translation machinery, could be observed upon treatment with NA. In parallel, it was found that the NA-induced increase in MCT2 protein was almost completely blocked by LY294002 (phosphoinositide 3-kinase inhibitor) as well as by rapamycin (mTOR inhibitor), while mitogen-activated protein kinase kinase and p38 MAPK inhibitors had much smaller effects. Taken together, these data reveal that NA induces an increase in neuronal MCT2 protein expression by a mechanism involving stimulation of phosphoinositide 3-kinase/Akt and translational activation via the mTOR/S6 kinase pathway. Moreover, considering the role of NA in synaptic plasticity, alterations in MCT2 expression as described in this study might represent an adaptation to face energy demands associated with enhanced synaptic transmission.
Collapse
Affiliation(s)
- Julie Chenal
- Département de Physiologie, Université de Lausanne, Lausanne, Switzerland
| | | |
Collapse
|
120
|
O'Brien J, Kla KM, Hopkins IB, Malecki EA, McKenna MC. Kinetic Parameters and Lactate Dehydrogenase Isozyme Activities Support Possible Lactate Utilization by Neurons. Neurochem Res 2006; 32:597-607. [PMID: 17006762 DOI: 10.1007/s11064-006-9132-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2006] [Indexed: 10/24/2022]
Abstract
Lactate is potentially a major energy source in brain, particularly following hypoxia/ischemia; however, the regulation of brain lactate metabolism is not well understood. Lactate dehydrogenase (LDH) isozymes in cytosol from primary cultures of neurons and astrocytes, and freshly isolated synaptic terminals (synaptosomes) from adult rat brain were separated by electrophoresis, visualized with an activity-based stain, and quantified. The activity and kinetics of LDH were determined in the same preparations. In synaptosomes, the forward reaction (pyruvate + NADH + H(+ )--> lactate + NAD(+)), which had a V (max) of 1,163 micromol/min/mg protein was 62% of the rate in astrocyte cytoplasm. In contrast, the reverse reaction (lactate + NAD(+ )--> pyruvate + NADH + H(+)), which had a V (max) of 268 micromol/min/mg protein was 237% of the rate in astrocytes. Although the relative distribution was different, all five isozymes of LDH were present in synaptosomes and primary cultures of cortical neurons and astrocytes from rat brain. LDH1 was 14.1% of the isozyme in synaptic terminals, but only 2.6% and 2.4% in neurons and astrocytes, respectively. LDH5 was considerably lower in synaptic terminals than in neurons and astrocytes, representing 20.4%, 37.3% and 34.8% of the isozyme in these preparations, respectively. The distribution of LDH isozymes in primary cultures of cortical neurons does not directly reflect the kinetics of LDH and the capacity for lactate oxidation. However, the kinetics of LDH in brain are consistent with the possible release of lactate by astrocytes and oxidative use of lactate for energy in synaptic terminals.
Collapse
Affiliation(s)
- Janet O'Brien
- Department of Pediatrics, University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, MD 21201-1559, USA
| | | | | | | | | |
Collapse
|
121
|
Prins ML, Giza CC. Induction of Monocarboxylate Transporter 2 Expression and Ketone Transport following Traumatic Brain Injury in Juvenile and Adult Rats. Dev Neurosci 2006; 28:447-56. [PMID: 16943667 DOI: 10.1159/000094170] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2005] [Accepted: 03/11/2006] [Indexed: 11/19/2022] Open
Abstract
Based on recent work demonstrating age-dependent ketogenic neuroprotection after traumatic brain injury (TBI), it was hypothesized that the neuroprotection among early post-weaned animals was related to induced cerebral transport of ketones after injury. Regional changes in monocarboxylate transporter 2 (MCT2) were acutely examined with immunohistochemistry after sham surgery or controlled cortical impact injury among postnatal day 35 and adult rats. Both ages showed elevated MCT2 expression in the ipsilateral cerebral vasculature after TBI. Using Western blotting, MCT2 expression was 80-88% greater in microvessels isolated from postnatal day 35 rats at all time points relative to adults. The increased MCT2 expression was temporally correlated with an age-related increase in cerebral uptake of ketones, when ketones were made available after injury.
Collapse
Affiliation(s)
- M L Prins
- Division of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-7039, USA.
| | | |
Collapse
|
122
|
Chiry O, Pellerin L, Monnet-Tschudi F, Fishbein WN, Merezhinskaya N, Magistretti PJ, Clarke S. Expression of the monocarboxylate transporter MCT1 in the adult human brain cortex. Brain Res 2006; 1070:65-70. [PMID: 16403470 DOI: 10.1016/j.brainres.2005.11.064] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2005] [Revised: 11/03/2005] [Accepted: 11/06/2005] [Indexed: 11/26/2022]
Abstract
Distribution of the monocarboxylate transporter MCT1 has been investigated in the cortex of normal adult human brain. Similarly to the glucose transporter GLUT1 55 kDa isoform, MCT1 was found to be strongly expressed on blood vessels in all cortical layers. In addition, laminar analysis revealed intense MCT1 expression in the neuropil of layer IV in primary auditory (AI) and visual (VI) areas, while this expression was more homogeneous in the non-primary auditory area STA. The cellular distribution shows that MCT1 is strongly expressed by glial cells often associated with blood vessels that were identified as astrocytes. The observed distribution of MCT1 supports the concept that, under certain circumstances, monocarboxylates could be provided as energy substrates to the adult human brain. Moreover, the distinct laminar pattern of MCT1 expression between primary and non-primary cortical areas may reflect different types of neuronal activity requiring adequate supply of specific energy substrates.
