Abstract
Solute carrier (SLC) transporters represent 52 families of membrane transport proteins that function in endogenous compound homeostasis and xenobiotic disposition, and have been exploited in drug delivery and therapeutic targeting strategies. In particular, the SLC16 family that encodes for the 14 isoforms of the monocarboxylate transporter (MCT) family plays a significant role in the absorption, tissue distribution, and clearance of both endogenous and exogenous compounds. MCTs are required for the transport of essential cell nutrients and for cellular metabolic and pH regulation. Recent publications have indicated their novel roles in disease, and thus their potential as biomarkers and new therapeutic targets in disease are under investigation. More research into MCT isoform function, specificity, expression, and regulation will allow researchers to exploit the potential utility of MCTs in the clinic as therapeutic targets and prognostic factors of disease.
Monocarboxylate transporters (MCTs) are members of the solute carrier (SLC) family (SLC16) of proteins and are comprised of 14 isoforms. Previously identified, based on cDNA isolation and homology sequencing, MCTs are present in a wide variety of tissues and are involved in the regulation of fundamental cellular processes, such as glycolysis, fatty acid homeostasis, as well as other key metabolic pathways.1,2 Recent research on MCTs has focused on their role in the pathophysiology of disease and as therapeutic targets.
Of the 14 isoforms identified, proton-dependent MCTs 1–4 have been extensively studied due to their importance in transporting L-lactate, pyruvate, and short-chain fatty acids in a wide variety of tissues. Most importantly, many of these MCT isoforms are upregulated in tumor tissues, making them attractive targets and biomarkers for a wide variety of cancers.3 Through the targeted disruption of pH homeostasis and balance of mono-carboxylate compounds, such as lactate, which are essential for oxidative and glycolytic metabolism, MCTs 1–4 are gaining more interest in the field of clinical pharmacology. In addition, MCTs 5–14, many of which have yet to be “de-orphanized” due to their unknown endogenous function and substrate specificity, are of interest due to their differential tissue localization and potential to be manipulated in drug delivery or targeting strategies. Of note, more recently developed high throughput metabolomics analyses and genetically modified preclinical models have been utilized and allowing researchers to decipher the physiological function of a number of orphan MCTs.4–6
This state of the art topical review will focus on MCTs, highlighting recent publications that have provided insight into their potential roles in clinical pharmacology and therapeutics. Although members of the SLC5 family of transporters encoding for the sodium-dependent monocarboxylate transporters have also demonstrated a fundamental role in maintaining monocarboxylate homeostasis, for the purpose of this review, the focus will be solely on the SLC16 family of transporters.
Structure, function, substrate specificity, and regulation
As members of the major facilitator superfamily, the SLC16 family of MCTs contains shared, conserved structural attributes, and sequence motifs. According to hydropathy plots, all MCT isoforms are predicted to have 12 transmembrane (TM) α-helices with a large intracellular loop between TMs 6 and 7, as well as intracellular C- and N-termini.7,8 Specifically, regarding their protein sequences, MCTs share the greatest conservation within the TM regions with more variability in the cytosolic C- and N-termini, as well as the loop between TMs 6 and 7. In addition, two highly conserved motifs include sequences preceding TM1 and TM5, which have been suggested to play a role in the molecular dynamics and conformational changes of MCTs.9 Several predicted 3D structures of the MCTs have been proposed from homology-based modeling approaches using the previously characterized Escherichia coli glycerol-3-phosphate transporter crystal structure (1PW4).10,11 Studies using MCT-transfected Xenopus laevis oocytes have been used to characterize the substrate specificity and functionality of specific MCT isoforms in the absence of other MCTs or other anionic transporters with overlapping substrate specificity.
Table 13–5,12–51 summarizes the substrate specificity, tissue localization, and the potential clinical relevance for the MCT isoforms. It is important to note that the discrepancy between MCT isoform number and SLC16A nomenclature evolved from the order in which each cDNA sequence was determined and characterized.2 It is apparent that there is a similarity in substrate specificity for lactate for MCTs 1–4 and a high degree of variability among tissue expression levels of all MCT isoforms. In addition, many of the MCT isoforms have demonstrated not just the uptake, but also efflux of their endogenous substrates in order to maintain homeostasis and ion balance. In general, the substrate specificity of MCTs 1–4 has demonstrated affinity for common, endogenous short chain monocarboxylates, including, but not limited to, lactate, pyruvate, butyrate, γ-hydroxybutyrate, and ketone bodies (such as acetoacetate and β-hydroxybutyrate).2 Importantly, the specificity for lactate remains restricted to the endogenous stereoisomer L-lactate, in contrast to D-lactate, which is perhaps the single most important substrate in the entire MCT family. L-lactate is not only important for oxidative/glycolytic metabolism and pH regulation, but also acts as a signaling agent to promote angiogenesis and immunosuppression.52 Other MCT isoforms, such as MCT6, have demonstrated affinity for a small group of xenobiotics, such as bumetanide, nateglinide, and probenecid,53 whereas MCT7 has been characterized as a transporter of ketone bodies.54 MCT8 and MCT10 have been reported to transport thyroid hormones,7 and more recently MCT9 has been characterized as a carnitine efflux transporter55 and MCT12 as a creatine transporter.6
Table 1.
The SLC16A family: substrates, tissue expression, and potential clinical relevance
| Protein | Gene | Key substrates | Human mRNA tissue/cellular expression | Potential clinical relevance (ref.) |
|---|---|---|---|---|
| MCT1 | SLC16A1 | Lactate, pyruvate, ketone bodies | Ubiquitous (except β-cells of the endocrine pancreas) | Cancera, immunosuppression,a lactate transporter defect,b glucose regulationa (12–24) |
| MCT2 | SLC16A7 | Pyruvate, lactate, ketone bodies | High expression in testis, moderate to low in spleen, heart, kidney, pancreas, skeletal muscle, brain, leucocytes | Cancer,a prostate cancer biomarker,a neurodegenerative disease,b male infertilityb (25–30) |
| MCT3 | SLC16A8 | Lactate | Retinal pigment epithelium, choroid plexus | Atherosclerosis,c altered visual function,b wound healingc (31–33) |
| MCT4 | SLC16A3 | Lactate, ketone bodies | Skeletal muscle, chondrocytes, leucocytes, testis, lung, ovary, placenta, heart | Cancera and prognosisa (3,34–40) |
| MCT5 | SLC16A4 | Unknown | Brain, muscle, liver, kidney, lung, ovary, placenta, heart | Unknown |
| MCT6 | SLC16A5 | Bumetanide, nateglinide, probenecid | Kidney, muscle, brain, heart, pancreas, prostate, lung, placenta | Diet-induced lipid/glucose homeostasisb (41–42) |
| MCT7 | SLC16A6 | Ketone bodies | Liver, brain, pancreas, muscle, prostate | Unknown |
| MCT8 | SLC16A2 | Thyroid hormones (T2, rT3, T3, T4) | Most tissues including liver, heart, brain, thymus, intestine, ovary, prostate, pancreas, placenta, lung, kidney, skeletal muscle | Allan-Herndon-Dudley syndromea (4,5,43–46) |
| MCT9 | SLC16A9 | Carnitine | Endometrium, testis, ovary, breast, brain, kidney, spleen, retina | Uric acid homeostasis,b renal overload goutb (47) |
| MCT10 | SLC16A10 | Aromatic amino acids, T3, T4 | Kidney, intestine, muscle, placenta, heart | Similar to that of MCT8a (48) |
| MCT11 | SLC16A11 | Unknown | Skin, lung, ovary, breast, lung, pancreas, retinal pigment epithelium, choroid plexus | Unknown |
| MCT12 | SLC16A12 | Creatine | Kidney, retina, lung, testis | Cataracts,b glucosuriab (49–51) |
| MCT13 | SLC16A13 | Unknown | Breast, bone marrow stem cells | Unknown |
| MCT14 | SLC16A14 | Unknown | Brain, heart, muscle, ovary, prostate, breast, lung, pancreas liver, spleen, thymus | Unknown |
MCT, monocarboxylate transporter; SLC, solute carrier.
