scispace - formally typeset
Search or ask a question

Showing papers on "Monocarboxylate transporter published in 2004"


Journal ArticleDOI
TL;DR: The monocarboxylate cotransporter 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 monoccarboxylates such as lactate, pyruvate and ketone bodies.
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.

964 citations


Journal ArticleDOI
TL;DR: For the first time, mutations in the monocarboxylate transporter 8 (MCT8) gene, located on the X chromosome, have been linked to a defect in cellular hormone transport as mentioned in this paper.
Abstract: Thyroid hormones are iodothyronines that control growth and development, as well as brain function and metabolism Although thyroid hormone deficiency can be caused by defects of hormone synthesis and action, it has not been linked to a defect in cellular hormone transport In fact, the physiological role of the several classes of membrane transporters remains unknown We now report, for the first time, mutations in the monocarboxylate transporter 8 (MCT8) gene, located on the X chromosome, that encodes a 613–amino acid protein with 12 predicted transmembrane domains The propositi of two unrelated families are males with abnormal relative concentrations of three circulating iodothyronines, as well as neurological abnormalities, including global developmental delay, central hypotonia, spastic quadriplegia, dystonic movements, rotary nystagmus, and impaired gaze and hearing Heterozygous females had a milder thyroid phenotype and no neurological defects These findings establish the physiological importance of MCT8 as a thyroid hormone transporter

616 citations


Journal ArticleDOI
TL;DR: The orphan cotransport protein expressed by the SLC5A8 gene, being expressed in colon and kidney, represents a novel means by which Na+, short‐chain fatty acids and other monocarboxylates are transported in these tissues.
Abstract: The orphan cotransport protein expressed by the SLC5A8 gene has been shown to play a role in controlling the growth of colon cancers, and the silencing of this gene is a common and early event in human colon neoplasia. We expressed this protein in Xenopus laevis oocytes and have found that it transports small monocarboxylic acids. The electrogenic activity of the cotransporter, which we have named SMCT (sodium monocarboxylate transporter), was dependent on external Na+ and was compatible with a 3 : 1 stoichiometry between Na+ and monocarboxylates. A portion of the SMCT-mediated current was also Cl− dependent, but Cl− was not cotransported. SMCT transports a variety of monocarboxylates (similar to unrelated monocarboxylate transport proteins) and most transported monocarboxylates demonstrated Km values near 100 μm, apart from acetate and d-lactate, for which the protein showed less affinity. SMCT was strongly inhibited by 1 mm probenecid or ibuprofen. In the absence of external substrate, a Na+-independent leak current was also observed to pass through SMCT. SMCT activity was strongly inhibited after prolonged exposure to high external concentrations of monocarboxylates. The transport of monocarboxylates in anionic form was confirmed by the observation of a concomitant alkalinization of the cytosol. SMCT, being expressed in colon and kidney, represents a novel means by which Na+, short-chain fatty acids and other monocarboxylates are transported in these tissues. The significance of a Na+–monocarboxylate transporter to colon cancer presumably stems from the transport of butyrate, which is well known for having anti-proliferative and apoptosis-inducing activity in colon epithelial cells.

159 citations


Journal ArticleDOI
TL;DR: X.13512 as discussed by the authors is a prodrug of gabapentin designed to be absorbed throughout the intestine by high-capacity nutrient transporters, and it has been shown to be a substrate for several high capacity absorption pathways.
Abstract: Gabapentin is thought to be absorbed from the intestine of humans and animals by a low-capacity solute transporter localized in the upper small intestine. Saturation of this transporter at doses used clinically leads to dose-dependent pharmacokinetics and high interpatient variability, potentially resulting in suboptimal drug exposure in some patients. XP13512 [(+/-)-1-([(alpha-isobutanoyloxyethoxy)carbonyl] aminomethyl)-1-cyclohexane acetic acid] is a novel prodrug of gabapentin designed to be absorbed throughout the intestine by high-capacity nutrient transporters. XP13512 was stable at physiological pH but rapidly converted to gabapentin in intestinal and liver tissue from rats, dogs, monkeys, and humans. XP13512 was not a substrate or inhibitor of major cytochrome P450 isoforms in transfected baculosomes or liver homogenates. The separated isomers of XP13512 showed similar cleavage in human tissues. The prodrug demonstrated active apical to basolateral transport across Caco-2 cell monolayers and pH-dependent passive permeability across artificial membranes. XP13512 inhibited uptake of (14)C-lactate by human embryonic kidney cells expressing monocarboxylate transporter type-1, and direct uptake of prodrug by these cells was confirmed using liquid chromatography-tandem mass spectrometry. XP13512 inhibited uptake of (3)H-biotin into Chinese hamster ovary cells overexpressing human sodium-dependent multivitamin transporter (SMVT). Specific transport by SMVT was confirmed by oocyte electrophysiology studies and direct uptake studies in human embryonic kidney cells after tetracycline-induced expression of SMVT. XP13512 is therefore a substrate for several high-capacity absorption pathways present throughout the intestine. Therefore, administration of the prodrug should result in improved gabapentin bioavailability, dose proportionality, and colonic absorption compared with administration of gabapentin.