Collapse
Affiliation(s)
- Oriana Chiry
- Département de Physiologie, Université de Lausanne, rue du Bugnon 7, 1005 Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
123
|
Mavroudis G, Prior MJW, Lister T, Nolan CC, Ray DE. Neurochemical and oedematous changes in 1,3-dinitrobenzene-induced astroglial injury in rat brain from a 1H-nuclear magnetic resonance perspective. J Neural Transm (Vienna) 2005; 113:1263-78. [PMID: 16362630 DOI: 10.1007/s00702-005-0395-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2005] [Accepted: 10/09/2005] [Indexed: 12/19/2022]
Abstract
1,3-Dinitrobenzene (1,3-DNB), an intermediate used in the chemical industry, has toxic effects in the brain and testes. It produces focal lesions with marked astroglial necrosis in the rat brain upon repeated administration. Astrocytic death occurs in parallel with elevated local blood flow and is followed by damage to the cerebral vasculature and neurones. (1)H-nuclear magnetic resonance spectroscopic analysis before the onset of astrocytic damage, showed a global elevation of lactate, whereas choline containing compounds increased in the non-vulnerable cerebral cortex, yet decreased in the vulnerable brainstem. Similarly, glutamate increased in the cerebral cortex, cerebellum and midbrain, but decreased in the susceptible brainstem. In vivo T2-weighted NMR imaging showed high signal intensities in brain nuclei shown to develop astroglial loss by conventional neuropathology at 24 hours after completion of dosing, but not at 6-10 hours. Hence the early neurochemical changes in susceptible areas contribute to the aetiology of degeneration, and those seen elsewhere may represent adaptive responses dependent on the particular phenotype of different cell groups and underlying metabolic relationships.
Collapse
Affiliation(s)
- G Mavroudis
- MRC Toxicology Unit, University of Leicester, Hodgkin Building, Leicester, United Kingdom.
| | | | | | | | | |
Collapse
|
124
|
Smith JP, Drewes LR. Modulation of monocarboxylic acid transporter-1 kinetic function by the cAMP signaling pathway in rat brain endothelial cells. J Biol Chem 2005; 281:2053-60. [PMID: 16301311 DOI: 10.1074/jbc.m511577200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
MCT1 (monocarboxylic acid transporter 1) facilitates bidirectional monocarboxylic acid transport across membranes. MCT1 function and regulation have not been characterized previously in cerebral endothelial cells but may be important during normal cerebral energy metabolism and during brain diseases such as stroke. Here, by using the cytoplasmic pH indicator 2',7'-bis-(2-carboxyethyl)-5-(and-6)-carboxyfluorescein-acetoxymethyl ester, the initial rates of monocarboxylate-dependent cytoplasmic acidification were measured as an indication of MCT1 kinetic function in vitro using the rat brain endothelial cell (RBE4) model of blood-brain transport. The initial rate of L-lactate-dependent acidification was significantly inhibited by 5-10-min incubations with agonists of intracellular cAMP-dependent cell signaling pathways as follows: dibutyryl cAMP, forskolin, and isoproterenol. Isoproterenol reduced V(max) but did not affect K(m) values. The effects of forskolin were completely reversed by the protein kinase A inhibitor H89, whereas H89 alone increased transport rates. Cytoplasmic cAMP levels, measured by radioimmunoassay, were increased by forskolin or isoproterenol, and the effect of isoproterenol was inhibited by propranolol. MCT1-independent intracellular pH control mechanisms did not contribute to the forskolin or H89 effects on MCT1 kinetic function as determined with amiloride, monocarboxylate-independent acid loading, or the transport inhibitor alpha-cyano-4-hydroxycinnamate. The data demonstrate the direct modulation of MCT1 kinetic function in cerebral endothelial cells by agents known to affect the beta-adrenergic receptor/adenylyl cyclase/cAMP/protein kinase A intracellular signaling pathway.
Collapse
Affiliation(s)
- Jeffrey P Smith
- Department of Biochemistry and Molecular Biology, University of Minnesota Medical School, 1035 University Drive, Duluth, MN 55812, USA
| | | |
Collapse
|
125
|
Tekkök SB, Brown AM, Westenbroek R, Pellerin L, Ransom BR. Transfer of glycogen-derived lactate from astrocytes to axons via specific monocarboxylate transporters supports mouse optic nerve activity. J Neurosci Res 2005; 81:644-52. [PMID: 16015619 DOI: 10.1002/jnr.20573] [Citation(s) in RCA: 170] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
It is hypothesized that L-lactate derived from astrocyte glycogen sustains axon excitability in mouse optic nerve (MON). This theory was tested by using a competitive antagonist of L-lactate transport and immunocytochemistry to determine whether transport proteins are appropriately distributed in adult MON. L-lactate sustained the compound action potential (CAP), indicating that exogenous L-lactate was an effective energy substrate. During 60 min of aglycemia, the CAP persisted for 30 min, surviving on a glycogen-derived substrate (probably lactate), before failing. After failing, the CAP could be partially rescued by restoring 10 mM glucose or 20 mM L-lactate. Aglycemia in the presence of 20 mM D-lactate, a metabolically inert but transportable monocarboxylate, resulted in accelerated CAP decline compared with aglycemia alone, suggesting that D-lactate blocked the axonal uptake of glycogen-derived L-lactate, speeding the onset of energy failure and loss of the CAP. The CAP was maintained for up to 2 hr when exposed to 20% of normal bath glucose (i.e., 2 mM). To test whether glycogen-derived L-lactate "supplemented" available glucose (2 mM) in supporting metabolism, L-lactate uptake into axons was reduced by the competitive inhibitor D-lactate. Indeed, in the presence of 20 mM D-lactate, the CAP was lost more rapidly in MONs bathed in 2 mM glucose artificial cerebrospinal fluid. Immunocytochemical staining demonstrated cell-specific expression of monocarboxylate transporter (MCT) subtypes, localizing MCT2 predominantly to axons and MCT1 predominantly to astrocytes, supporting the idea that L-lactate is released from astrocytes and taken up by axons as an energy source for sustaining axon excitability.
Collapse
Affiliation(s)
- Selva Baltan Tekkök
- Department of Neurology, University of Washington School of Medicine, Seattle, Washington, USA
| | | | | | | | | |
Collapse
|
126
|
Pellerin L. How astrocytes feed hungry neurons. Mol Neurobiol 2005; 32:59-72. [PMID: 16077184 DOI: 10.1385/mn:32:1:059] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2004] [Accepted: 12/17/2004] [Indexed: 11/11/2022]
Abstract
For years glucose was thought to constitute the sole energy substrate for neurons; it was believed to be directly provided to neurons via the extracellular space by the cerebral circulation. It was recently proposed that in addition to glucose, neurons might rely on lactate to sustain their activity. Therefore, it was demonstrated that lactate is a preferred oxidative substrate for neurons not only in vitro but also in vivo. Moreover, the presence of specific monocarboxylate transporters on neurons as well as on astrocytes is consistent with the hypothesis of a transfer of lactate from astrocytes to neurons. Evidence has been provided for a mechanism whereby astrocytes respond to glutamatergic activity by enhancing their glycolytic activity, resulting in increased lactate release. This is accomplished via the uptake of glutamate by glial glutamate transporters, leading to activation of the Na+/K+ ATPase and a stimulation of astrocytic glycolysis. Several recent observations obtained both in vitro and in vivo with different approaches have reinforced this view of brain energetics. Such an understanding might be critically important, not only because it forms the basis of some classical functional brain imaging techniques but also because several neurodegenerative diseases exhibit diverse alterations in energy metabolism.