Table adapted from ref. 2.
Supported by in vitro and in vivo data.
Supported by in vivo data.
Supported by in vitro data.
Studies examining MCT1 molecular dynamics suggest a significant role of a lysine residue (K38) at the extracellular region of the protein in the transport function of MCT1.56 The proton-coupled symport activity exhibited by MCT1 is facilitated by sequential proton and lactate binding through electrostatic interactions, altering the conformation of the protein from a “closed” to an “open” state.10 Amino acid identity to MCT1 is low (~20–40%) for MCTs 5–14, and detailed mechanistic information regarding transporter functionality of other MCT isoforms has yet to be elucidated.
Regulation and trafficking of MCTs 1–4 have been associated with several ancillary proteins, as well as a variety of transcriptional and recently characterized post-transcriptional modulators; information is lacking for the other MCT isoforms. In numerous studies examining MCT trafficking and localization, two proteins, basigin (CD147) and embigin (gp70), have been identified as chaperone proteins important in the trafficking of MCTs 1–4 to the plasma membrane.57,58 Depending on the tissue and the MCT isoform, the interactions between these accessory proteins and MCTs have been experimentally confirmed and evaluated through in vitro approaches, such as immunoprecipitation and fluorescence resonance energy transfer, as well as in silico approaches using homology-based models. More commonly, basigin seems to be the most important protein for trafficking MCTs 1–4, with the exception of MCT2 in which embigin plays the major role in proper trafficking to the membrane.57 Although MCTs 5–14 seem to demonstrate independence from these two proteins (or the significance of this interaction has yet to be studied), it is evident that MCTs 1–4 are largely dependent on coexpression of these ancillary proteins for proper localization to the plasma membrane. Besides trafficking to the membrane, the activity of MCTs has also been shown to be modulated by carbonic anhydrases II and IX.59,60 The catalytic reaction performed by carbonic anhydrases, involved in the interconversion of carbon dioxide and water to bicarbonate and protons, was hypothesized to provide proton-coupled MCT isoforms 1–4 with enhanced functional activity through a “proton-collecting antenna” and not based on catalytic activity. However, further studies are needed prior to understanding this mechanism in more detail. More recently, the noncovalent interaction between cytosolic carbonic anhydrase II and an intracellular anionic amino acid moiety present in MCTs was investigated, which suggested the importance of the MCT C-terminus in regulatory protein-protein interactions.61 However, these interactions and their binding kinetics also need to be further evaluated in order to confirm this paradigm.
Regarding the transcriptional regulatory mechanisms of MCT1, nuclear factor of activated T-cells has been implicated in the regulatory binding in the MCT1 gene promoter region in skeletal muscle.52 The upregulation of MCT1 has been evaluated with increased exercise-induced Ca2+ -dependent protein phosphatase calcineurin activity, which is responsible for the dephosphorylation and downstream activation of nuclear factor of activated T-cells. This downstream activation is important considering that several consensus sequences for nuclear factor of activated T-cell binding have been identified in the promoter region of MCT1, which have been associated with activation of T-lymphocytes and increasing rates of glycolysis along with the upregulation of MCT1 gene expression.2 In addition, rising levels of AMP in response to exercise have also been associated with stimulation of AMP-activated protein kinase, which may potentially lead to increased gene expression of MCT1.1 Hypoxiainduced transcriptional regulatory mechanisms have also been associated with changes in the gene expression of MCT4 in a wide variety of tissues in the body. In contrast to MCT1 and MCT2, the MCT4 promoter region contains two response elements that are responsible for the binding of hypoxia-inducible factor-1α, leading to the increase in MCT4 gene expression in response to hypoxic conditions.62 Unfortunately, there is very limited information regarding the transcriptional regulation of other members of the MCT family and more research is required in order to elucidate the importance of these regulatory mechanisms. More recently, with the improved abilities to study post-transcriptional modifications and other modes of epigenetic regulation, MCT1, MCT3, and MCT4 genes have been shown to be regulated in response to DNA methylation in certain disease states, including breast cancer, atherosclerosis, and renal cancer.31,34,63 The miR-29a and miR-29b can contribute to the β-cell silencing of Mct1 gene in mouse islets, thus demonstrating that miRNA-mediated repression may be primarily involved in regulating the ubiquitous gene expression of Mct1 in tissues that lack expression, such as the pancreas.64 In addition, selective demethylation at the MCT2 promoter occurs in patients with prostate cancer, such that MCT2 gene expression may be utilized as a potential biomarker of prostate cancer.25 As more information becomes available regarding the regulation of MCTs 1–4, as well as the other isoforms, a better understanding of their potential significance in the prognosis and treatment of disease may be attained.
Proton-coupled MCTs (MCT1–4) and roles in disease
MCT1 has been utilized in drug delivery due to its high protein expression in the intestine: the drug product Horizant (gabapentin enacarbil) targets MCT1 to increase the bioavailability of gabapentin. In addition, MCT1 has been recognized as a useful tool for improving the availability of poorly permeable compounds to the central nervous system due to its protein expression at the blood brain barrier. MCT1-mediated drug delivery has been of particular interest most likely due to the cellular localization of MCT1 protein in the brain and its high protein expression compared with the other MCTs. MCT1 protein has been identified in brain endothelial cells, astrocytes, oligodendrocytes, microglial cells, and there is potential evidence for its neuronal expression in specific areas of the brain.52 4-phenylbutyrate (marketed in the United States as Buphenyl) has been used in the treatment of various conditions, including hyperammonemia, and more recently investigated for its neuroprotective effects. One recent study has demonstrated the likely importance of MCT1 in the facilitation of 4-phenylbutyrate across the blood brain barrier.65 For the purpose of this review of MCTs in clinical pharmacology and therapeutics, the primary focus will be on the more recently established roles of MCTs 1, 2, and 4 as targets in a wide variety of cancers. A review written by Pinheiro et al.3 in 2012 outlines the role of these transporters in a wide variety of human cancers based on protein expression and their ability to regulate cancer metabolism. Cancer cells have an intrinsic ability to undergo aerobic metabolic processes by which an abundance of lactic acid is produced as a byproduct of glucose metabolism, otherwise known as the “Warburg” effect.66 As summarized in Figure 1,52 lactate plays a major role in the symbiotic functioning between glycolytic and oxidative cancer cells. Seeing as lactate is a fundamental metabolic product and signaling agent, controlling its mechanism of transport through the administration of potent MCT inhibitors has been of considerable interest. Also, it has been reported that genes representing major roles in glycolysis are overexpressed in a set of 24 cancers, which encompasses >70% of all human cancer cases in the world.67 Several reports have also characterized the protein expression of MCTs 1, 2, and 4 in several types of common cancers.3,52,68 In particular, tumor cells are often markedly different than normal cells when comparing their highly glycolytic profile and abnormal hypoxic environments. The majority of the evidence of MCTs 1–4 in treatment of cancer relies heavily on preclinical data suggesting safety and efficacy, as well as from in vitro experiments in a wide variety of cancer cell lines. The primary points addressed in this section relate to the interplay of gene expression of MCTs 1–4, their regulation, and association to cancer cell metabolism and other diseased states, which make them attractive targets for the development of novel therapeutics.