145 citations


Journal ArticleDOI
TL;DR: The results suggest that the facilitation of MCT1 transport activity is mainly due to the increase in apparent buffer capacity contributed by the NBC, and thereby suppresses the build-up of intracellular H+ during the influx of lactate/H+, which would reduce M CT1 activity.

98 citations


Journal ArticleDOI
TL;DR: Comparison of results indicates that the most likely source of discrepancy in the mitochondrial localization of MCT1 is the cell fractionation procedure utilized.
Abstract: We sought to test the hypothesis that monocarboxylate transporter isoform 1 (MCT1) is the inner mitochondrial membrane lactate/pyruvate transporter, and, as such, contributes to functioning of the intracellular lactate shuttle. However, presence of a mammalian mitochondrially localized MCT1 (mMCT1) has been contested. We sought to confirm by Western blotting the mitochondrial localization of MCT1 in rat cardiac, soleus, and extensor digitorum longus muscles utilizing three different cell fractionation methods and three different antibodies. We performed Western blotting using antibodies to cell membrane glucose transporter isoform GLUT1, inner mitochondrial constituent cytochrome oxidase, the monocarboxylate transporter protein chaperone CD147, as well as custom and commercially available MCT1 antibodies. Western blots demonstrated similar results with each MCT1 antibody and two of three methods of fractionation. MCT1 was found in the mitochondria, as well as in the sarcolemmal membrane and whole muscle homogenates. Probing with GLUT1 and CD147 demonstrated that mitochondrial fractions were not contaminated with sarcolemmal remnants. Probing with cytochrome oxidase showed mitochondrial localization of MCT1. Comparison of these results to the findings of others indicates that the most likely source of discrepancy is the cell fractionation procedure utilized.

86 citations


Journal ArticleDOI
01 Jul 2004-Placenta
TL;DR: It is demonstrated that the opposing plasma membranes of human syncytiotrophoblast are polarized with respect to both MCT activity and expression.

56 citations


Journal ArticleDOI
TL;DR: It is concluded that hypoxic-ischemic brain injury is associated with transient compensatory changes targeted at protecting glucose delivery to fuel cellular energy metabolism, which then may delay the processes of apoptosis and cell necrosis.
Abstract: We examined the effect of hypoxic ischemia and hypoxia vs. normoxia on postnatal murine brain substrate transporter concentrations and function. We detected a transient increase in the neuronal brain glucose transporter isoform (GLUT-3) in response to hypoxic ischemia after 4 h of reoxygenation. This increase was associated with no change in GLUT-1 (blood-brain barrier/glial isoform), monocarboxylate transporter isoforms 1 and 2, synapsin I (neuronal marker), or Bax (proapoptotic protein) but with a modest increase in Bcl-2 (antiapoptotic mitochondrial protein) protein concentrations. At 24 h of reoxygenation, the increase in GLUT-3 disappeared but was associated with a decline in Bcl-2 protein concentrations and the Bcl2:Bax ratio, an increase in caspase-3 enzyme activity (apoptotic effector enzyme), and extensive DNA fragmentation, which persisted later in time (48 h) only in the hippocampus. Hypoxia alone in the absence of ischemia was associated with a transient but modest increase in GLUT-3 and synapsin I protein concentrations, which did not cause significant apoptosis and/or necrosis. Assessment of glucose transporter function by 2-deoxyglucose (2-DG) uptake using two distinct techniques, namely positron emission tomography (PET) and the modified Sokoloff method, revealed a discrepancy due to glucose uptake by extracranial Harderian glands that masked the accurate detection of intracranial brain glucose uptake by PET scanning. The modified Sokoloff method assessing 2-DG uptake revealed that the transient increase in GLUT-3 was critical in protecting against a decline in brain glucose uptake. We conclude that hypoxic-ischemic brain injury is associated with transient compensatory changes targeted at protecting glucose delivery to fuel cellular energy metabolism, which then may delay the processes of apoptosis and cell necrosis.