Collapse
Affiliation(s)
- Luc Pellerin
- Département de Physiologie, Université de Lausanne, Switzerland.
| |
Collapse
|
127
|
Pierre K, Pellerin L. Monocarboxylate transporters in the central nervous system: distribution, regulation and function. J Neurochem 2005; 94:1-14. [PMID: 15953344 DOI: 10.1111/j.1471-4159.2005.03168.x] [Citation(s) in RCA: 508] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Monocarboxylate transporters (MCTs) are proton-linked membrane carriers involved in the transport of monocarboxylates such as lactate, pyruvate, as well as ketone bodies. They belong to a larger family of transporters composed of 14 members in mammals based on sequence homologies. MCTs are found in various tissues including the brain where three isoforms, MCT1, MCT2 and MCT4, have been described. Each of these isoforms exhibits a distinct regional and cellular distribution in rodent brain. At the cellular level, MCT1 is expressed by endothelial cells of microvessels, by ependymocytes as well as by astrocytes. MCT4 expression appears to be specific for astrocytes. By contrast, the predominant neuronal monocarboxylate transporter is MCT2. Interestingly, part of MCT2 immunoreactivity is located at postsynaptic sites, suggesting a particular role of monocarboxylates and their transporters in synaptic transmission. In addition to variation in expression during development and upon nutritional modifications, new data indicate that MCT expression is regulated at the translational level by neurotransmitters. Understanding how transport of monocarboxylates is regulated could be of particular importance not only for neuroenergetics but also for areas such as functional brain imaging, regulation of food intake and glucose homeostasis, or for central nervous system disorders such as ischaemia and neurodegenerative diseases.
Collapse
Affiliation(s)
- Karin Pierre
- Département de Physiologie, Université de Lausanne, Switzerland
| | | |
Collapse
|
128
|
Abstract
Astrocytes are multifunctional cells that interact with neurons and other astrocytes in signaling and metabolic functions, and their resistance to pathophysiological conditions can help restrict loss of tissue after an ischemic event provided adequate nutrients are supplied to support their requirements. Astrocytes have substantial oxidative capacity and mechanisms to upregulate glycolytic capability when respiration is impaired. An astrocytic enzyme that synthesizes a powerful activator of glycolysis is not present in neurons, endowing astrocytes with the ability to sustain ATP production under restrictive conditions. The monocarboxylic acid transporter (MCT) isoforms predominating in astrocytes are optimized to facilitate very large increases in lactate flux as lactate concentration increases within (1-3 mM) and above (>3 mM) the normal range. In sharp contrast, the major neuronal MCT serves as a barrier to increased transmembrane transport as lactate rises above 1 mM, restricting both entry and efflux. Lactate can serve as fuel during recovery from ischemia but direct evidence that lactate is oxidized by neurons (vs. astrocytes) to maintain synaptic function is lacking. Astrocytes have critical roles in regulation of ionic homeostasis and control of extracellular glutamate levels, and spreading depression associated with ischemia places high demands on energy supplies in astrocytes and contributes to metabolic exhaustion and demise. Disruption of Ca2+ homeostasis, generation of oxygen free radicals and nitric oxide, and mitochondrial depolarization contribute to astrocyte death during and after a metabolic insult. Novel pharmaceutical agents targeted to astrocytes and hyperoxic therapy that restores penumbral oxygen level during energy failure might improve postischemic outcome.
Collapse
Affiliation(s)
- Gerald A Dienel
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Leif Hertz
- College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
129
|
Abstract
Aquaporin 9 (AQP9) is a member of the aquaporin channel family involved in water flux through plasma membranes and exhibits the distinct feature of being also permeable to monocarboxylates, such as lactate, and various solutes, including glycerol, carbamides, purines, pyrimidines, and urea. AQP9 is constitutively expressed at high levels in the liver. In the brain under physiological conditions, AQP9 was first observed in tanycytes, and then in astrocytes. Only recently, its expression was also shown in neurons. Neurons expressing AQP9 are catecholaminergic and glucose sensitive. The expression of neuronal AQP9 can be negatively regulated by insulin and in diabetic animals an increase in AQP9 expression is observed in the catecholaminergic nuclei of the hindbrain, similar to the regulation of AQP9 by insulin in the liver. Furthermore, after transient brain ischemia, AQP9 expression is increased in astrocytes and its regulation may implicate the MAP-kinase pathways stimulated in such pathological conditions. Despite these new data, the exact role of AQP9 in the brain is still unclear. However, we may hypothesize that AQP9 is implicated in brain energy metabolism, as a neutral solute channel. AQP9 could facilitate the diffusion of lactate from the astrocyte to the neuron. In glucose sensitive neurons, diffusion of lactate and glycerol could stimulate these neurons in a similar manner to glucose and could regulate the energy balance. In pathological conditions, induction of AQP9 in astrocytes could participate in the clearance of excess lactate in the extracellular space. These hypotheses concerning the function of brain AQP9 are still speculative and open new areas of investigation.