Figure 1.
A scheme illustrating the lactate shuttling pathway in glycolytic and oxidative cancer cells. Glycolytic cancer cells (Left), dependent on glucose uptake via glucose transporters (GLUTs), undergo glycolysis to produce pyruvate and ATP, in which pyruvate undergoes further metabolism into lactate via lactate dehydrogenase A (LDHA). Proton symport-mediated lactate efflux occurs by MCT4. In oxidative cancer cells (Right), proton symport-mediated lactate influx and efflux occurs by MCT1. In these cells, lactate is transformed into pyruvate via lactate dehydrogenase B (LDHB) with the generation of NADH as a byproduct. These two metabolic products are then incorporated into the TCA cycle in mitochondria, to supplement the production of ATP via the oxidative phosphorylation pathway. This figure and caption was adapted from Perez-Escuredo et al.52
MCT1
Out of all the proton-dependent MCT isoforms investigated for their association in disease and therapeutic potential, none are more common than MCT1, which has been investigated for its role in cancer. Most notably, AstraZeneca developed a series of potent MCT1 inhibitors in the search for small molecules for immunosuppression that do not exhibit the same extent of adverse effects as current drugs12,13; one of these inhibitors is currently being evaluated for cancer treatment. A Cancer Research UK Phase I clinical trial of AZD3965 is recruiting patients to examine the safety and evaluate dose tolerance in solid tumors and diffuse large B-cell lymphoma (NCT01791595). The rationale to proceed with this clinical trial arose primarily from AZD3965’s efficacy in seven small cell lung cancer cell lines and its ability to reduce tumor growth in an small cell lung cancer tumor xenograft model.14,15 The experimental Ki value for this compound was demonstrated to be 1.6 nM for MCT1, exhibiting much less potency for MCT2. One of the primary problems that has been noted from these studies is the ability of MCT4 protein to be upregulated as a compensatory mechanism following MCT1 inhibition. In this regard, other mechanisms of inhibition and different classes of compounds are being evaluated for their clinical utility. Another class of compounds, the carboxycoumarins, has also recently been evaluated for their potential to disrupt the synergistic effect of glycolytic and oxidative metabolism in cancer cells.16 More specifically, the compound 7ACC2 demonstrated superior inhibition of lactate influx in cancer cells with an IC50 of 11 nM. Further investigation of these MCT1 inhibitors in preclinical models and phase I clinical studies is needed. Besides these newly developed compounds, the antitumor agent and mitochondrial hexokinase inhibitor, lonidamine, was found to be a potent inhibitor of the MCTs and mitochondrial pyruvate carrier at the plasma membrane, consistent with the effects of lonidamine on cancer cell pH, and lactate/ATP concentrations.17 In addition, the cancer chemotherapeutic agent 3-bromopyruvate is dependent on MCT1 for uptake and chemotherapeutic sensitivity.18
Perturbations in MCT1 gene expression levels have also been associated with a range of debilitating diseases. Exercise-induced hypoglycemia, caused by the failed silencing of the MCT1 gene in pancreatic β-cells, was found to be an inherited genetic disease mapped to chromosome 1.19,20 In normal pancreatic β-cells, most gene expression of MCT1 and other MCT isoforms is negligible9; however, in this genetic disorder, MCT1 gene expression is activated in response to mutations within the binding sites of several transcription factors in the promoter region. This inappropriate MCT1 expression results in a signaling cascade promoting ATP synthesis and secretion of insulin leading to hypoglycemia. Additionally, several missense mutations in the MCT1 gene can result in lactate transport deficiency.21 However, further studies evaluating the clinical relevance are needed in order to confirm the importance of these genetic mutations. One of the most commonly studied polymorphic variants of MCT1 is the MCT1-T1470A polymorphism (rs1049434), which has been previously suggested to influence changes in blood lactate concentrations during weight training and muscle injury in football players.22,23 This single nucleotide polymorphism results in the conversion of aspartic acid to glutamic acid at the 490 position, and patients carrying the T-allele exhibited a reduction of 35–40% in erythrocyte lactate transport rate.21 Although this reduction is large, more extensive studies are required to determine the clinical utility of this variant. The role of MCT1 in diet-induced obesity was also investigated recently in a preclinical haploinsufficient mouse model, which resulted in findings suggesting a role of MCT1 in dietary regulatory processes.24 Further studies into the clinical significance of MCT1 in diet and obesity are needed.
MCT2
In addition to MCT1, MCT2 has been investigated regarding its clinical relevance and potential as a drug target and biomarker in cancer, as well as other diseases. MCT2 generally has a higher affinity than MCT1 for monocarboxylates, such as lactate and pyruvate, which make it an ideal candidate in targeting cancer cell metabolism. Interestingly, findings from studies evaluating MCT2 protein expression in various cancer cells indicate that it is expressed primarily in the cytosol, suggesting its potential localization within intracellular organelles, such as mitochondria.3 More recently, MCT2 protein expression was demonstrated to be localized to the peroxisomes of human prostate cancer cells, and associated with malignant transformation processes.26 Of the cancerous tumors investigated for MCT2 protein expression, MCT2 was found to be expressed in colon, brain, breast, lung, and prostate cancers.
MCT2 protein expression has been recently evaluated as a potential candidate as a biomarker for prostate cancer.25,27 In one study, MCT2 was demonstrated to be comparable to α-methylacyl-CoA racemase, a previously established biomarker for prostate cancer.27 According to the American Cancer Society, prostate cancer is one of the most common cancers in men, making the investigation into new biomarkers an even more significant area of research. In neurodegenerative diseases, MCT2 protein expression was markedly downregulated with decreased lactate content in the cerebral cortex and hippocampus of an Aβ25–35-treated rat model of Alzheimer’s disease.28 However, in the MPTP mouse model of Parkinson’s disease, MCT2 protein expression levels were found to be unaltered and the metabolic impairments associated with Parkinson disease were not related to abnormalities of MCT2 protein expression.29 Considering MCT2 protein expression remains relatively high in neurons in comparison to other cell types in the brain, further investigation into MCT2’s role in neurodegeneration could prove fruitful as a future drug targeting modality.52 Although the clinical relevance of MCT2 in neurodegenerative diseases needs to be further investigated, it is apparent that there may also be variability in different preclinical models of neurodegenerative diseases that adds uncertainty in determining the association of MCT2 with neurodegeneration.