53 citations


Journal ArticleDOI
TL;DR: It appears that mild exercise training fails to increase MCT4 and that changes in MCT1 are complex, depending not only the accumulated exercise but also on the stage of training.
Abstract: We examined whether the quantity of exercise performed influences the expression of monocarboxylate transporter (MCT) 1 and MCT4 in mouse skeletal muscles (plantaris, tibialis anterior, soleus) and...

39 citations


Journal ArticleDOI
TL;DR: The results indicate that the electrogenic NBCe1 renders the cell membrane potential an effective regulator of intracellular H+ buffering and acid/base-coupled metabolite transport.

38 citations


Journal ArticleDOI
TL;DR: The transport and metabolism of lactate, another monocarboxylate, was examined in these two preparations, and the results were compared to those for acetate.
Abstract: Astrocytes transport the monocarboxylate acetate, but synaptosomes do not. The reason for this is unknown, because both preparations express monocarboxylate transporters (MCT). The transport and metabolism of lactate, another monocarboxylate, was examined in these two preparations, and the results were compared to those for acetate. Lactate transport is more rapid in astrocytes than in synaptosomes, but of lower affinity (Kms of 17 and 4 mM, respectively). Lactate (0.2 mM) is metabolized to CO2 more rapidly in synaptosomes than in astrocytes (rates of 0.37 and 0.07 nmol · mg protein−1 · min−1, respectively). The reason for this is unclear, but cellular differences in lactate dehydrogenase isotype expression may be involved. Acetate is metabolized to CO2 more rapidly in astrocytes than in synaptosomes (rates of 0.43 and 0.02 nmol · mg protein−1 · min−1, respectively). This is likely due to cellular differences in the expression of monocarboxylate transporter subtypes.

Journal ArticleDOI
TL;DR: It is suggested that EIH could modify lactate influx into erythrocyte and was not accompanied by an increase in RBC rigidity, as well as higher monocarboxylate transporter-1-mediated lactATE influx in the EIh subjects.
Abstract: This study investigated 1) red blood cells (RBC) rigidity and 2) lactate influxes into RBCs in endurance-trained athletes with and without exercise-induced hypoxemia (EIH). Nine EIH and six non-EIH subjects performed a submaximal steady-state exercise on a cyclo-ergometer at 60% of maximal aerobic power for 10 min, followed by 15 min at 85% of maximal aerobic power. At rest and at the end of exercise, arterialized blood was sampled for analysis of arterialized pressure in oxygen, and venous blood was drawn for analysis of plasma lactate concentrations and hemorheological parameters. Lactate influxes into RBCs were measured at three labeled [U-14C]lactate concentrations (1.6, 8.1, and 41 mM) on venous blood sampled at rest. The EIH subjects had higher maximal oxygen uptake than non-EIH (P < 0.05). Total lactate influx was significantly higher in RBCs from EIH compared with non-EIH subjects at 8.1 mM (1,498.1 +/- 87.8 vs. 1,035.9 +/- 114.8 nmol.ml(-1).min(-1); P < 0.05) and 41 mM (2,562.0 +/- 145.0 vs. 1,618.1 +/- 149.4 nmol.ml(-1).min(-1); P < 0.01). Monocarboxylate transporter-1-mediated lactate influx was also higher in EIH at 8.1 mM (P < 0.05) and 41 mM (P < 0.01). The drop in arterial oxygen partial pressure was negatively correlated with total lactate influx measured at 8.1 mM (r = -0.82, P < 0.05) and 41 mM (r = -0.84, P < 0.05) in the two groups together. Plasma lactate concentrations and hemorheological data were similar in the two groups at rest and at the end of exercise. The results showed higher monocarboxylate transporter-1-mediated lactate influx in the EIH subjects and suggested that EIH could modify lactate influx into erythrocyte. However, higher lactate influx in EIH subjects was not accompanied by an increase in RBC rigidity.