Collapse
Affiliation(s)
- J Badaut
- Department of Neurosurgery, CHUV BH19-208, 1011 Lausanne, Switzerland.
| | | |
Collapse
|
130
|
Zhang F, Vannucci SJ, Philp NJ, Simpson IA. Monocarboxylate transporter expression in the spontaneous hypertensive rat: effect of stroke. J Neurosci Res 2005; 79:139-45. [PMID: 15578721 DOI: 10.1002/jnr.20312] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The expression of the monocarboxylate transporters (MCT) 1, 2, and 4 have been studied in the brains of spontaneous hypertensive (SH) rats after an ischemic insult induced by a permanent occlusion of the left middle cerebral artery (MCAO). Profound temporal changes in MCT1 expression were observed in various regions of the ipsilateral hemisphere over the period of 1 hr to 5 days after MCAO. Initially, a very rapid and transient increase in MCT1 mRNA was observed in neurons in the second layer of the cortex and in the piriform cortex at 1-3 hr. A slower but sustained increase in MCT1 mRNA expression was observed in astrocytes in the peri-infarct region beginning at 6 hr after MCAO and persisting over a period of 120 hr coinciding with their activation, migration, and involvement in scar formation. An increase in MCT1 expression in endothelial cells was seen over the same period. These increases in MCT1 expression in astrocytes and endothelial cells were accompanied by a corresponding increase in MCT1 protein. Finally at 120 hr post-MCAO, increases in MCT1, MCT2, and MCT4 expression were observed in cells within the infarct and bordering the scar, the identity of which remains to be determined. Consistent with cell death, the levels of MCT1, MCT2, and MCT4 mRNA decreased with cell death within the infarcted area but unlike MCT1, no increases in either MCT2 or 4 were observed within the remaining ipsilateral hemisphere. These studies reveal that the pattern of regulation of MCT1 expression after MCAO is similar to that observed previously for glucose transporter 1 (GLUT1) and suggest that the regulation of MCT1 mRNA expression is mediated by the hypoxia-inducible transcription factor HIF1.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | | | | | | |
Collapse
|
131
|
Bliss TM, Ip M, Cheng E, Minami M, Pellerin L, Magistretti P, Sapolsky RM. Dual-gene, dual-cell type therapy against an excitotoxic insult by bolstering neuroenergetics. J Neurosci 2005; 24:6202-8. [PMID: 15240812 PMCID: PMC6729663 DOI: 10.1523/jneurosci.0805-04.2004] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Increasing evidence suggests that glutamate activates the generation of lactate from glucose in astrocytes; this lactate is shuttled to neurons that use it as a preferential energy source. We explore this multicellular "lactate shuttle" with a novel dual-cell, dual-gene therapy approach and determine the neuroprotective potential of enhancing this shuttle. Viral vector-driven overexpression of a glucose transporter in glia enhanced glucose uptake, lactate efflux, and the glial capacity to protect neurons from excitotoxicity. In parallel, overexpression of a lactate transporter in neurons enhanced lactate uptake and neuronal resistance to excitotoxicity. Finally, overexpression of both transgenes in the respective cell types provided more protection than either therapy alone, demonstrating that a dual-cell, dual-gene therapy approach gives greater neuroprotection than the conventional single-cell, single-gene strategy.
Collapse
Affiliation(s)
- Tonya M Bliss
- Department of Biological Sciences, Stanford University, Stanford, California 94305-5020, USA
| | | | | | | | | | | | | |
Collapse
|
132
|
|
133
|
Serres S, Bezancon E, Franconi JM, Merle M. Ex Vivo Analysis of Lactate and Glucose Metabolism in the Rat Brain under Different States of Depressed Activity. J Biol Chem 2004; 279:47881-9. [PMID: 15361523 DOI: 10.1074/jbc.m409429200] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Brain metabolism of glucose and lactate was analyzed by ex vivo NMR spectroscopy in rats presenting different cerebral activities induced after the administration of pentobarbital, alpha-chloralose, or morphine. The animals were infused with a solution of either [1-(13)C]glucose plus lactate or glucose plus [3-(13)C]lactate for 20 min. Brain metabolite contents and enrichments were determined from analyses of brain tissue perchloric acid extracts according to their post-mortem evolution kinetics. When amino acid enrichments were compared, both the brain metabolic activity and the contribution of blood glucose relative to that of blood lactate to brain metabolism were linked with cerebral activity. The data also indicated the production in the brain of lactate from glycolysis in a compartment other than the neurons, presumably the astrocytes, and its subsequent oxidative metabolism in neurons. Therefore, a brain electrical activity-dependent increase in the relative contribution of blood glucose to brain metabolism occurred via the increase in the metabolism of lactate generated from brain glycolysis at the expense of that of blood lactate. This result strengthens the hypothesis that brain lactate is involved in the coupling between neuronal activation and metabolism.
Collapse
Affiliation(s)
- Sébastien Serres
- Unité de Résonance Magnétique des Systèmes Biologiques, Unité Mixte de Recherche 5536, CNRS-Université Victor Segalen, 146 Rue Léo-Saignat, 33076 Bordeaux, France
| | | | | | | |
Collapse
|
134
|
Reinert M, Schaller B, Widmer HR, Seiler R, Bullock R. Influence of oxygen therapy on glucose-lactate metabolism after diffuse brain injury. J Neurosurg 2004; 101:323-9. [PMID: 15309926 DOI: 10.3171/jns.2004.101.2.0323] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Severe traumatic brain injury (TBI) imposes a huge metabolic load on brain tissue, which can be summarized initially as a state of hypermetabolism and hyperglycolysis. In experiments O2 consumption has been shown to increase early after trauma, especially in the presence of high lactate levels and forced O2 availability. In recent clinical studies the effect of increasing O2 availability on brain metabolism has been analyzed. By their nature, however, clinical trauma models suffer from a heterogeneous injury distribution. The aim of this study was to analyze, in a standardized diffuse brain injury model, the effect of increasing the fraction of inspired O2 on brain glucose and lactate levels, and to compare this effect with the metabolism of the noninjured sham-operated brain. METHODS A diffuse severe TBI model developed by Foda and Maramarou, et al., in which a 420-g weight is dropped from a height of 2 m was used in this study. Forty-one male Wistar rats each weighing approximately 300 g were included. Anesthesized rats were monitored by placing a femoral arterial line for blood pressure and blood was drawn for a blood gas analysis. Two time periods were defined: Period A was defined as preinjury and Period B as postinjury. During Period B two levels of fraction of inspired oxygen (FiO2) were studied: air (FiO2 0.21) and oxygen (FiO2 1). Four groups were studied including sham-operated animals: air-air-sham (AAS); air-O2-sham (AOS); air-air-trauma (AAT); and air-O2-trauma (AOT). In six rats the effect of increasing the FiO2 on serum glucose and lactate was analyzed. During Period B lactate values in the brain determined using microdialysis were significantly lower (p < 0.05) in the AOT group than in the AAT group and glucose values in the brain determined using microdialysis were significantly higher (p < 0.04). No differences were demonstrated in the other groups. Increasing the FiO2 had no significant effect on the serum levels of glucose and lactate. CONCLUSIONS Increasing the FiO2 influences dialysate glucose and lactate levels in injured brain tissue. Using an FiO2 of 1 influences brain metabolism in such a way that lactate is significantly reduced and glucose significantly increased. No changes in dialysate glucose and lactate values were found in the noninjured brain.