Additionally, investigation into genetic variants of MCT2 has also suggested the potential functional and clinical relevance of MCT2 in male spermatology and infertility.30 In this study, two single nucleotide polymorphisms (rs10506398 and rs10506399) were associated with male infertility in Korean men, resulting in a 2.4-fold decrease in sperm count with men exhibiting the homozygous rs10506399 variant. The findings of this study suggest that MCT2 single nucleotide polymorphisms may help us to understand its regulatory role in reproductive diseases, as well as aid in genetic risk assessments.
MCT3
MCT3 is distinct from other MCT isoforms because it is expressed predominantly in retinal pigmented epithelial cells and the choroid plexus epithelium.69–71 Although MCT3 remains less studied than MCTs 1, 2, and 4, preliminary evidence suggests that MCT3 may play a role in maintaining pH balance, ion homeostasis, and modulating wound healing in the retinal pigment epithelium.32,33 In 2008, Daniele et al.33 generated Mct3−/− knockout mice in order to examine the physiological role of MCT3 in the retina. The authors concluded that the disruption of pH homeostasis and lactate transport in this animal model resulted in increased lactate levels in the retina, suggesting an influence on the light responses of photoreceptors. Following this study, researchers evaluated the protein expression of MCT3 in chick and human fetal retinal pigmented epithelium (RPE) cell cultures in response to wounding and cell differentiation.32 It was noted that the mechanical disruption of the RPE resulted in decreased protein expression of MCT3 and positive MCT4 protein expression at the leading edge of the wound, followed by the re-expression of MCT3 protein in RPE cells dependent on the re-establishment of cell-cell contacts and the composition of the basement membrane.32 The changes in levels of expression of MCT3 protein suggest its utility as a biomarker for differentiated RPE cells. It is important to note that, with regards to the clinical relevance of this study, the authors concluded that by better understanding the changes in MCT expression within different RPE cell populations, we may be able to better understand retinal pathophysiology in cases, such as vitreoretinopathy, in which cell therapies could be utilized to treat damaged RPEs.
MCT4
With respect to MCT4 gene and protein expression in different cell types, MCT4 tends to have a much more abundant expression profile than MCT1 in cells where rates of glycolysis are high, such as lymphocytes, astrocytes, white muscle fibers, and tumor cells.2 The high affinity of MCT4 for pyruvate supports the theory that MCT4 promotes the uptake and subsequent metabolism of pyruvate to L-lactate. In addition, the protein expression of MCT4 in multiple cancer types shed light on its potential as a useful prognostic factor and potential cancer treatment strategy.3 Considerable data regarding MCT4 and its association with cancer have been generated in numerous experiments over the past decade, using a wide variety of in vitro and preclinical in vivo xenograft models. One of the major findings has been the association between breast cancer and MCT4 protein expression. In one experiment, it was shown that upregulation of MCT4 protein expression occurs in cancer-associated fibroblasts co-cultured with the MCF7 breast cancer cell line, in which oxidative stress activates hypoxia-inducible factor-1α inducing MCT4 gene transcription.35,62 In addition, the differential protein expression of MCT1 was also found to be upregulated; however, it was mainly localized to epithelial cancer cells, whereas MCT4 remained in stromal cells. This makes MCT4 a potential candidate target in blocking cancer metabolite homeostasis between glycolytic cancer fibroblasts and metabolic coupling to adjacent cancer cells using lactate as a fuel for energy production. This phenomenon, known as the “reverse Warburg effect,” has been evaluated more recently in the literature as a cancer cell targeting modality and for prognosis.36,37 By testing 181 patients with triple-negative breast cancer for MCT4 protein expression using immunostained tissue microarrays, stromal MCT4 was concluded to be a predictor of high-risk patients.37 The authors concluded that MCT4 inhibitors should be further developed in order to treat aggressive human breast cancer, while indicating the potential for MCT4 targeting in other cancers. In a comprehensive study investigating 17 breast cancer cell lines, MCT4 was found to play a major role in breast cancer cell survival, despite the large genetic diversity of the disease.36 Indeed, cancer cell metabolic adaptations are necessary for their growth and survival, suggesting that MCT4 represents a novel cancer treatment opportunity.
Besides the interest in MCT4 as a novel therapeutic target, its utility as a potential clinical biomarker for a wide variety of cancers has been investigated by several groups. As mentioned previously, MCT4 gene expression has been shown to be upregulated in clear cell renal cell carcinoma mediated by DNA methylation in the promoter region, also associated with cancer-related death.34 Data from this study showed that MCT4 gene expression and DNA methylation were predictive in assessing outcomes of patients with clear cell renal cell carcinomas, as well as predicting cancer-related death. More detailed and larger studies, however, are necessary in order to clinically determine the DNA methylation of the MCT4 gene as a predictive biomarker. MCT4 gene expression has also been shown to be highly upregulated in a glycolytic subset of pancreatic cancer, and the data demonstrated that elevated gene expression of MCT4 was associated with poor survival.38 Similarly, experimental evidence also suggests metabolic differences between adeno- and squamous cell non-small cell lung cancers, in which high MCT4 gene expression was indicated in more aerobic and glycolytic adenocarcinomas.39 In all these cases, it has been suggested that MCT4 and its association with glycolytic, metabolically active cancer cells can be used as not only a prognostic tool, but also a novel targeting strategy to inhibit the growth and function of these diseases.
More recently, a group at the British Columbia Cancer Research Centre investigated the utility of targeting the MCT4 gene using antisense oligonucleotides in castration-resistant prostate cancer.40 This study investigated the protein expression of MCT4, as well as the safety and efficacy of these antisense oligonucleotides in human castration-resistant prostate cancer cells and a PC-3 tumor xenograft mouse model. Results suggested that along with the elevated MCT4 protein expression in higher grade castration-resistant prostate cancer biopsies, there was increased immune cell penetration and activation following antisense oligonucleotide treatment.40 The results indicated by the study are exciting because there has been little in vivo evidence thus far suggesting the utility of MCT4 as a drug target in cancer. Considering there has been little success at generating small molecules as specific inhibitors of the MCT4 isoform, the generation of highly specific forms of treatment, such as oligonucleotides, represents an exciting advancement in the therapeutic targeting of MCT4.
MCT isoforms 6, 8–10, and 12 and roles in disease
MCT isoforms 5–14 have been substantially less addressed throughout the literature with regard to their clinical relevance and functional roles, compared with MCTs 1–4. More specifically, there is no information in the literature regarding MCTs 5, 11, and 13–14 other than data regarding the relative mRNA expression in human tissues, and little information on MCT7. Limited information is available regarding substrate specificity, functional characterization, and potential roles in vivo for the other MCT isoforms. This section will focus on the effect of MCTs 6, 8–10, and 12 in pathophysiology and disease and their potential clinical relevance as novel drug targets.
MCT6
With its endogenous function unknown and the lack of clarity surrounding MCT6 substrate specificity and functionality, MCT6 still remains an orphan transporter. Two major studies performed by the Yamamoto laboratory have attempted to classify MCT6 substrates, inhibitors, as well as its functional characterization using MCT6-transfected X. laevis oocytes.53,72 The results suggested that MCT6 is neither proton-, sodium-, nor bicarbonate-dependent, and primarily relies on pH and membrane potential as regulators and major driving forces for transport. Only a handful of drug substrates were identified (e.g. bumetanide, nateglinide, and probenecid), which suggests that MCT6 is a xenobiotic transporter.