Journal ArticleDOI
TL;DR: The results demonstrate a new strategy for the production of properly folded and correctly targeted membrane proteins in a microbial expression system by co-expression of appropriate accessory proteins.
Abstract: We have developed a new heterologous expression system for monocarboxylate transporters. The system is based on a Saccharomyces cerevisiae pyk1 mae1 jen1 triple-deletion strain that is auxotrophic for pyruvate and deficient in monocarboxylate uptake. Growth of the yeast cells on ethanol medium supplemented with pyruvate or lactate was dependent on the expression of a suitable monocarboxylate transporter. We have used the system to characterize the functional significance of interactions between the rat MCT1 transporter and its ancillary protein CD147. CD147 was shown to improve trafficking of MCT1 to the plasma membrane and its uptake activity. Our results demonstrate a new strategy for the production of properly folded and correctly targeted membrane proteins in a microbial expression system by co-expression of appropriate accessory proteins.

Journal ArticleDOI
TL;DR: Results suggest that L-lactic acid efflux in L6 cells is mediated by MCT other than MCT1, and that an MCT participates in the uptake of L- lactic acid by L 6 cells.

Journal ArticleDOI
TL;DR: The results suggest that AmcA could be involved not only in monocarboxylate transport, but also in drug secretion, the first report suggesting a possible involvement of MCT transporters with drug resistance in eukaryotic cells.
Abstract: The Monocarboxylate Transporter (MCT) is a family of membrane proteins from the Major Facilitator Superfamily (MFS) of transporters. Here, we report the first identified Aspergillus nidulans MCT homologue, AmcA. The amcA gene was isolated by high copy number suppression of the hydroxyurea (HU) sensitivity of an A. nidulans camptothecin-sensitive mutant. Expression of amcA is increased when hyphae are grown in media containing acetate or pyruvate as single carbon source, and after exposure to several unrelated drugs. Our results suggest that AmcA could be involved not only in monocarboxylate transport, but also in drug secretion. To our knowledge, this is the first report suggesting a possible involvement of MCT transporters with drug resistance in eukaryotic cells.

Book ChapterDOI
01 Jan 2004
TL;DR: This chapter addresses the importance of the transcriptional regulation of intestinal nutrient transporter genes, and highlights its relevance to nutrition, health and disease.
Abstract: Nutrient transport across the apical plasma membrane of enterocytes is mediated by highly specialised membrane proteins, the majority of which are adaptively regulated by dietary substrates. The nutrient signals are either transmitted by the transporter itself, regulating the external nutrient access to the intracellular environment, or via distinct luminal membrane nutrient sensors. This chapter addresses the importance of the transcriptional regulation of intestinal nutrient transporter genes, and highlights its relevance to nutrition, health and disease. Examples are given of nutrient transporters from both the small and large intestine for which the underlying transcriptional mechanisms have been identified. These include the Na+/glucose cotransporter (SGLT1) and the bile salt transporter (ABST) expressed in the small intestine, and the monocarboxylate transporter (MCT1), residing in the large intestine. A better understanding of transcriptional regulation of intestinal nutrient transporters will undoubtedly have important clinical and nutritional implications.

Journal ArticleDOI
TL;DR: The existence of MCT1 is proved in aortic smooth muscle cells (HASMC) and pressure stress changed the localization of M CT1, which may transport lactate for energy metabolism in cells.
Abstract: Lactate is formed and utilized continuously under fully aerobic conditions. Lactate is oxidized actively at all times, especially during exercise. Family of monocarboxylate transport proteins (MCTs) that are differentially expressed in cells and tissues accomplishes the facilitated transport of lactate across membranes. Previously we reported that there is MCT1 in blood circulation. We also reported the pressure stress stimulated cell proliferation in aortic smooth muscle cells (HASMC). In this experiment we attempted to prove the existence of MCT1 in HASMC and to clarify the effect of pressure stress on MCT1 localization in HASMC. We determined succinate dehydrogenase (SDH) activity as a marker of energy metabolism in cells. SDH activity was increased by pressure stress. Lactate enhanced the SDH activity under pressure stress (160 mmHg for 3 h) as dose dependent manner. On the other hand, lactate excretion was suppressed by the addition of lactate. We could detect MCT1 in the cytosolic and the membrane fractions of HASMC. The pressure stress increased MCT1 in the membrane fraction in the presence of extracellular lactate. In summary, we proved the existence of MCT1 in HASMC. Pressure stress changed the localization of MCT1. The increased membranous MCT1 may transport lactate for energy metabolism in cells.