Collapse
Affiliation(s)
- Michael Reinert
- Department of Neurological Surgery, University of Bern, Inselspital Bern, Switzerland.
| | | | | | | | | |
Collapse
|
135
|
Abstract
The mammalian brain contains glycogen, which is located predominantly in astrocytes, but its function is unclear. A principal role for brain glycogen as an energy reserve, analogous to its role in the periphery, had been universally dismissed based on its relatively low concentration, an assumption apparently reinforced by the limited duration that the brain can function in the absence of glucose. However, during insulin-induced hypoglycaemia, where brain glucose availability is limited, glycogen content falls first in areas with the highest metabolic rate, suggesting that glycogen provides fuel to support brain function during pathological hypoglycaemia. General anaesthesia results in elevated brain glycogen suggesting quiescent neurones allow glycogen accumulation, and as long ago as the 1950s it was shown that brain glycogen accumulates during sleep, is mobilized upon waking, and that sleep deprivation results in region-specific decreases in brain glycogen, implying a supportive functional role for brain glycogen in the conscious, awake brain. Interest in brain glycogen has recently been re-awakened by the first continuous in vivo measurements using NMR spectroscopy, by the general acceptance of metabolic coupling between glia and neurones involving intercellular transfer of energy substrate, and by studies supporting a prominent physiological role for brain glycogen as a provider of supplemental energy substrate during periods of increased tissue energy demand, when ambient normoglycaemic glucose is unable to meet immediate energy requirements.
Collapse
Affiliation(s)
- Angus M Brown
- Department of Neurology, University of Washington School of Medicine, Seattle, Washington, USA.
| |
Collapse
|
136
|
Pellerin L, Magistretti PJ. Neuroenergetics: calling upon astrocytes to satisfy hungry neurons. Neuroscientist 2004; 10:53-62. [PMID: 14987448 DOI: 10.1177/1073858403260159] [Citation(s) in RCA: 193] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Classical neuroenergetics states that glucose is the exclusive energy substrate of brain cells and its full oxidation provides all the necessary energy to support brain function. Recent data have revealed a more intricate picture in which astrocytes play a key role in supplying lactate as an additional energy substrate in register with glutamatergic activity.
Collapse
Affiliation(s)
- Luc Pellerin
- Institut de Physiologie, Université de Lausanne, Switzerland.
| | | |
Collapse
|
137
|
Yudkoff M, Daikhin Y, Nissim I, Lazarow A, Nissim I. Ketogenic diet, brain glutamate metabolism and seizure control. Prostaglandins Leukot Essent Fatty Acids 2004; 70:277-85. [PMID: 14769486 DOI: 10.1016/j.plefa.2003.07.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2003] [Accepted: 07/01/2003] [Indexed: 11/23/2022]
Abstract
We do not know the mode of action of the ketogenic diet in controlling epilepsy. One possibility is that the diet alters brain handling of glutamate, the major excitatory neurotransmitter and a probable factor in evoking and perpetuating a convulsion. We have found that brain metabolism of ketone bodies can furnish as much as 30% of glutamate and glutamine carbon. Ketone body metabolism also provides acetyl-CoA to the citrate synthetase reaction, in the process consuming oxaloacetate and thereby diminishing the transamination of glutamate to aspartate, a pathway in which oxaloacetate is a reactant. Relatively more glutamate then is available to the glutamate decarboxylase reaction, which increases brain [GABA]. Ketosis also increases brain [GABA] by increasing brain metabolism of acetate, which glia convert to glutamine. GABA-ergic neurons readily take up the latter amino acid and use it as a precursor to GABA. Ketosis also may be associated with altered amino acid transport at the blood-brain barrier. Specifically, ketosis may favor the release from brain of glutamine, which transporters at the blood-brain barrier exchange for blood leucine. Since brain glutamine is formed in astrocytes from glutamate, the overall effect will be to favor the release of glutamate from the nervous system.
Collapse
Affiliation(s)
- Marc Yudkoff
- Department of Pediatrics, University of Pennsylvania School of Medicine, Children's Hospital of Philadelphia, 34th Street and Civic Center Boulevard, Philadelphia, PA 19104, USA.
| | | | | | | | | |
Collapse
|
138
|
Hosoi R, Okada M, Hatazawa J, Gee A, Inoue O. Effect of astrocytic energy metabolism depressant on 14C-acetate uptake in intact rat brain. J Cereb Blood Flow Metab 2004; 24:188-90. [PMID: 14747745 DOI: 10.1097/01.wcb.0000098606.42140.02] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Fluorocitrate, a selective astrocytic toxin, was microinjected into the right striatum of rat brain, and the regional distribution of 14C-acetate was measured using autoradiography. A significant reduction (more than 80%) in 14C-acetate uptake over a 5-minute period was observed in the right striatum, compared with that in the left striatum (saline infused), 4 hours after fluorocitrate (1 nmol/microL) infusion. This effect was transient, and 14C-acetate uptake had almost returned to normal at 24 hours after the fluorocitrate infusion. In contrast, the regional blood flow in the striatum, as determined using 14C-iodoamphetamine, was significantly increased by the fluorocitrate infusion. The present observations indicate that 14C-acetate uptake might be a useful characteristic for examining astrocytic energy metabolism in the intact brain.