Transcriptomic analyses performed in mouse liver homogenates in two separate studies indicate a role of MCT6 in regulating hepatic diet-induced changes in lipid and glucose homeostasis.41,42 Protein expression data available from the Human Protein Atlas suggests a high expression of MCT6 in human hepatocytes, which does not correlate to human mRNA expression data. These data suggest that MCT6 may have a fundamental role in the regulation of dietary fatty acids in the liver. It is important to note that further studies are required in vivo, potentially using a genetically deficient Mct6 knockout mouse model, prior to confirming the role of MCT6 and its potential importance in a clinical setting.
MCT8 and MCT10
MCT8 and MCT10 have both demonstrated affinity for thyroid hormones; however, mutations associated with the MCT8 gene have been associated with an X-linked mental retardation syndrome, known as Allan-Herndon-Dudley syndrome.4,5,43–46 Localized primarily to the neurons, as determined in mouse studies, MCT8 seems to be responsible for the specific neuronal uptake of a variety of thyroid hormones, such as diiodothyronine, triiodothyronine, and thyroxine. Although there is some evidence suggesting MCT8 mRNA and protein expression in human cerebral microvessels, more studies are required prior to confirming its significance in facilitating thyroid hormone transport across the blood brain barrier, compared with other transporters.73 These mutations in the MCT8 gene locus result in abnormal levels of thyroid hormones in the blood at an early age, which result in phenotypes, such as hypotonia, muscle hypoplasia, weakness, and developmental delays.45 Studies performed in Mct8-deficient mice have also demonstrated similar abnormalities to humans suffering from this disease.4,46 Due to the difficulty of studying this disease in humans, models such as these represent useful tools in studying the abnormalities associated with MCT8-deficiency and thyroid hormone disease. Although there is currently no defined treatment of this syndrome, a phase II clinical trial is ongoing using a triiodothyronine analog, Triac, to assist in normalizing serum thyroid hormone levels in patients with Allan-Herndon-Dudley syndrome (NCT02060474). Although Allan-Herndon-Dudley syndrome is a rare disorder with a low frequency of occurrence, the findings from these studies suggest that MCT8 plays a major role in regulating tissue-specific thyroid hormone levels and physiological development.
MCT10 (also referred to as TAT1) shows overlapping substrate specificity with MCT8 for transporting thyroid hormones; however, unlike MCT8, MCT10 demonstrates low affinity aromatic amino acid transport with compounds, such as tryptophan, tyrosine, and phenylalanine.74,75 Investigation into the contribution of MCT10 to thyroid hormone transport was performed in Mct10 knockout mice, and interestingly, no effects were seen in thyroid hormone levels in blood and tissue.48 However, the compensatory effects of Mct8 protein expression could potentially be playing a role in this model. In addition, the authors demonstrated that thyroid hormone concentrations within the livers and kidneys of double Mct8/Mct10 knockout mice were elevated compared to the Mct8 knockout mice, suggesting that both transporters contribute to the efflux of thyroid hormones. The clinical relevance of MCT10 has yet to be established, considering that it does not seem to be a major thyroid hormone transporter. However, further research into MCT10 substrate specificity and tissue-specific regulatory mechanisms may reveal its role in certain disease states.
MCT9
To date, very little evidence is available to suggest MCT9’s potential clinical relevance. However, one study suggested that a frequently occurring missense variant (rs2242206) of the MCT9 gene, resulting in a lysine change to a threonine at the 258 amino acid position, was significantly associated with an increase in risk of renal overload gout.47 Although studies have characterized MCT9 as a carnitine transporter,55 no evidence has suggested that it is responsible for the transport of urate, which would suggest its physiological association to gout. The importance of MCT9 in vivo has yet to be evaluated using a preclinical genetic knockout mouse model, which would assist in characterizing the clinical contribution to maintaining urate homeostasis.
MCT12
Recent evidence has suggested the potential physiological role and importance of MCT12 in humans; however, there still remains some debate surrounding its association with glucosuria and cataract formation.49–51 A nonsense mutation in the MCT12 gene that results in a Q215X amino acid mutation was found in a group of 12 subjects suffering from cataracts and elevated glucose levels in the urine.50 The premature termination of the MCT12 protein sequence is suggested to yield a highly or fully dysfunctional protein, which could lead to tissue-specific defects. Further studies were performed assessing the functional mechanism of this mutation in the development of cataracts.49 In vitro evidence and in vivo studies performed using genetically-modified Mct12 in rats in this study suggested that the human Q215X mutation in MCT12 resulted in cataract formation most likely due to protein misfolding and the inability to traffic to the plasma membrane. Interestingly, transposon-modified Mct12 rats resulting in the complete loss of Mct12 did not result in any detectable abnormal ocular phenotype, which suggests that there could be interspecies differences. More recently, MCT12 was characterized as a creatine transporter using MCT12-transfected X. laevis oocytes and the utilization of a high-resolution metabolomics approach.6 In order to further clarify the association of the MCT12 mutation and its relation to glucosuria, detailed in vitro studies and analysis of patients with the MCT12 mutation were performed.51 Although individuals with an MCT12 mutation displayed unaltered creatine levels from the wild-type patients, there was increased renal secretion of creatine’s precursor, guanidinoacetate. The glucosuria observed in the MCT12 mutation was also evaluated and demonstrated to be more directly correlated to a heterozygous SLGT2 mutation, and not MCT12.51 Further studies and more detailed data collection in vivo are still necessary, however, prior to characterizing the mechanistic effect of the MCT12 mutation on guanidinoacetate homeostasis.
Conclusions and future directions: Where are we headed?
Research targeting SLC transporters as therapeutic strategies in disease76 support the need for further investigations of SLC transport proteins as promising therapeutic targets in disease and drug therapy. This review highlights the emerging roles of the SLC16A family of transporters in drug targeting and as prognostic factors in disease, roles that will impact the field of clinical pharmacology. Currently, although MCTs 1–4 have been well-characterized, there still remain many undefined orphan MCTs, having no known endogenous substrates. MCTs 1–4 are lactate transporters, with MCT 1, 2, and 4 proteins being overexpressed in a variety of cancers and representing potential therapeutic targets for cancer treatment. MCT1, quantitatively the most important MCT, is a target for drug delivery both at the intestine and at the blood brain barrier. Investigations have suggested an important role of MCT3 in retinal pathophysiology, MCT6 in glucose and lipid homeostasis, MCT8/10 in thyroid hormone regulation, MCT9 in renal pathophysiology, and MCT12 as a creatine transporter that is potentially important in cataract development. Further research is needed for the characterization of these MCT enzymes, defining their roles in both xenobiotic disposition and in endogenous compound homeostasis, and for the determination of specific inhibitors of the MCT isoforms. The identification of specific substrates and inhibitors of the MCT isoforms has been limited, partly due to the lack of experimentally characterized 3D protein crystal structure resulting in the inability to reliably perform in silico ligand docking simulations. In addition, there remains very limited information available regarding genetic variability of these transporters, their roles in physiology and pathophysiology, as well as the clinical significance of the MCT family members in disease and their potential as disease biomarkers, prognostic factors, or therapeutic targets for the treatment of disease. With the exception of MCT1, the majority of the potential clinical relevance of MCTs as therapeutic targets is based primarily on preclinical and in vitro evidence. Therefore, more research is needed in order to fully understand the role of MCTs in disease and as drug targets.