Collapse
Affiliation(s)
- Rie Hosoi
- School of Allied Health Sciences, Faculty of Medicine, Osaka University, Suita, Osaka, Japan
| | | | | | | | | |
Collapse
|
139
|
Halestrap AP, Meredith D. The SLC16 gene family-from monocarboxylate transporters (MCTs) to aromatic amino acid transporters and beyond. Pflugers Arch 2004; 447:619-28. [PMID: 12739169 DOI: 10.1007/s00424-003-1067-2] [Citation(s) in RCA: 756] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2003] [Accepted: 03/27/2003] [Indexed: 11/30/2022]
Abstract
The monocarboxylate cotransporter (MCT) family now comprises 14 members, of which only the first four (MCT1-MCT4) have been demonstrated experimentally to catalyse the proton-linked transport of metabolically important monocarboxylates such as lactate, pyruvate and ketone bodies. SLC16A10 (T-type amino-acid transporter-1, TAT1) is an aromatic amino acid transporter whilst the other members await characterization. MCTs have 12 transmembrane domains (TMDs) with intracellular N- and C-termini and a large intracellular loop between TMDs 6 and 7. MCT1 and MCT4 require a monotopic ancillary protein, CD147, for expression of functional protein at the plasma membrane. Lactic acid transport across the plasma membrane is fundamental for the metabolism of and pH regulation of all cells, removing lactic acid produced by glycolysis and allowing uptake by those cells utilizing it for gluconeogenesis (liver and kidney) or as a respiratory fuel (heart and red muscle). The properties of the different MCT isoforms and their tissue distribution and regulation reflect these roles.
Collapse
Affiliation(s)
- Andrew P Halestrap
- Department of Biochemistry, University of Bristol, BS8 1TD, Bristol, UK.
| | | |
Collapse
|
140
|
Fayol L, Baud O, Monier A, Pellerin L, Magistretti P, Evrard P, Verney C. Immunocytochemical expression of monocarboxylate transporters in the human visual cortex at midgestation. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2004; 148:69-76. [PMID: 14757520 DOI: 10.1016/j.devbrainres.2003.10.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Lactate and the other monocarboxylates are a major energy source for the developing brain. We investigated the immunocytochemical expression of two monocarboxylate transporters, MCT1 and MCT2, in the human visual cortex between 13 and 26 post-ovulatory weeks. We used immunoperoxidase and immunofluorescence techniques to determine whether these transporters co-localized with markers for blood vessels (CD34), neurons (microtubule-associated protein 2 [MAP2], SMI 311), radial glia (vimentin), or astrocytes (glial fibrillary acidic protein [GFAP], S100beta protein). MCT1 immunoreactivity was visible in blood vessel walls as early as the 13th week of gestation mainly in the cortical plate and subplate. At this stage, less than 10% of vessels in the ventricular layer expressed MCT1, whereas all blood vessels walls showed this immunoreactivity at the 26th gestational week. Starting at the 19th week of gestation, sparse MCT1 positive cell bodies were detected, some of them co-localized with MAP2 immunoreactivity. MCT2 immunoreactivity was noted in astrocytic cell bodies from week 19 and spread subsequently to the astrocyte end-feet in contact with blood vessels. MCTs immunoreactivities were most marked in the subplate and deep cortical plate, where the most differentiated neurons were located. Our findings suggest that monocarboxylate trafficking between vessels (MCT1), astrocytes (MCT2) and some postmitotic neurons (MCT1) could develop gradually toward 20 gestational weeks (g.w.). These data suggest that lactate or other monocarboxylates could represent a significant energy source for the human visual cortex at this early stage.
Collapse
Affiliation(s)
- Laurence Fayol
- INSERM E9935, Hôpital Robert Debré, 48, Bd Sérurier 75019, Paris, France
| | | | | | | | | | | | | |
Collapse
|
141
|
Pellerin L, Bergersen LH, Halestrap AP, Pierre K. Cellular and subcellular distribution of monocarboxylate transporters in cultured brain cells and in the adult brain. J Neurosci Res 2004; 79:55-64. [PMID: 15573400 DOI: 10.1002/jnr.20307] [Citation(s) in RCA: 193] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Monocarboxylate transporters (MCTs) are involved in the uptake and release of lactate, pyruvate, and ketone bodies. Studies of their distribution at both the mRNA and protein levels have highlighted the specific expression of MCT1, MCT2, and more recently MCT4 in the central nervous system. MCT1 was found strongly expressed by cortical astrocytes both in vitro and in vivo. It was also found at high levels on blood vessels, ependymocytes, and glia limitans. A subset of neurons in vitro exhibited a weak but significant MCT1 expression. In contrast, it was determined that MCT2 represents the predominant neuronal MCT on cultured neurons as well as on neurons throughout the brain parenchyma. At the subcellular level, part of MCT2 is located in postsynaptic densities. Specific populations of astrocytes in the white matter also exhibited MCT2 expression in the rat, but not in the mouse brain. MCT4 was found exclusively in astrocytes in several areas including the cortex, the hippocampus, and the cerebellum. MCT2 expression increased in cultured neurons with days in vitro commensurate with increased synapse formation. Moreover, a significant increase in MCT2 expression was observed in cultured neurons exposed to noradrenaline, an effect involving a regulation at the translational level. The description of MCTs on different cell types in the central nervous system together with clear evidence for regulation of their expression further emphasize the important role that monocarboxylates, and particularly lactate, might play in brain energy metabolism not only during development but also in the adult.
Collapse
Affiliation(s)
- Luc Pellerin
- Department of Physiology, University of Lausanne, 7 rue du Bugnon, 1005 Lausanne, Switzerland.
| | | | | | | |
Collapse
|
142
|
Hertz L, Dienel GA. Lactate transport and transporters: General principles and functional roles in brain cells. J Neurosci Res 2004; 79:11-8. [PMID: 15586354 DOI: 10.1002/jnr.20294] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Lactate is transported across cell membranes by diffusional, saturable cotransport with protons, mediated by monocarboxylate transporters (MCTs). This transport is bidirectional and in the absence of a transcellular H(+) gradient, it can increase the intracellular concentration of lactate up to but not beyond the extracellular level (or vice versa). If extra- and intracellular pH differ, however, the equilibrium level is determined by the gradients of both lactate anions and protons. Rates of lactate uptake are determined most often by measuring uptake of labeled lactate, e.g., [U-14C]lactate. In the case of lactate and other compounds that are metabolized, errors are introduced easily because continuing inwardly directed diffusional net transport of label can be achieved by intracellular metabolism, reducing the intracellular level of the nonmetabolized lactate and thus maintaining a concentration gradient between extra- and intracellular concentrations of the nonmetabolized compound (metabolism-driven uptake). For measurement of facilitated diffusion kinetics, it is essential that the period during which the uptake is measured is short enough that little or no metabolism-driven uptake contributes to the measured uptake (or that first-order regression analysis is carried out to obtain initial uptake rates from nonlinear traces). To achieve initial uptake rates, incubation periods well below 1 min are generally required. Lactate uptake is fast in astrocytes, which express powerful, low-affinity MCTs, i.e., MCT1 and MCT4. Due to the low affinity of these transporters, they respond to increased lactate gradients with enhanced transporter activity. The predominant MCT in neurons is the high-affinity MCT2, which can only increase its activity to a limited extent in the face of an increased lactate gradient. This is reflected by a high-affinity lactate uptake, although most investigators also have demonstrated a component of lactate uptake with lower affinity. In both neurons and astrocytes, however, facilitated diffusion is fast enough that under most conditions lactate fluxes will be determined mainly by the rate of metabolism-driven uptake, and MCT-mediated transport only will be rate-limiting after establishment of large transmembrane gradients.
Collapse
Affiliation(s)
- Leif Hertz
- College of Basic Medical Sciences, China Medical University, Shenyang, P.R. China
| | | |
Collapse
|
143
|
Mengual R, El Abida K, Mouaffak N, Rieu M, Beaudry M. Pyruvate shuttle in muscle cells: high-affinity pyruvate transport sites insensitive to trans-lactate efflux. Am J Physiol Endocrinol Metab 2003; 285:E1196-204. [PMID: 12915395 DOI: 10.1152/ajpendo.00034.2003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The specificity of the transport mechanisms for pyruvate and lactate and their sensitivity to inhibitors were studied in L6 skeletal muscle cells. Trans- and cis-lactate effects on pyruvate transport kinetic parameters were examined. Pyruvate and lactate were transported by a multisite carrier system, i.e., by two families of sites, one with low affinity and high capacity (type I sites) and the other with high affinity and low capacity (type II). The multisite character of transport kinetics was not modified by either hydroxycinnamic acid (CIN) or p-chloromercuribenzylsulfonic acid (PCMBS), which exert different types of inhibition. The transport efficiency (TE) ratios of maximal velocity to the trans-activation dissociation constant (Kt) showed that lactate and pyruvate were preferentially transported by types I and II sites, respectively. The cis-lactate effect was observed with high Ki values for both sites. The trans-lactate effect on pyruvate transport occurred only on type I sites and exhibited an asymmetric interaction pattern (Kt of inward lactate > Kt of outward lactate). The inability of lactate to trans-stimulate type II sites suggests that intracellular lactate cannot recruit these sites. The high-affinity type II sites act as a specific pyruvate shuttle and constitute an essential relay for the intracellular lactate shuttle.
Collapse
Affiliation(s)
- Raymond Mengual
- Laboratoire de Physiologie des Adaptations, Unité de Formation et de Recherche Cochin Port Royal, Université René Descartes, 75014 Paris, France.
| | | | | | | | | |
Collapse
|
144
|
Cater HL, Chandratheva A, Benham CD, Morrison B, Sundstrom LE. Lactate and glucose as energy substrates during, and after, oxygen deprivation in rat hippocampal acute and cultured slices. J Neurochem 2003; 87:1381-90. [PMID: 14713294 DOI: 10.1046/j.1471-4159.2003.02100.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The effects of raised brain lactate levels on neuronal survival following hypoxia or ischemia is still a source of controversy among basic and clinical scientists. We have sought to address this controversy by studying the effects of glucose and lactate on neuronal survival in acute and cultured hippocampal slices. Following a 1-h hypoxic episode, neuronal survival in cultured hippocampal slices was significantly higher if glucose was present in the medium compared with lactate. However, when the energy substrate during the hypoxic period was glucose and then switched to lactate during the normoxic recovery period, the level of cell damage in the CA1 region of organotypic cultures was significantly improved from 64.3 +/- 2.1 to 74.6 +/- 2.1% compared with cultures receiving glucose during and after hypoxia. Extracellular field potentials recorded from the CA1 region of acute slices were abolished during oxygen deprivation for 20 min, but recovered almost fully to baseline levels with either glucose (82.6 +/- 10.0%) or lactate present in the reperfusion medium (108.1 +/- 8.3%). These results suggest that lactate alone cannot support neuronal survival during oxygen deprivation, but a combination of glucose followed by lactate provides for better neuroprotection than either substrate alone.
Collapse
Affiliation(s)
- Heather L Cater
- Clinical Neurosciences, University of Southampton, Southampton Neurology Centre of Excellence for Drug Discovery, GlaxoSmithKline, Harlow, UK.
| | | | | | | | | |
Collapse
|
145
|
Chih CP, Roberts EL. Energy substrates for neurons during neural activity: a critical review of the astrocyte-neuron lactate shuttle hypothesis. J Cereb Blood Flow Metab 2003; 23:1263-81. [PMID: 14600433 DOI: 10.1097/01.wcb.0000081369.51727.6f] [Citation(s) in RCA: 236] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Glucose had long been thought to fuel oxidative metabolism in active neurons until the recently proposed astrocyte-neuron lactate shuttle hypothesis (ANLSH) challenged this view. According to the ANLSH, activity-induced uptake of glucose takes place predominantly in astrocytes, which metabolize glucose anaerobically. Lactate produced from anaerobic glycolysis in astrocytes is then released from astrocytes and provides the primary metabolic fuel for neurons. The conventional hypothesis asserts that glucose is the primary substrate for both neurons and astrocytes during neural activity and that lactate produced during activity is removed mainly after neural activity. The conventional hypothesis does not assign any particular fraction of glucose metabolism to the aerobic or anaerobic pathways. In this review, the authors discuss the theoretical background and critically review the experimental evidence regarding these two hypotheses. The authors conclude that the experimental evidence for the ANLSH is weak, and that existing evidence and theoretical considerations support the conventional hypothesis.