References
- 1.Halestrap AP, Wilson MC. The monocarboxylate transporter family–role and regulation. IUBMB Life. 2012;64:109–119. doi: 10.1002/iub.572. [DOI] [PubMed] [Google Scholar]
- 2.Halestrap AP. The SLC16 gene family – structure, role and regulation in health and disease. Mol Aspects Med. 2013;34:337–349. doi: 10.1016/j.mam.2012.05.003. [DOI] [PubMed] [Google Scholar]
- 3.Pinheiro C, Longatto-Filho A, Azevedo-Silva J, Casal M, Schmitt FC, Baltazar F. Role of monocarboxylate transporters in human cancers: state of the art. J Bioenerg Biomembr. 2012;44:127–139. doi: 10.1007/s10863-012-9428-1. [DOI] [PubMed] [Google Scholar]
- 4.Dumitrescu AM, Liao XH, Weiss RE, Millen K, Refetoff S. Tissue-specific thyroid hormone deprivation and excess in monocarboxylate transporter (mct) 8-deficient mice. Endocrinology. 2006;147:4036–4043. doi: 10.1210/en.2006-0390. [DOI] [PubMed] [Google Scholar]
- 5.Trajkovic M, et al. Abnormal thyroid hormone metabolism in mice lacking the monocarboxylate transporter 8. J Clin Invest. 2007;117:627–635. doi: 10.1172/JCI28253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Abplanalp J, et al. The cataract and glucosuria associated monocarboxylate transporter MCT12 is a new creatine transporter. Hum Mol Genet. 2013;22:3218–3226. doi: 10.1093/hmg/ddt175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Halestrap AP, Meredith D. The SLC16 gene family – from monocarboxylate transporters (MCTs) to aromatic amino acid transporters and beyond. Pflugers Arch. 2004;447:619–628. doi: 10.1007/s00424-003-1067-2. [DOI] [PubMed] [Google Scholar]
- 8.Halestrap AP. The monocarboxylate transporter family–structure and functional characterization. IUBMB Life. 2012;64:1–9. doi: 10.1002/iub.573. [DOI] [PubMed] [Google Scholar]
- 9.Price NT, Jackson VN, Halestrap AP. Cloning and sequencing of four new mammalian monocarboxylate transporter (MCT) homologues confirms the existence of a transporter family with an ancient past. Biochem J. 1998;329(Pt 2):321–328. doi: 10.1042/bj3290321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Wilson MC, Meredith D, Bunnun C, Sessions RB, Halestrap AP. Studies on the DIDS-binding site of monocarboxylate transporter 1 suggest a homology model of the open conformation and a plausible translocation cycle. J Biol Chem. 2009;284:20011–20021. doi: 10.1074/jbc.M109.014217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Ovens MJ, Manoharan C, Wilson MC, Murray CM, Halestrap AP. The inhibition of monocarboxylate transporter 2 (MCT2) by AR-C155858 is modulated by the associated ancillary protein. Biochem J. 2010;431:217–225. doi: 10.1042/BJ20100890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Påhlman C, et al. Immunosuppressive properties of a series of novel inhibitors of the monocarboxylate transporter MCT-1. Transpl Int. 2013;26:22–29. doi: 10.1111/j.1432-2277.2012.01579.x. [DOI] [PubMed] [Google Scholar]
- 13.Murray CM, et al. Monocarboxylate transporter MCT1 is a target for immunosuppression. Nat Chem Biol. 2005;1:371–376. doi: 10.1038/nchembio744. [DOI] [PubMed] [Google Scholar]
- 14.Polanski R, et al. Activity of the monocarboxylate transporter 1 inhibitor AZD3965 in small cell lung cancer. Clin Cancer Res. 2014;20:926–937. doi: 10.1158/1078-0432.CCR-13-2270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Bola BM, et al. Inhibition of monocarboxylate transporter-1 (MCT1) by AZD3965 enhances radiosensitivity by reducing lactate transport. Mol Cancer Ther. 2014;13:2805–2816. doi: 10.1158/1535-7163.MCT-13-1091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Draoui N, et al. Synthesis and pharmacological evaluation of carboxycoumarins as a new antitumor treatment targeting lactate transport in cancer cells. Bioorg Med Chem. 2013;21:7107–7117. doi: 10.1016/j.bmc.2013.09.010. [DOI] [PubMed] [Google Scholar]
- 17.Nancolas B, et al. The anti-tumour agent lonidamine is a potent inhibitor of the mitochondrial pyruvate carrier and plasma membrane monocarboxylate transporters. Biochem J. 2016;473:929–936. doi: 10.1042/BJ20151120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Birsoy K, et al. MCT1-mediated transport of a toxic molecule is an effective strategy for targeting glycolytic tumors. Nat Genet. 2013;45:104–108. doi: 10.1038/ng.2471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Otonkoski T, et al. Physical exercise-induced hypoglycemia caused by failed silencing of monocarboxylate transporter 1 in pancreatic beta cells. Am J Hum Genet. 2007;81:467–474. doi: 10.1086/520960. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Otonkoski T, et al. Physical exercise-induced hyperinsulinemic hypoglycemia is an autosomal-dominant trait characterized by abnormal pyruvate-induced insulin release. Diabetes. 2003;52:199–204. doi: 10.2337/diabetes.52.1.199. [DOI] [PubMed] [Google Scholar]
- 21.Merezhinskaya N, Fishbein WN, Davis JI, Foellmer JW. Mutations in MCT1 cDNA in patients with symptomatic deficiency in lactate transport. Muscle Nerve. 2000;23:90–97. doi: 10.1002/(sici)1097-4598(200001)23:1<90::aid-mus12>3.0.co;2-m. [DOI] [PubMed] [Google Scholar]
- 22.Massidda M, et al. Influence of the MCT1 rs1049434 on indirect muscle disorders/injuries in elite football players. Sports Med Open. 2015;1:33. doi: 10.1186/s40798-015-0033-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Cupeiro R, et al. Influence of the MCT1-T1470A polymorphism (rs1049434) on blood lactate accumulation during different circuit weight trainings in men and women. J Sci Med Sport. 2012;15:541–547. doi: 10.1016/j.jsams.2012.03.009. [DOI] [PubMed] [Google Scholar]
- 24.Lengacher S, et al. Resistance to diet-induced obesity and associated metabolic perturbations in haploinsufficient monocarboxylate transporter 1 mice. PLoS One. 2013;8:e82505. doi: 10.1371/journal.pone.0082505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Pertega-Gomes N, et al. Epigenetic and oncogenic regulation of SLC16A7 (MCT2) results in protein over-expression, impacting on signalling and cellular phenotypes in prostate cancer. Oncotarget. 2015;6:21675–21684. doi: 10.18632/oncotarget.4328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Valença I, et al. Localization of MCT2 at peroxisomes is associated with malignant transformation in prostate cancer. J Cell Mol Med. 2015;19:723–733. doi: 10.1111/jcmm.12481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Pértega-Gomes N, et al. Monocarboxylate transporter 2 (MCT2) as putative biomarker in prostate cancer. Prostate. 2013;73:763–769. doi: 10.1002/pros.22620. [DOI] [PubMed] [Google Scholar]