Collapse
Affiliation(s)
- Ching-Ping Chih
- Geriatric Research, Education, and Clinical Center, and Research Office, Miami VA Medical Center, Miami, Florida, USA
| | | |
Collapse
|
146
|
Abstract
Normal development of both human and rat brain is associated with a switch in metabolic fuel from a combination of glucose and ketone bodies in the immature brain to a nearly total reliance on glucose in the adult. The delivery of glucose, lactate, and ketone bodies from the blood to the brain requires specific transporter proteins, glucose and monocarboxylic acid transporter proteins (GLUTs and MCTs), respectively. Developmental expression of the GLUTs in rat brain, i.e., 55-kDa GLUT1 in the blood-brain barrier (BBB), 45-kDa GLUT1 and GLUT3 in vascular-free brain, corresponds to maturational increases in cerebral glucose uptake and utilization. It has been suggested that MCT expression peaks during suckling and sharply declines thereafter, although a comparable detailed study has not been done. This study investigated the temporal and regional expression of MCT1 and MCT2 mRNA and protein in the BBB and the nonvascular brain during postnatal development in the rat. The results confirmed maximal MCT1 mRNA and protein expression in the BBB during suckling and a decline with maturation, coincident with the switch to glucose as the predominant cerebral fuel. However, nonvascular MCT1 and MCT2 levels do not reflect changes in cerebral energy metabolism, suggesting a more complex regulation. Although MCT1 assumes a predominantly glial expression in postweanling brain, the concentration remains fairly constant, as does that of MCT2 in neurons. The maintenance of nonvascular MCT levels in the adult brain implies a major role for these proteins, in concert with the GLUTs in both neurons and astrocytes, to transfer glycolytic intermediates during cerebral energy metabolism.
Collapse
Affiliation(s)
- Susan J Vannucci
- Department of Pediatrics, University, New York, New York 10032, USA.
| | | |
Collapse
|
147
|
Bouzier-Sore AK, Voisin P, Canioni P, Magistretti PJ, Pellerin L. Lactate is a preferential oxidative energy substrate over glucose for neurons in culture. J Cereb Blood Flow Metab 2003; 23:1298-306. [PMID: 14600437 DOI: 10.1097/01.wcb.0000091761.61714.25] [Citation(s) in RCA: 235] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The authors investigated concomitant lactate and glucose metabolism in primary neuronal cultures using 13C- and 1H-NMR spectroscopy. Neurons were incubated in a medium containing either [1-13C]glucose and different unlabeled lactate concentrations, or unlabeled glucose and different [3-13C]lactate concentrations. Overall, 13C-NMR spectra of cellular extracts showed that more 13C was incorporated into glutamate when lactate was the enriched substrate. Glutamate 13C-enrichment was also found to be much higher in lactate-labeled than in glucose-labeled conditions. When glucose and lactate concentrations were identical (5.5 mmol/L), relative contributions of glucose and lactate to neuronal oxidative metabolism amounted to 21% and 79%, respectively. Results clearly indicate that when neurons are in the presence of both glucose and lactate, they preferentially use lactate as their main oxidative substrate.
Collapse
Affiliation(s)
- Anne-Karine Bouzier-Sore
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536 CNRS/Université, Bordeaux, France.
| | | | | | | | | |
Collapse
|
148
|
Affiliation(s)
- Luc Pellerin
- Institut de Physiologie, Université de Lausanne, Switzerland.
| | | |
Collapse
|
149
|
Baud O, Fayol L, Gressens P, Pellerin L, Magistretti P, Evrard P, Verney C. Perinatal and early postnatal changes in the expression of monocarboxylate transporters MCT1 and MCT2 in the rat forebrain. J Comp Neurol 2003; 465:445-54. [PMID: 12966567 DOI: 10.1002/cne.10853] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In addition to glucose, monocarboxylates including lactate represent a major source of energy for the brain, especially during development. We studied the immunocytochemical expression of the monocarboxylate transporters MCT1 and MCT2 in the rat brain between embryonic day (E) 16 and postnatal day (P) 14. At E16-18, MCT1-like immunoreactivity was found throughout the cortical anlage, being particularly marked medially in the hippocampal anlage next to the ventricle. In a complementary pattern, MCT2-like immunoreactivity was expressed along the medial and ventral border of the ventricle in the medial septum and habenula before birth. The hypothalamic area exhibited MCT2 and MCT1 positive areas from E18 on. These transient labelings revealed four main sites of monocarboxylate and/or glucose exchange: the brain parenchyma, the epithelial cells, the ependymocytes, and the glia limitans. During the first postnatal week, MCT1 immunoreactivity extended massively to the vessel walls and moderately to the developing astrocytes in the cortex. In contrast, MCT2 immunoreactivity was faint in blood vessels but massive in developing astrocytes from P3 to P7. Neither MCT2 nor MCT1 colocalized with neuronal, microglial, or oligodendrocytic markers during the first postnatal week. At P14, a part of the scattered punctate MCT2 staining could be associated with astrocytes and postsynaptic dendritic labeling. The transient pattern of expression of MCTs throughout the perinatal period suggests a potential relationship with the maturation of the blood-brain barrier.
Collapse
Affiliation(s)
- Olivier Baud
- Institut National de la Santé et de la Recherche Médicale E9935, Laboratoire de Neurologie du Développement, Hôpital Robert Debré, 48 boulevard Sérurier, 75019 Paris, France
| | | | | | | | | | | | | |
Collapse
|
150
|
Abstract
Lactate has been considered for a long time as a metabolic waste and/or a sign of hypoxia in the central nervous system. Nevertheless, clear evidence that lactate can constitute an adequate energy substrate for brain tissue has been provided as early as in the 1950s with the pioneering work of McIlwain in brain slices. Over the years, several studies using different approaches have confirmed that lactate is efficiently oxidized by brain cells in vitro. Moreover, lactate has been shown under certain circumstances to have a neuroprotective effect and support neuronal activity. Similar confirmation of lactate utilization in vivo as well as putative neuroprotection in various excitotoxic models has been provided. Lactate was even shown to restore cognitive performance upon an hypoglycemic episode in humans. More recently, it was proposed that lactate could be produced by astrocytes and released in the extracellular space to form a pool readily available for neurons in case of high energy demands. Several elements support the concept of a lactate shuttle between astrocytes and neurons in the central nervous system. Among them, the description of specific monocarboxylate transporters found on both astrocytes and neurons is an important observation consistent with this concept. Interestingly, lactate shuttles between different cell types within the same organ have been described outside the central nervous system, notably in muscle and testis. Thus, lactate is emerging as a valuable intercellular exchange molecule in different systems including the brain where it might be an essential element of neuron-glia metabolic interactions.
Collapse
Affiliation(s)
- Luc Pellerin
- Institut de Physiologie, 7 rue du Bugnon, 1005 Lausanne, Switzerland.
| |
Collapse
|