- 28.Lu W, et al. Changes in lactate content and monocarboxylate transporter 2 expression in Aβ25–35-treated rat model of Alzheimer’s disease. Neurol Sci. 2015;36:871–876. doi: 10.1007/s10072-015-2087-3. [DOI] [PubMed] [Google Scholar]
- 29.Puchades M, Sogn CJ, Maehlen J, Bergersen LH, Gundersen V. Unaltered lactate and glucose transporter levels in the MPTP mouse model of Parkinson’s disease. J Parkinsons Dis. 2013;3:371–385. doi: 10.3233/JPD-130190. [DOI] [PubMed] [Google Scholar]
- 30.Lee J, Lee DR, Lee S. The genetic variation in monocarboxylic acid transporter 2 (MCT2) has functional and clinical relevance with male infertility. Asian J Androl. 2014;16:694–697. doi: 10.4103/1008-682X.124561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Zhu S, Goldschmidt-Clermont PJ, Dong C. Inactivation of monocarboxylate transporter MCT3 by DNA methylation in atherosclerosis. Circulation. 2005;112:1353–1361. doi: 10.1161/CIRCULATIONAHA.104.519025. [DOI] [PubMed] [Google Scholar]
- 32.Gallagher-Colombo S, Maminishkis A, Tate S, Grunwald GB, Philp NJ. Modulation of MCT3 expression during wound healing of the retinal pigment epithelium. Invest Ophthalmol Vis Sci. 2010;51:5343–5350. doi: 10.1167/iovs.09-5028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Daniele LL, Sauer B, Gallagher SM, Pugh EN, Jr, Philp NJ. Altered visual function in monocarboxylate transporter 3 (Slc16a8) knockout mice. Am J Physiol Cell Physiol. 2008;295:C451–C457. doi: 10.1152/ajpcell.00124.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Fisel P, et al. DNA methylation of the SLC16A3 promoter regulates expression of the human lactate transporter MCT4 in renal cancer with consequences for clinical outcome. Clin Cancer Res. 2013;19:5170–5181. doi: 10.1158/1078-0432.CCR-13-1180. [DOI] [PubMed] [Google Scholar]
- 35.Whitaker-Menezes D, et al. Evidence for a stromal-epithelial “lactate shuttle” in human tumors: MCT4 is a marker of oxidative stress in cancer-associated fibroblasts. Cell Cycle. 2011;10:1772–1783. doi: 10.4161/cc.10.11.15659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Baenke F, et al. Functional screening identifies MCT4 as a key regulator of breast cancer cell metabolism and survival. J Pathol. 2015;237:152–165. doi: 10.1002/path.4562. [DOI] [PubMed] [Google Scholar]
- 37.Witkiewicz AK, et al. Using the “reverse Warburg effect” to identify high-risk breast cancer patients: stromal MCT4 predicts poor clinical outcome in triple-negative breast cancers. Cell Cycle. 2012;11:1108–1117. doi: 10.4161/cc.11.6.19530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Baek G, et al. MCT4 defines a glycolytic subtype of pancreatic cancer with poor prognosis and unique metabolic dependencies. Cell Rep. 2014;9:2233–2249. doi: 10.1016/j.celrep.2014.11.025. [DOI] [PubMed] [Google Scholar]
- 39.Meijer TW, et al. Differences in metabolism between adeno- and squamous cell non-small cell lung carcinomas: spatial distribution and prognostic value of GLUT1 and MCT4. Lung Cancer. 2012;76:316–323. doi: 10.1016/j.lungcan.2011.11.006. [DOI] [PubMed] [Google Scholar]
- 40.Choi SY, et al. The MCT4 gene: a novel, potential target for therapy of advanced prostate cancer. Clin Cancer Res. 2016;22:2721–2733. doi: 10.1158/1078-0432.CCR-15-1624. [DOI] [PubMed] [Google Scholar]
- 41.Zhang F, Xu X, Zhou B, He Z, Zhai Q. Gene expression profile change and associated physiological and pathological effects in mouse liver induced by fasting and refeeding. PLoS One. 2011;6:e27553. doi: 10.1371/journal.pone.0027553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Lu Y, Boekschoten MV, Wopereis S, Müller M, Kersten S. Comparative transcriptomic and metabolomic analysis of fenofibrate and fish oil treatments in mice. Physiol Genomics. 2011;43:1307–1318. doi: 10.1152/physiolgenomics.00100.2011. [DOI] [PubMed] [Google Scholar]
- 43.Friesema EC, Ganguly S, Abdalla A, Manning Fox JE, Halestrap AP, Visser TJ. Identification of monocarboxylate transporter 8 as a specific thyroid hormone transporter. J Biol Chem. 2003;278:40128–40135. doi: 10.1074/jbc.M300909200. [DOI] [PubMed] [Google Scholar]
- 44.Dumitrescu AM, Liao XH, Best TB, Brockmann K, Refetoff S. A novel syndrome combining thyroid and neurological abnormalities is associated with mutations in a monocarboxylate transporter gene. Am J Hum Genet. 2004;74:168–175. doi: 10.1086/380999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Schwartz CE, et al. Allan-Herndon-Dudley syndrome and the monocarboxylate transporter 8 (MCT8) gene. Am J Hum Genet. 2005;77:41–53. doi: 10.1086/431313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Wirth EK, et al. Neuronal 3′,3,5-triiodothyronine (T3) uptake and behavioral phenotype of mice deficient in Mct8, the neuronal T3 transporter mutated in Allan-Herndon-Dudley syndrome. J Neurosci. 2009;29:9439–9449. doi: 10.1523/JNEUROSCI.6055-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Nakayama A, et al. Common missense variant of monocarboxylate transporter 9 (MCT9/SLC16A9) gene is associated with renal overload gout, but not with all gout susceptibility. Hum Cell. 2013;26:133–136. doi: 10.1007/s13577-013-0073-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Müller J, et al. Tissue-specific alterations in thyroid hormone homeostasis in combined Mct10 and Mct8 deficiency. Endocrinology. 2014;155:315–325. doi: 10.1210/en.2013-1800. [DOI] [PubMed] [Google Scholar]
- 49.Castorino JJ, et al. Juvenile cataract-associated mutation of solute carrier SLC16A12 impairs trafficking of the protein to the plasma membrane. Invest Ophthalmol Vis Sci. 2011;52:6774–6784. doi: 10.1167/iovs.10-6579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kloeckener-Gruissem B, et al. Mutation of solute carrier SLC16A12 associates with a syndrome combining juvenile cataract with microcornea and renal glucosuria. Am J Hum Genet. 2008;82:772–779. doi: 10.1016/j.ajhg.2007.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Dhayat N, et al. Mutation in the monocarboxylate transporter 12 gene affects guanidinoacetate excretion but does not cause glucosuria. J Am Soc Nephrol. 2016;27:1426–1436. doi: 10.1681/ASN.2015040411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Pérez-Escuredo J, et al. Monocarboxylate transporters in the brain and in cancer. Biochim Biophys Acta. 2016 doi: 10.1016/j.bbamcr.2016.03.013. e-pub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Murakami Y, et al. Functional characterization of human monocarboxylate transporter 6 (SLC16A5) Drug Metab Dispos. 2005;33:1845–1851. doi: 10.1124/dmd.105.005264. [DOI] [PubMed] [Google Scholar]
- 54.Hugo SE, Cruz-Garcia L, Karanth S, Anderson RM, Stainier DY, Schlegel A. A monocarboxylate transporter required for hepatocyte secretion of ketone bodies during fasting. Genes Dev. 2012;26:282–293. doi: 10.1101/gad.180968.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Suhre K, et al. Human metabolic individuality in biomedical and pharmaceutical research. Nature. 2011;477:54–60. doi: 10.1038/nature10354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Manoharan C, Wilson MC, Sessions RB, Halestrap AP. The role of charged residues in the transmembrane helices of monocarboxylate transporter 1 and its ancillary protein basigin in determining plasma membrane expression and catalytic activity. Mol Membr Biol. 2006;23:486–498. doi: 10.1080/09687860600841967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Wilson MC, Meredith D, Fox JE, Manoharan C, Davies AJ, Halestrap AP. Basigin (CD147) is the target for organomercurial inhibition of monocarboxylate transporter isoforms 1 and 4: the ancillary protein for the insensitive MCT2 is EMBIGIN (gp70) J Biol Chem. 2005;280:27213–27221. doi: 10.1074/jbc.M411950200. [DOI] [PubMed] [Google Scholar]
- 58.Poole RC, Halestrap AP. Interaction of the erythrocyte lactate transporter (monocarboxylate transporter 1) with an integral 70-kDa membrane glycoprotein of the immunoglobulin superfamily. J Biol Chem. 1997;272:14624–14628. doi: 10.1074/jbc.272.23.14624. [DOI] [PubMed] [Google Scholar]
- 59.Klier M, Andes FT, Deitmer JW, Becker HM. Intracellular and extracellular carbonic anhydrases cooperate non-enzymatically to enhance activity of monocarboxylate transporters. J Biol Chem. 2014;289:2765–2775. doi: 10.1074/jbc.M113.537043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Klier M, Schüler C, Halestrap AP, Sly WS, Deitmer JW, Becker HM. Transport activity of the high-affinity monocarboxylate transporter MCT2 is enhanced by extracellular carbonic anhydrase IV but not by intracellular carbonic anhydrase II. J Biol Chem. 2011;286:27781–27791. doi: 10.1074/jbc.M111.255331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Noor SI, et al. Analysis of the binding moiety mediating the interaction between monocarboxylate transporters and carbonic anhydrase II. J Biol Chem. 2015;290:4476–4486. doi: 10.1074/jbc.M114.624577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Ullah MS, Davies AJ, Halestrap AP. The plasma membrane lactate transporter MCT4, but not MCT1, is upregulated by hypoxia through a HIF-1alpha-dependent mechanism. J Biol Chem. 2006;281:9030–9037. doi: 10.1074/jbc.M511397200. [DOI] [PubMed] [Google Scholar]
- 63.Asada K, et al. Reduced expression of GNA11 and silencing of MCT1 in human breast cancers. Oncology. 2003;64:380–388. doi: 10.1159/000070297. [DOI] [PubMed] [Google Scholar]
- 64.Pullen TJ, da Silva Xavier G, Kelsey G, Rutter GA. miR-29a and miR-29b contribute to pancreatic beta-cell-specific silencing of monocarboxylate transporter 1 (Mct1) Mol Cell Biol. 2011;31:3182–3194. doi: 10.1128/MCB.01433-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Lee NY, Kang YS. In vivo and in vitro evidence for brain uptake of 4-phenylbutyrate by the monocarboxylate transporter 1 (MCT1) Pharm Res. 2016;33:1711–1722. doi: 10.1007/s11095-016-1912-6. [DOI] [PubMed] [Google Scholar]
- 66.Warburg O. On the origin of cancer cells. Science. 1956;123:309–314. doi: 10.1126/science.123.3191.309. [DOI] [PubMed] [Google Scholar]
- 67.Altenberg B, Greulich KO. Genes of glycolysis are ubiquitously overexpressed in 24 cancer classes. Genomics. 2004;84:1014–1020. doi: 10.1016/j.ygeno.2004.08.010. [DOI] [PubMed] [Google Scholar]
- 68.Pinheiro C, Reis RM, Ricardo S, Longatto-Filho A, Schmitt F, Baltazar F. Expression of monocarboxylate transporters 1, 2, and 4 in human tumours and their association with CD147 and CD44. J Biomed Biotechnol. 2010;2010:427694. doi: 10.1155/2010/427694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Yoon H, Fanelli A, Grollman EF, Philp NJ. Identification of a unique monocarboxylate transporter (MCT3) in retinal pigment epithelium. Biochem Biophys Res Commun. 1997;234:90–94. doi: 10.1006/bbrc.1997.6588. [DOI] [PubMed] [Google Scholar]
- 70.Philp NJ, Yoon H, Grollman EF. Monocarboxylate transporter MCT1 is located in the apical membrane and MCT3 in the basal membrane of rat RPE. Am J Physiol. 1998;274(6 Pt 2):R1824–R1828. doi: 10.1152/ajpregu.1998.274.6.R1824. [DOI] [PubMed] [Google Scholar]
- 71.Philp NJ, Yoon H, Lombardi L. Mouse MCT3 gene is expressed preferentially in retinal pigment and choroid plexus epithelia. Am J Physiol Cell Physiol. 2001;280:C1319–C1326. doi: 10.1152/ajpcell.2001.280.5.C1319. [DOI] [PubMed] [Google Scholar]
- 72.Kohyama N, Shiokawa H, Ohbayashi M, Kobayashi Y, Yamamoto T. Characterization of monocarboxylate transporter 6: expression in human intestine and transport of the antidiabetic drug nateglinide. Drug Metab Dispos. 2013;41:1883–1887. doi: 10.1124/dmd.113.051854. [DOI] [PubMed] [Google Scholar]
- 73.Roberts LM, et al. Expression of the thyroid hormone transporters monocarboxylate transporter-8 (SLC16A2) and organic ion transporter-14 (SLCO1C1) at the blood-brain barrier. Endocrinology. 2008;149:6251–6261. doi: 10.1210/en.2008-0378. [DOI] [PubMed] [Google Scholar]
- 74.Kim DK, Kanai Y, Chairoungdua A, Matsuo H, Cha SH, Endou H. Expression cloning of a Na+-independent aromatic amino acid transporter with structural similarity to H+/monocarboxylate transporters. J Biol Chem. 2001;276:17221–17228. doi: 10.1074/jbc.M009462200. [DOI] [PubMed] [Google Scholar]
- 75.Ramadan T, et al. Basolateral aromatic amino acid transporter TAT1 (Slc16a10) functions as an efflux pathway. J Cell Physiol. 2006;206:771–779. doi: 10.1002/jcp.20531. [DOI] [PubMed] [Google Scholar]
- 76.Lin L, Yee SW, Kim RB, Giacomini KM. SLC transporters as therapeutic targets: emerging opportunities. Nat Rev Drug Discov. 2015;14:543–560. doi: 10.1038/nrd4626. [DOI] [PMC free article] [PubMed] [Google Scholar]

